17.12.2012 Views

dermatology - DermaAmin

dermatology - DermaAmin

dermatology - DermaAmin

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

DERMATOLOGY: CLINICAL & BASIC SCIENCE SERIES<br />

COPPER and<br />

the SKIN


DERMATOLOGY: CLINICAL & BASIC SCIENCE SERIES<br />

Series Editor Howard I. Maibach, M.D.<br />

Published Titles:<br />

Bioengineering of the Skin: Cutaneous Blood Flow and Erythema<br />

Enzo Berardesca, Peter Elsner, and Howard I. Maibach<br />

Bioengineering of the Skin: Methods and Instrumentation<br />

Enzo Berardesca, Peter Elsner, Klaus P. Wilhelm, and Howard I. Maibach<br />

Bioengineering of the Skin: Skin Biomechanics<br />

Peter Elsner, Enzo Berardesca, Klaus-P. Wilhelm, and Howard I. Maibach<br />

Bioengineering of the Skin: Skin Surface, Imaging, and Analysis<br />

Klaus P. Wilhelm, Peter Elsner, Enzo Berardesca, and Howard I. Maibach<br />

Bioengineering of the Skin: Water and the Stratum Corneum,<br />

Second Edition<br />

Joachim W. Fluhr, Peter Elsner, Enzo Berardesca, and Howard I. Maibach<br />

Contact Urticaria Syndrome<br />

Smita Amin, Arto Lahti, and Howard I. Maibach<br />

Copper and the Skin<br />

Jurij J. Host´ynek and Howard I. Maibach<br />

Cutaneous T-Cell Lymphoma: Mycosis Fungoides and Sezary Syndrome<br />

Herschel S. Zackheim and Howard I. Maibach<br />

Dermatologic Botany<br />

Javier Avalos and Howard I. Maibach<br />

Dermatologic Research Techniques<br />

Howard I. Maibach<br />

Dry Skin and Moisturizers: Chemistry and Function, Second Edition<br />

Marie Lodén and Howard I. Maibach<br />

The Epidermis in Wound Healing<br />

David T. Rovee and Howard I. Maibach<br />

Hand Eczema, Second Edition<br />

Torkil Menné and Howard I. Maibach<br />

Human Papillomavirus Infections in Dermatovenereology<br />

Gerd Gross and Geo von Krogh<br />

The Irritant Contact Dermatitis Syndrome<br />

Pieter van der Valk, Pieter Coenrads, and Howard I. Maibach


Latex Intolerance: Basic Science, Epidemiology, and Clinical Management<br />

Mahbub M. V. Chowdhry and Howard I. Maibach<br />

Nickel and the Skin: Absorption, Immunology, Epidemiology,<br />

and Metallurgy<br />

Jurij J. Host´ynek and Howard I. Maibach<br />

Pesticide Dermatoses<br />

Homero Penagos, Michael O’Malley, and Howard I. Maibach<br />

Protective Gloves for Occupational Use, Second Edition<br />

Anders Boman, Tuula Estlander, Jan E. Wahlberg, and Howard I. Maibach<br />

Sensitive Skin Syndrome<br />

Enzo Berardesca, Joachim W. Fluhr, and Howard I. Maibach<br />

Skin Cancer: Mechanisms and Human Relevance<br />

Hasan Mukhtar<br />

Skin Reactions to Drugs<br />

Kirsti Kauppinen, Kristiina Alanko, Matti Hannuksela, and Howard I. Maibach


DERMATOLOGY: CLINICAL & BASIC SCIENCE SERIES<br />

COPPER and<br />

the SKIN<br />

Edited by<br />

Jurij J. Host´ynek<br />

University of California at San Francisco School of Medicine<br />

San Francisco, California, U.S.A.<br />

Howard I. Maibach<br />

University of California at San Francisco School of Medicine<br />

San Francisco, California, U.S.A.<br />

New York London


Informa Healthcare USA, Inc.<br />

270 Madison Avenue<br />

New York, NY 10016<br />

© 2006 by Informa Healthcare USA, Inc.<br />

Informa Healthcare is an Informa business<br />

No claim to original U.S. Government works<br />

Printed in the United States of America on acid‑free paper<br />

10 9 8 7 6 5 4 3 2 1<br />

International Standard Book Number‑10: 0‑8493‑9532‑1 (Hardcover)<br />

International Standard Book Number‑13: 978‑0‑8493‑9532‑1 (Hardcover)<br />

This book contains information obtained from authentic and highly regarded sources. Reprinted material<br />

is quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable<br />

efforts have been made to publish reliable data and information, but the author and the publisher cannot<br />

assume responsibility for the validity of all materials or for the consequences of their use.<br />

No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,<br />

mechanical, or other means, now known or hereafter invented, including photocopying, microfilming,<br />

and recording, or in any information storage or retrieval system, without written permission from the<br />

publishers.<br />

For permission to photocopy or use material electronically from this work, please access www.copyright.<br />

com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222 Rosewood<br />

Drive, Danvers, MA 01923, 978‑750‑8400. CCC is a not‑for‑profit organization that provides licenses and<br />

registration for a variety of users. For organizations that have been granted a photocopy license by the<br />

CCC, a separate system of payment has been arranged.<br />

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are<br />

used only for identification and explanation without intent to infringe.<br />

Visit the Informa Web site at<br />

www.informa.com<br />

and the Informa Healthcare Web site at<br />

www.informahealthcare.com


This monograph on copper is dedicated to Dr. Roberto Milanino of the<br />

University of Verona, Italy. A scientist and teacher, he spent the past 30 years<br />

of his academic career investigating the biological function and importance of<br />

copper in the mammalian organism while navigating the narrows of Italy’s<br />

budget allocated to science. Among the leading authorities on this subject,<br />

he was a guiding light, set the proper accents, and kept things in perspective<br />

over the period of much of our work and writing in assembling this monograph.<br />

The whole is a clear reflection of his qualities as teacher.<br />

J. J. Hosty´nek<br />

H. I. Maibach


Preface<br />

Preface<br />

Metals have long interested the dermatologic community in terms of their<br />

toxicity and efficacy. A century ago, mercury enjoyed widespread usage in<br />

the treatment of syphilis.<br />

As the dermatologic sciences evolved, sufficient knowledge also evolved,<br />

leading to the clear assertion that metals may be toxic when applied to the<br />

skin. Nickel has enjoyed the greatest amount of study, especially because of<br />

its frequent induction of clinical allergic contact dermatitis in humans. Cobalt<br />

is also a common contact allergen, but its clinical significance is less clearly<br />

explored. Chromate in cement, leather, and other applications also enjoys<br />

considerable study. Most recently, gold salts have been recognized as a common<br />

inducer of cell-mediated immunity; however, this clinical significance is<br />

currently being investigated in terms of dermatitis and even restenosis.<br />

These data have led to several textbooks dedicated to individual metals.<br />

The first was on chromate (by Desmond Burrows), many metals and the skin<br />

(by R. Guy and J. J. Hosty´nek), and most recently nickel (by J. J. Hosty´nek<br />

and H. I. Maibach). The current volume on copper presents sufficient information<br />

to place it among the pantheon of the metallic gods and <strong>dermatology</strong>.<br />

Our aim was to mold contributions from individuals widely spread over<br />

several disparate disciplines into a cohesive, readily digestible text. The<br />

individual disciplines include basic chemistry (metallurgy), dermatotoxicology<br />

(irritant and allergic contact dermatitis), and membrane transport<br />

(percutaneous penetration).<br />

v


vi Preface<br />

Our specific objective is to allow scientists in many fields to more<br />

efficiently focus their attention on this essential element (copper) and the<br />

skin. Hopefully, the parts will simplify understanding the whole.<br />

This volume differs from the others, not only in its extensive dermatotoxicologic<br />

profile of copper and its salts, but also as an equally impressive<br />

data on copper’s possible anti-inflammatory actions in man.<br />

The editors welcome suggestions for the next edition.<br />

Jurij J. Hosty´ nek<br />

Howard I. Maibach


Acknowledgment<br />

We gratefully acknowledge the partial financial support by the International<br />

Copper Association, Ltd. (ICA) towards publication of this book.<br />

vii


Preface . . . . v<br />

Acknowledgment . . . . vii<br />

Contributors . . . . xv<br />

Contents<br />

1. Copper and Copper Alloys<br />

Harold T. Michels<br />

. . . . . . . . ................ 1<br />

Introduction . . . . 1<br />

Copper: Properties of the Element . . . . 1<br />

Pure Copper . . . . 2<br />

Copper Alloys . . . . 2<br />

Properties of Copper Alloys . . . . 2<br />

Copper Alloy Families . . . . 4<br />

The High Coppers . . . . 4<br />

Conclusions . . . . 6<br />

2. Corrosion Chemistry of Copper: Formation of Potentially<br />

Skin-Diffusible Compounds . . . . . . . . ................<br />

Jurij J. Hosty´nek<br />

7<br />

Introduction . . . . 7<br />

Electron Configuration and Reactivity of Copper . . . . 8<br />

Corrosion of Copper in the Environment . . . . 8<br />

Corrosion of Copper in Physiologic Media . . . . 9<br />

Conclusions . . . . 15<br />

Glossary . . . . 16<br />

Abbreviations . . . . 16<br />

References . . . . 16<br />

ix


x Contents<br />

3. Basics of Metal Skin Penetration:<br />

Scope and Limitations . . . . . . .................... 21<br />

Jurij J. Hosty´ nek and Howard I. Maibach<br />

Introduction . . . . 21<br />

Structure of Skin and Its Function as<br />

Diffusion Barrier . . . . 23<br />

Descriptors of Dermal Absorption . . . . 25<br />

Permeant Categories and Paths of Diffusion . . . . 28<br />

Compounds Formed by Metals in Contact<br />

with the Skin . . . . 31<br />

Variables Determining Skin Diffusion<br />

of Metal Compounds . . . . 35<br />

Methods for Measuring Percutaneous Absorption . . . . 45<br />

Analytical Methods for Metal Detection . . . . 53<br />

Summary and Conclusions . . . . 56<br />

Abbreviations . . . . 57<br />

References . . . . 58<br />

4. Percutaneous Absorption of Copper Compounds . . . . . . . . 67<br />

Jurij J. Hosty´ nek and Howard I. Maibach<br />

Introduction . . . . 67<br />

Qualitative Diffusion Data . . . . 68<br />

Semiquantitative Data . . . . 70<br />

Quantitative Data . . . . 71<br />

Discussion and Conclusions . . . . 73<br />

Limitations in Measuring Copper Absorption<br />

In Vivo . . . . 74<br />

Interdependence of Systemic Copper and Zinc Levels . . . . 75<br />

Recommendations for Research to Fill Existing<br />

Data Gaps . . . . 76<br />

Conclusions . . . . 77<br />

Glossary . . . . 78<br />

Abbreviations . . . . 78<br />

References . . . . 79<br />

5. Diffusion of Copper Through Human Skin<br />

In Vivo . . .................................. 81<br />

Jurij J. Hosty´ nek, Howard I. Maibach, and Frank Dreher<br />

Introduction . . . . 81<br />

Experimental . . . . 84<br />

Results . . . . 85


Contents xi<br />

Discussion . . . . 88<br />

Conclusions . . . . 92<br />

Glossary . . . . 93<br />

Abbreviations . . . . 93<br />

References . . . . 94<br />

6. Irritation Potential of Copper Compounds . . . . ........ 97<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Introduction . . . . 97<br />

Exposure to Copper . . . . 97<br />

Solubilization of Copper Metal . . . . 98<br />

Incidence and Epidemiology of Irritation<br />

Due to Copper . . . . 100<br />

Pharmacology of Copper . . . . 101<br />

Copper Irritancy in Skin and Mucosa . . . . 103<br />

Conclusions . . . . 111<br />

Abbreviations . . . . 112<br />

References . . . . 112<br />

7. Copper Hypersensitivity: Dermatologic<br />

Aspects—Overview . . . ........................ 115<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Introduction . . . . 115<br />

Metallurgy of Copper and Its Alloys, and Its Role<br />

as Sensitizer . . . . 117<br />

Predictive Immunology Test Results<br />

for Copper . . . . 119<br />

Diagnostic Tests for Hypersensitivity . . . . 119<br />

Test Concentrations for Copper ACD . . . . 123<br />

Immunogenic Potential of Copper . . . . 123<br />

Summaries of Population-Based Studies . . . . 134<br />

Summary of Selected Case Reports of Immune Reactions<br />

to Copper . . . . 138<br />

Selection of Individual Reports of Immune Reactions<br />

to Copper . . . . 138<br />

Comments . . . . 140<br />

Conclusions . . . . 140<br />

Abbreviations . . . . 141<br />

References . . . . 141


xii Contents<br />

8. Copper in Medicine and Personal Care:<br />

A Historical Overview . . . . . . ................... 149<br />

Roberto Milanino<br />

Introduction . . . . 149<br />

The Sumeric Culture: Circa 4000–2300 B.C. .... 150<br />

The Ancient Egyptian Culture . . . . 150<br />

The Babylonian–Assyrian Culture:<br />

Circa 1750–539 B.C. .... 152<br />

The Ancient Indian Culture: Circa 2800–1000 B.C. .... 152<br />

The Ancient Chinese Culture: Circa 3000 B.C.<br />

to 1100 A.D. .... 152<br />

The Pre-Columbian Meso- and South-American Cultures:<br />

Circa 600 B.C. to 1500 A.D. .... 153<br />

The Ancient Greek Culture . . . . 153<br />

The Ancient Roman Culture: Circa 600 B.C.<br />

to 476 A.D. .... 155<br />

From the High-Medieval Age to the Early<br />

20th Century . . . . 156<br />

Beginning of the Scientific Age for Copper:<br />

1928–1976 . . . . 157<br />

Conclusions . . . . 158<br />

Abbreviations . . . . 159<br />

References . . . . 159<br />

9. The Role of Copper in Onset, Development, and<br />

Control of Acute and Chronic Inflammation . . . . . . . . . . 161<br />

Roberto Milanino<br />

Introduction . . . . 161<br />

Studies on Copper-Deficient, Experimentally<br />

Inflamed Animals . . . . 163<br />

Laboratory Animals: Studies on ‘‘Endogenous’’ Copper<br />

Metabolism in Acute and Chronic Inflammation . . . . 170<br />

Human Subjects: Studies on ‘‘Endogenous’’ Copper Metabolism<br />

in Acute and Chronic Inflammations, with a Particular<br />

Reference to Rheumatoid Arthritis . . . . 179<br />

Effects of ‘‘Exogenous’’ Copper Administration on the<br />

Inflammatory Process . . . . 184<br />

Copper Anti-inflammatory Activity: Hypotheses Explaining<br />

the Possible Mechanisms of Action . . . . 203<br />

Conclusions . . . . 216


Contents xiii<br />

Abbreviations . . . . 219<br />

References . . . . 220<br />

10. Copper Jewelry and Arthritis . . . . . . .............. 237<br />

Brenda J. Harrison<br />

Introduction . . . . 237<br />

The Copper Bracelet ‘‘Myth’’ and Hypothesis . . . . 239<br />

The Copper Bracelet Trial . . . . 243<br />

The Present State of the Copper Bracelets ‘‘Issue’’ . . . . 251<br />

Is There Likely to Be a Future for Copper Bracelets<br />

in Arthritis Care? . . . . 256<br />

Appendix A: Position Statements of Support Organizations,<br />

Government Agencies, Etc. . . . . 257<br />

Appendix B: Miscellany . . . . 259<br />

References . . . . 261<br />

11. Role of Copper in Anti-inflammatory Therapy and the<br />

Potential for Its Transdermal Application . . . . . ....... 267<br />

Jurij J. Hosty´nek and Roberto Milanino<br />

Introduction . . . . 267<br />

Traditional and Modern Therapies for RA<br />

and Related Disorders . . . . 268<br />

Drug Therapy . . . . 271<br />

Precedents in Topical Delivery of Anti-inflammatory<br />

Agents . . . . 275<br />

Role of Copper in AI Activity . . . . 275<br />

Past Use of Copper Chelates in the Treatment<br />

of Rheumatoid Arthritis . . . . 278<br />

Transdermal Delivery of Anti-inflammatory Copper<br />

Chelates vs. Conventional (Systemic)<br />

Anti-inflammatory Therapy . . . . 278<br />

Conclusions . . . . 286<br />

Outlook . . . . 288<br />

Abbreviations . . . . 288<br />

References . . . . 289<br />

Index . . . . 295


Contributors<br />

Frank Dreher Neocutis, Inc., San Francisco, California, U.S.A.<br />

Brenda J. Harrison Department of Earth and Ocean Sciences, Copper<br />

Research Information Flow Project, University of British Columbia,<br />

Vancouver, British Columbia, Canada<br />

Jurij J. Hosty´nek Department of Dermatology, University of California<br />

at San Francisco School of Medicine, San Francisco, California, U.S.A.<br />

Howard I. Maibach Department of Dermatology, University of California<br />

at San Francisco School of Medicine, San Francisco, California, U.S.A.<br />

Harold T. Michels Copper Development Association, Inc., New York,<br />

New York, U.S.A.<br />

Roberto Milanino Facoltà di Medicina e Chirurgia, Sezione di<br />

Farmacologia, Dipartimento di Medicina e Salute Pubblica, Università di<br />

Verona, Verona, Italy<br />

xv


1<br />

Copper and Copper Alloys<br />

Harold T. Michels<br />

Copper Development Association, Inc., New York, New York, U.S.A.<br />

INTRODUCTION<br />

This first chapter describes copper, its properties and characteristics, and<br />

where it is used, both in its pure form and as alloys. The emphasis is on<br />

materials that come into contact with human skin. This chapter provides<br />

the background for the second chapter, which gives a detailed discussion of<br />

the corrosion resistance of these materials and how that relates to their<br />

interaction with humans by sweat. Common items made of copper and<br />

copper alloys that are touched by humans every day include copper and copper–nickel<br />

coins, copper–nickel–zinc door keys, and brass door knobs, door<br />

push plates, and sink faucet handles.<br />

COPPER: PROPERTIES OF THE ELEMENT<br />

Copper, atomic number 29, is classified as a metal in the periodic table of<br />

elements. It is the first element in the group containing silver and gold,<br />

and thus it is considered to be a semiprecious metal.<br />

Some of the properties of copper include:<br />

melting point of 1083 C<br />

metallic luster and reddish color<br />

high electrical and thermal conductivity<br />

nonmagnetic<br />

alloys readily as both a solute and a solvent<br />

1


2 Michels<br />

good corrosion resistance and durability<br />

forms a protective oxide in air and water<br />

face-centered crystal structure<br />

high malleability, formability, and ductility<br />

good machinability<br />

readily electroplated<br />

essential nutrient for life<br />

highly recyclable<br />

These unique properties of copper account for its widespread and<br />

long-term use as an industrial material.<br />

PURE COPPER<br />

Copper is refined from ore shipped to fabricators, mainly as cathode, wire<br />

rod, billet, cake (slab), or ingot. Through extrusion, drawing, rolling, forging,<br />

melting, or atomization, fabricators form wire, rod, tube, sheet, plate,<br />

strip, castings, powder, and other shapes. These copper and copper alloys<br />

are then shipped to manufacturing plants where they are used to make<br />

products to meet society’s needs.<br />

COPPER ALLOYS<br />

Copper alloys are widely used in many applications, ranging from electrical<br />

wiring and connectors to musical instruments, from household plumbing<br />

tube and fixtures to keys, locks, doorknobs, and handrails. The applications<br />

are almost endless. The wide use of copper alloys is attributable to a long<br />

history of successful use, ready availability from a multitude of sources, the<br />

attainability of a wide range of physical and mechanical properties, and<br />

amenability to subsequent processing, such as machining, brazing, soldering,<br />

polishing, and plating. The properties of copper alloys, which occur<br />

in unique combinations found in no other alloy system, include high<br />

thermal and electrical conductivity, a wide range of attainable strength properties<br />

and excellent ductility and toughness, as well as superior corrosion<br />

resistance in many different environments.<br />

Nevertheless, to the uninitiated, copper alloys appear to be confusing<br />

and complex. They are generically described by such terms as brass, bronze,<br />

copper–nickel, and copper–nickel–zincs, which are called nickel silvers<br />

because of their shiny white color, even though they contain no silver.<br />

PROPERTIES OF COPPER ALLOYS<br />

Copper alloys provide important properties and characteristics including:<br />

Good corrosion resistance—which contributes to durability, and<br />

leads to long-term cost effectiveness.


Copper and Copper Alloys 3<br />

Favorable mechanical properties—ranging from pure copper that is<br />

soft and ductile to other alloys, such as the manganese bronzes that<br />

can rival the mechanical properties of quenched and tempered<br />

steel. Furthermore, almost all copper alloys retain their mechanical<br />

properties, including impact toughness, at very low temperatures.<br />

High thermal and electrical conductivity—copper has higher conductivity<br />

than any other metal except silver. Conductivity drops<br />

when copper is alloyed. However, even the copper alloys with relatively<br />

low conductivity transfer both heat and electricity far better<br />

than other corrosion-resistant materials, such as titanium, aluminum,<br />

and stainless steel.<br />

Biofouling resistance—copper inhibits the growth of marine organisms<br />

including algae and barnacles. This property, unique to<br />

copper, decreases when alloyed. However, it is retained at a useful<br />

level in copper alloys, such as the copper–nickels, which are routinely<br />

found in marine applications.<br />

Antimicrobial action—copper chemicals have been historically used as<br />

bactericides, algicides, and fungicides. However, recent studies indicate<br />

that bacteria, including certain harmful strains of Escherichia<br />

coli, and MRSA, or methicillin-resistant Staphylococcus aureus<br />

(a serious nosocomial or hospital-acquired infection) simply die in a<br />

few hours when placed on copper alloy surfaces at room temperature.<br />

Low friction and wear rates—copper alloys, such as the high-leaded<br />

tin bronzes, are cast into sleeve bearings and exhibit low wear rates<br />

against steel. Both the nickel bronze and the tin bronze are the<br />

industry standards for worm gears, an application in which low<br />

wear rates are important.<br />

Good castability—all are sand castable and almost all can be<br />

centrifugally and continuously cast. Many copper alloys can be permanently<br />

molded and precision or die cast. Wrought copper alloys<br />

are initially cast and subsequently hot and cold rolled.<br />

High fabricability—copper alloys are readily hot rolled extruded or<br />

forged. They then may be cold rolled to the desired thickness.<br />

Sheet, plate, strip, and bar products are readily forged, stamped,<br />

and bent into desired shapes.<br />

High machinability—good surface finish and high tolerance control<br />

is readily achieved. While the leaded copper alloys are free-cutting<br />

at high machining speeds, many unleaded alloys such as nickel–<br />

aluminum bronze are readily machinable at recommended feeds<br />

and speeds with proper tooling.<br />

Ease of subsequent processing—many copper alloys are routinely<br />

polished to a high luster, especially those with an esthetically pleasant<br />

color, such as the yellow brasses. Plating, soldering, brazing,<br />

and welding are also routinely performed.


4 Michels<br />

Availability of a range of alloys—in a given application, any one of<br />

several alloys may be a suitable candidate, depending on design<br />

loads and corrosivity of the environment.<br />

Reasonable cost—high processing yield and low machining costs<br />

make copper alloy very economical. Gates and risers from castings<br />

and chips from machining are also recycled, which leads to additional<br />

overall cost reductions. In addition, copper alloys do not<br />

require surface coatings, such as paints. The avoidance of surface<br />

coatings further reduces initial costs and provides additional maintenance<br />

savings. In addition, when the component reaches the end<br />

of its useful life, it too is readily and routinely recycled.<br />

COPPER ALLOY FAMILIES<br />

From a metallurgical viewpoint, many copper alloys are single-phase solid<br />

solutions in which the alloying elements, such as zinc, tin, and nickel, are<br />

substituted for copper in the copper matrix. Examples of single-phase<br />

solution alloys include the brasses that contain up to 35% zinc, and copper–<br />

nickels that contain up to 33% nickel. As alloy content is increased, a second<br />

phase may form. In the case of brass, when the zinc content is increased, a<br />

hard second phase called beta forms within the alpha copper-rich matrix.<br />

This second phase is found in yellow brass that contains up to 41% zinc.<br />

Beta, which slightly impairs room temperature ductility, markedly increases<br />

ductility at elevated temperatures. One of the most common systems used<br />

to designate specific copper alloys is the UNS system. Copper alloys are<br />

either wrought or cast. Wrought alloys range in UNS number from C10100<br />

through C79999. They are subjected to hot and usually cold work after initial<br />

melting and solidification, and are generally available as wire, rod, bar,<br />

sheet, strip, and plate. Cast alloys range in UNS number from C80000<br />

through C99999. They are typically cast into a mold in a variety of specific<br />

shapes and then machined without any hot or cold working.<br />

The Coppers<br />

The coppers (wrought: C10100–C15999, and cast: C80000–C81399) are<br />

essentially pure copper (99.7% min) with traces of silver or phosphorus.<br />

The presence of silver imparts annealing resistance, while phosphorus, a deoxidizer,<br />

aids in welding. These alloys, which have relatively low strength, are used<br />

in applications where high thermal and electrical conductivity are desirable,<br />

such as in electrical connectors and in hot metal handling. Copper is used<br />

in low denomination coinage. Some individuals wear copper wrist bracelets.<br />

THE HIGH COPPERS<br />

High copper alloys (wrought: C16000–C19999, and cast: C81400–C83299),<br />

which contain 95.1% copper (min), are unique in that they combine high


Copper and Copper Alloys 5<br />

strength with high thermal and electrical conductivity, two properties that<br />

are seldom found together in the same material.<br />

Chromium coppers are alloys containing up to 1.2% chromium. The<br />

strength of chromium copper is approximately twice that of pure copper,<br />

but its electrical conductivity remains high (80% of pure copper). Applications<br />

for chromium copper include welding clamps and high-strength<br />

electrical connectors.<br />

The Brasses<br />

The brasses (wrought: C21000–C49999, and cast: C83300–C89999) are the<br />

most commonly used casting alloys, and are essentially alloys of copper<br />

and zinc.<br />

The red brasses are alloys of up to 15% zinc and may contain varying<br />

amounts of tin if they are cast alloys. Lead may be present in various<br />

amounts to promote pressure tightness in castings in service and to facilitate<br />

free machining during the manufacturing process. The color of red brass is<br />

attributable to its relatively low zinc content. The largest-volume cast red<br />

brass alloy, C83600 (commonly known as 85-5-5-5), contains 85% copper,<br />

5% tin, 5% lead, and 5% zinc. It has been used commercially for several<br />

hundred years and accounts for more tonnage than any other cast alloy.<br />

The yellow brasses are even lower in cost than the red brasses because<br />

their zinc content is higher, at 20–39% Zn. Yellow brass has a pleasant<br />

yellow color, which can be polished to a high luster. This accounts in part<br />

for its selection as decorative hardware.<br />

The Bronzes<br />

Bronze (wrought: C50000–C69999, and cast: C90000–C95999) is a very<br />

imprecise term. Strictly speaking, it originally referred to alloys in which<br />

tin was the major alloying element. Today, the term bronze applies to a<br />

broader class of alloys, which may contain little, if any, tin.<br />

The silicon bronzes and silicon brasses are essentially alloys of up to<br />

20% zinc and up to 5% silicon. They have low melting points and high fluidity,<br />

which favor permanent molding and pressure die casting.<br />

Modern day cast tin bronzes are very similar to the alloys found in<br />

many relics from the ‘‘Bronze Age,’’ over 3500 years ago. They are basically<br />

alloys of copper and tin, where tin content can be up to 20%. The good<br />

aqueous corrosion resistance of tin bronzes accounts, in part, for the survival<br />

of these Bronze Age relics to this day. Additional attributes of tin<br />

bronzes include reasonably high strength, good wear resistance, and a low<br />

coefficient of friction versus steel, making them very useful for bearings,<br />

piston rings, and gear parts.<br />

Aluminum bronzes have complex metallurgical structures. Alloying<br />

elements always include aluminum and varying amounts of manganese,


6 Michels<br />

iron, and, in some versions, nickel. Aluminum imparts both strength and<br />

oxidization resistance by virtue of the formation of alumina (Al2O3)-rich<br />

protective films. These alloy are very wear resistant, and exhibit good casting<br />

and welding characteristics. Their corrosion resistance is superior in<br />

seawater, chloride, and in dilute acids. Applications are varied and include<br />

propellers and valves, pickling hooks, pickling baskets, and wear rings. Aluminum<br />

bronzes and especially nickel–aluminum bronzes are desirable alloys<br />

for fluid moving applications such as pump impellers, because of superior<br />

erosion-corrosion and cavitation resistance.<br />

Copper Nickels<br />

The copper–nickel alloys (wrought: C70000–C73499, and cast: C96000–<br />

C96999) are simple solid solutions of nickel in copper. Nickel content varies<br />

from 9% to 33%. A small amount of manganese (0.05–1.5%) and iron<br />

(0.4–1.8%) is present. Their excellent corrosion resistance in seawater, combined<br />

with their high strengths and good fabricability, accounts for their<br />

wide use in piping, heat exchangers, valves, ship tail-shaft sleeves, and other<br />

marine applications. They are also used in coins.<br />

Nickel Silvers<br />

Nickel silver alloys (wrought: C73500–C79999, and cast: C97000–C97999)<br />

contain nickel (11–27%) and zinc (1–25%). The presence of nickel primarily<br />

accounts for their pleasant silver luster but, in contrast to their name, the<br />

nickel silvers do not contain silver. In spite of their high degree of alloying,<br />

the nickel silvers are simple solid solution alloys. They offer good corrosion<br />

resistance, ease of castability, and good machinability. Major uses include<br />

hardware for food processing, seals, architectural trim, musical instrument<br />

valves, and door keys.<br />

Other Copper Alloys<br />

In the interest of completeness, two other types of copper alloys, which are<br />

both cast alloys, should be mentioned. They are the leaded coppers<br />

(C89000–C98000), which are typically employed where their high lead provides<br />

lubricity, and the special alloys (C99000–C99999), which have unique<br />

specific properties needed for specialized applications.<br />

CONCLUSIONS<br />

The copper alloys offer a wide range of properties to meet the needs of many<br />

applications that humans touch. The list is almost endless. End-use applications<br />

are limited only by the knowledge, creativity, and imagination of those<br />

who specify, design, produce, and process these copper alloy products.


2<br />

Corrosion Chemistry of Copper:<br />

Formation of Potentially<br />

Skin-Diffusible Compounds<br />

Jurij J. Hostynek<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

INTRODUCTION<br />

In the background of this review lies the question, what effect, if any, does<br />

copper metal have when kept in contact with the skin, on inflammation, and<br />

arthritis in particular—i.e., concerning the unsettled issue of the ‘‘copper<br />

bangle.’’ Copper complexes given systemically have a well-documented therapeutic<br />

effect on inflammatory conditions (1), and anti-inflammatory agents<br />

(NSAIDs) have also proven successful by dermal delivery (2,3). Investigations<br />

demonstrating the efficacy of dermal assimilation of copper in arthritic and<br />

rheumatoid conditions by skin contact with the metal or its derivatives in<br />

humans have been few, poorly conducted, and have only brought qualitative,<br />

mostly subjective, evidence of benefits to be expected from the ‘‘mere’’ skin contact<br />

with the metal (4). The review of the microenvironment prevailing on the<br />

skin surface and the chemical agents present there aims to delineate acceptable<br />

chemical arguments that support the contention that copper, solubilized in that<br />

environment, is apt to be assimilated by the mammalian organism.<br />

This chapter, in part, was reprinted from Hosty´nek JJ. Factors determining percutaneous metal<br />

absorption. Food Chem Toxicol 2003; 41:327–345, with permission of Elsevier.<br />

7


8 Hostynek<br />

ELECTRON CONFIGURATION AND REACTIVITY OF COPPER<br />

The transition metals in the periodic table have partially filled d (or f) electron<br />

shells, in which an inner shell is being filled with electrons, two in each of five<br />

orbitals, while the outer s-electron shell of slightly lower energy is complete.<br />

Characteristic for transition metals is a low ionization potential (electropositivity),<br />

i.e., the ease with which they can lose electrons to yield a variety of<br />

positive ionic oxidation states, and the ability to form complexes. This occurs<br />

mainly with sulfur, (e.g., in histidine), oxygen, or nitrogen groups (e.g., in<br />

cysteine), particularly in biological systems and chelates (cyclic coordination<br />

compounds) that are stable and likely to remain unchanged in physical<br />

processes, such as membrane transport.<br />

Copper has a single s electron outside the filled 3d shell, but the d electrons<br />

also are involved in metallic bonding, and the (most common) Cu(II)<br />

ion has the configuration 3d 9 , having lost the single s electron from the<br />

outermost shell and one d electron. While further oxidation to Cu(III) is<br />

difficult, the lower oxidation states can interchange easily. For instance,<br />

the equilibrium 2Cu(I) D Cu(0)þ Cu(II) can be readily displaced in either<br />

direction. Copper complexes with proteins—molecules composed entirely<br />

of alpha-amino acid residues covalently united head to tail by peptide<br />

bonds—to form unbranched polymers. Examples are copper metallothionein<br />

(a small, ubiquitous protein in the organism fulfilling a multifunctional role<br />

in absorption and reversible storage, transport and detoxification of the<br />

highly toxic free copper ion) ceruloplasmin (the enzyme involved in the antiinflammatory<br />

action of copper), blood clotting factors V and VIII, and<br />

copper glycyl-L-histidyl-L-lysine, Cu(II)-GHK, which may prevent the inhibition<br />

of mitochondrial glucose oxidation (5–8).<br />

CORROSION OF COPPER IN THE ENVIRONMENT<br />

For metals and their alloys exposed to the environment, the major reactions<br />

are oxidation to the ionic form and liberation of electrons:<br />

M ! M xþ þ x e<br />

and the reduction of the (obligatory) oxygen present to form water:<br />

O2 þ 4H þ þ 4e ! 2H2O<br />

In presence of oxygen, on the surface of pure copper metal cuprous<br />

oxide is formed, whereby copper ion persists in the monovalent Cu(I) state,<br />

due to the reducing action of the metal. In the presence of saline (water),<br />

oxygen dissolved in the medium can oxidize Cu(I) further to yield the cupric<br />

ion, Cu(II), with formation of free radicals (9).<br />

Although copper and its alloys have been known and widely used since<br />

prehistoric times, appropriately named the Bronze Age, corrosion of the


Corrosion Chemistry of Copper 9<br />

pure metal has hardly been investigated and is poorly documented. The corrosive<br />

action of the environment (e.g., seawater) has focused on its alloys,<br />

the bronzes and brasses, but not on pure metal itself (10). Well established<br />

is the corrosive action of saline, a prime factor in the dissolution of the metal<br />

(11), due to which copperware used in cooking is usually lined with tin.<br />

While main sources of exposure to copper occurs through the diet (animal<br />

organs, seafood, certain vegetables, and nuts), unsuspected exposure<br />

may be due to release of copper corrosion products from copper cooking<br />

and tableware (12), or the metal released into the plumbing system of water<br />

conduits (13). Low hardness, low alkalinity, chlorinated tap water in municipal<br />

water supplies, particularly of low pH and at elevated temperatures<br />

(as in hot water conduits), increases the rate of corrosion. The effects of temperature,<br />

chlorine, and organic matter on copper alloys have been investigated<br />

in a number of studies (14).<br />

Corrosion products of copper, brass, or bronze that are formed on<br />

exposure to air, of different hues of blue and green, are collectively referred<br />

to as verdigris, a term possibly derived from the term ‘‘vert de Grece,’’ mentioned<br />

in ancient texts. A mixture of cupric acetate Cu(CH3COO)2, basic<br />

cupric acetate Cu(OH) (CH3COO), and carbonate CuCO3, verdigris salts<br />

occurring naturally are mostly noticeable as patina on weathering copper<br />

objects, such as roofs and statues, described in Egyptian records dating<br />

back to 1500 B.C. In post-Renaissance Europe, verdigris was used in oil-based<br />

paint pigments to decorate interiors of homes, and as wood preservative.<br />

Formed by the action of acetic acid vapors on metallic copper since the<br />

Middle Ages and until the early 20th century, verdigris was produced in<br />

the wine-growing regions of France as a cottage industry. Flat copper strips<br />

were rubbed with verdigris, then layered with fermented grape husks for<br />

several weeks until crystals formed on the metal surface. Dripping wine on<br />

the surface promoted the formation of spongy green crystals that, scraped<br />

from the copper, were dissolved in vinegar, recrystallized on wooden sticks,<br />

crushed, and sold as pigment (15).<br />

CORROSION OF COPPER IN PHYSIOLOGIC MEDIA<br />

Copper and its alloys are present in numerous articles of everyday use, and<br />

thus come in regular, sometimes extended and intimate contact with the skin,<br />

or through systemic exposure by implanted prostheses or contraceptive<br />

devices [intrauterine devices (IUDs)]. Heat, moisture, sweating, and friction<br />

promote chemical reactions on the surface of objects in contact with the skin.<br />

Skin: The Action of Sweat and Sebum<br />

To gain insight onto the phenomenon of skin penetration by metals in<br />

general, it is important to account for the factors involved in that process


10 Hostynek<br />

stemming from the chemical microenvironment prevailing on the skin<br />

surface, as the oxidative action encountered there is a determinant factor<br />

in the formation of skin-diffusible derivatives. By the action of salts and<br />

acids present in sweat and sebum, metals in their elemental state are converted<br />

to the hydrophilic or lipophilic derivatives, respectively. Only as such<br />

do metals become diffusible via the transcellular, intercellular, or transappendageal<br />

route.<br />

Leaching or release of copper ion from metal objects (alloys) in contact<br />

with sweat is a multifactorial event that defeats prediction. Aside from<br />

immediate environmental factors, the microenvironment within the particular<br />

alloy in contact with a conducting medium (electrolyte, e.g., sweat) is a<br />

principal determinant, due to the action of electromotive forces prevailing at<br />

the interface of atoms with differing electromotive potential (16).<br />

Such metals alloyed with copper form a galvanic element (or pile),<br />

whereby an electron current flows from the more electronegative to the more<br />

electropositive one, resulting in oxidation or solution (‘‘corrosion’’) of the<br />

more electronegative metal. These effects, unpredictable quantitatively, vary<br />

in function of actual metals present and their ratios in alloy composition.<br />

A recent example with potential impact on public health is the recently minted,<br />

bimetallic (cupronickel) 1- and 2-Euro coins, consisting of 25% nickel.<br />

That bimetallic structure, which generates a potential difference of 30–40 mV<br />

between the components, increases nickel release, due to galvanic corrosion,<br />

facilitated in the medium of conducting sweat. As a consequence, reports of<br />

nickel sensitizations from <strong>dermatology</strong> clinics are on the increase in a number<br />

of countries where the new currency is in circulation, as nickel release in handling<br />

the coins may elicit contact dermatitis in those allergic to nickel (17–22).<br />

While amino acids, such as glycine or histidine, or proteins that can<br />

complex with cupric ions enhance the rate of copper released, the presence<br />

of saline and oxygen are prime factors in the dissolution of the metal. In<br />

their absence no copper is released (9,23).<br />

As formulated for the general reaction of a metal with amino acids or<br />

fatty acids present in sweat, the process can be presented as:<br />

2M 0 þ O2 þ 4HL ! 2ML2 þ 2H2O<br />

where M 0 is the metal in elemental form and HL the endogenous ligand.<br />

The Major Sweat Components<br />

Electrolytes<br />

The considerable variation in electrolyte composition occurring naturally in<br />

sweat became evident in a number of studies conducted on the subject.<br />

In one study on normal subjects, in Palmar sweat both sodium and<br />

chloride levels fell below 50 milliequivalents/L (mEq/L) for 99% of subjects,<br />

and below 65 mEq/L in all 649 volunteers tested (24).


Corrosion Chemistry of Copper 11<br />

Using direct reading ion-selective electrodes, mean sodium concentration<br />

in forearm transudate was 1.7 mEq/L 0.7, and chloride concentration<br />

2.8 mEq/L 3.5 (n ¼ 6) (25).<br />

Mean body sweat induced through exercise contained 24.2 2.2 mEq/L<br />

chloride ion in men, and 26.0 4.6 in women (26).<br />

Sodium and potassium content of pharmacologically stimulated sweat<br />

in men (pilocarpine, methylcholine, and acetylcholine) was seen to be higher<br />

than that measured in thermal sweat: mean values (in mEq/L) are 8.3 0.66<br />

and 4.9 0.17, respectively (27).<br />

Sweat osmolality values in normal adults were seen to increase with<br />

increasing age. Range/mean (SD) values for men are 49–151/117 mmol/kg<br />

(33.4) and for women 66–187/134 mmol/kg (38.6). An increase in osmolality<br />

was observed to increase in tandem with the (normal) increase in<br />

sodium concentration in sweat among an aging population (28).<br />

The main cause for corrosion of metal surfaces from skin contact in<br />

individuals referred to as ‘‘rusters’’ is not due to elevated electrolyte concentration,<br />

as generally assumed, but rather seems to coincide with palmar<br />

hyperhydrosis. When the sodium concentration measured in normal subjects<br />

was compared to that of ‘‘rusters,’’ in fact no significant difference<br />

could be observed (mean values of 49.6 vs. 49.1 mEq/L, respectively) (29).<br />

Amino Acids<br />

Proteins and AAs are normal components of mammalian sweat. The data<br />

documented for humans differ significantly, probably due to differences<br />

in the stimulation methods applied, to regional differences in anatomic site,<br />

or the sampling methods used. Substantial variations were observed in relative<br />

concentrations of AAs in sweat collected from various areas of the<br />

skin, and their concentrations increased markedly in blood and urine on<br />

oral protein intake (30). No differences in AA patterns were seen between<br />

young and middle-aged adults, or between men and women (31). In<br />

contrast to essential elements, AAs are neither selectively excreted nor<br />

reabsorbed (32).<br />

Large individual differences occur in AA composition between eccrine<br />

forearm sweat from men under controlled exercise conditions. Comparison<br />

of AA excretions analyzed in sweat and urine by ion exchange chromatography<br />

showed comparable losses in those two media. In general, AA<br />

concentrations were considerably higher in the exercise sweat of untrained<br />

men than in the sweat of trained men determined by thermal and physiological<br />

stimulation. Total average AA values collected from 20 trained<br />

men and 20 untrained men were 12,797 and 24,855 mmol/L, respectively.<br />

For trained versus untrained men, highest values were seen for serine<br />

(3954/7782), glycine (2239/4392), alanine (1556/3028) and threonine<br />

(1057/1856), respectively (33).


12 Hostynek<br />

Lactate and Pyruvate<br />

Lactate is the major organic compound secreted in sweat. At rest and low<br />

sweating rates, the range in lactate concentrations is 30–40 mmol/L, at<br />

higher sweat rate levels 10–15 mmol/L. Under thermal stimulation and also<br />

during exercise, lactate concentration in sweat decreases with increasing<br />

sweating rate, but remains constant in blood. Differences in the excretion<br />

of lactate were also observed in function of physical fitness of three male<br />

volunteers: mean values for the sedentary individual was 21.71 0.85, for<br />

the fit individual 16.75 0.99, and the very fit individual was 12.75<br />

0.50 mmol, respectively (34).<br />

In contrast, pyruvic acid (pyruvate) concentrations in sweat are low,<br />

found to vary between 0.1 and 1.2 mmol/L. The ratio of the two metabolites,<br />

lactate and pyruvate, was observed to increase with rising heart rate (35).<br />

Sebum<br />

Human skin features an acid mantle of pH 4–6 at the surface of the SC in<br />

normal, healthy subjects, which increases with depth to pH 7 at the juncture<br />

with live tissue (36). Determinants of this pH are protons originating in the<br />

epidermis or as products of sebaceous gland activity, which gradually reach<br />

the surface of the skin. They stem from three classes of compounds:<br />

amino acids (e.g., urocanic acid, pyrrolidone carboxylic acid)<br />

alpha-hydroxy acids (e.g., lactic and butyric acid, also present in<br />

sweat)<br />

acidic lipids (e.g., cholesteryl sulfate and free fatty acids, primarily<br />

oleic, linoleic, and behenic) (37–40)<br />

Sebum as secreted by the sebaceous glands is a complex mixture of<br />

lipids consisting of glycerides, but no free fatty acids. The occurrence<br />

of free acids in the SC and on the skin surface is the result of hydrolysis of<br />

phospholipids and glycerides by lipolytic enzymes occurring in the sebaceous<br />

ducts and on the skin surface, and of bacterial decomposition. On<br />

the skin surface, lipids of epidermal origin contain up to 20% free fatty<br />

acids, those originating in the pilosebaceous glands, 16% (40). They consist<br />

for the greater part of C16 and C18 acids, but their full range reaches<br />

from C5 to C22, with an average length of C16 (39,41). Such an acid<br />

milieu plays a regulating role for SC homeostasis, with relevance to the<br />

integrity of the skin’s barrier function and regeneration of the SC (42).<br />

It is believed that the acid environment on the skin surface both controls<br />

moisture loss from the epidermis and protects the skin from fungal and<br />

bacterial infection.<br />

These acid components making up the sebum also play an important<br />

role in solubilizing (‘‘corroding’’) metal surfaces in measurable amounts.


Corrosion Chemistry of Copper 13<br />

The Release of Copper Ion in Artificial Sweat<br />

The definition and thus reconstitution of human sweat for experimental purposes<br />

fluctuate in function of numerous factors, which is why experimental<br />

results are not consistent. Most striking are changes in sweat composition,<br />

due to the rate of sweat secretion. Sodium and chloride content, one decisive<br />

factor in the corrosion of copper and metals in general, is as low as 5 mEq/L<br />

under quiescent conditions, due to a reabsorption (conservation) mechanism<br />

(43,44). As sweating rate increases, that control mechanism is overwhelmed<br />

and the sodium concentration can rise to approximate that occurring in<br />

plasma. Also other significant components of sweat such as urea, lactic acid,<br />

and potassium ions, increase at high rates of secretion and their concentrations<br />

can also reach the levels of plasma.<br />

Release of copper ions from various copper alloys in synthetic sweat<br />

was investigated in model experiments.<br />

Over a period of 24 hours at 35 C and a pH of 5.1, the range of Cu 2þ<br />

liberated in artificial sweat was 80–100 mg/mL sweat (45). Walker and<br />

Keats performed a number of experiments to determine the solubility of<br />

copper metal in artificial sweat. In samples where initial copper concentration<br />

was of the order of 2 10 5 M, after 24 hours the samples turned blue<br />

and the concentration had increased to 2 10 3 mol Cu (46).<br />

Oxidation of Copper in Contact with the Skin<br />

Walker and Keats also measured the loss from copper bracelets worn by<br />

volunteers. Bracelets measuring 22 1.3 0.1 cm lost 80 mg in 50 days when<br />

worn around the ankles, and 90 mg when worn around the wrist (4).<br />

Weight losses of 0.1% to 0.8% were observed for copper bracelets<br />

weighing 14 g used in a trial involving volunteers and lasting one month<br />

(47). Such variation can be explained by differential mechanical wear, but<br />

a more likely alternative explanation is the variability in subjective sweat<br />

composition and sweating rate.<br />

Skin Penetration Data<br />

Although limited in number, experiments have demonstrated that copper<br />

compounds will penetrate the integument in humans and animals, mostly<br />

in a qualitative manner. Such information is based on clinical observations<br />

of remedial action in inflammatory diseases, describing therapeutic effects,<br />

and qualitative observation by spectroscopic methods (Warner RR, 1993,<br />

personal communication) (48–51).<br />

One experiment only measured diffusivity of a skin-identical complex,<br />

bis(glycinato) copper(II), in vitro on cat skin (52). A radioactive ( 64 Cu)<br />

0.05 M solution in physiological saline applied to excised cat skin revealed,


14 Hostynek<br />

after a lag time of nine hours, a steady-state transport rate described by a<br />

Kp¼ 24 10 4 cm/hr.<br />

Electron micrographs of skin sections stained for copper revealed the<br />

metal in all layers of the skin in exposed samples. In 24 hours, 3.3% of<br />

the applied copper(II) complex had completely penetrated the skin. This is<br />

equivalent to 3.0 mg of the complex. Atomic absorption analysis found<br />

47 ppm copper in the washing solution beneath the skin (20 mL isotonic<br />

saline). This corresponds to 0.94 mg, which, in agreement with the radioactivity,<br />

is equivalent to 3.1 mg of the complex.<br />

To examine whether skin exudates will oxidize the surface of copper<br />

metal in contact with skin, forming potentially diffusible compounds, copper<br />

powder was applied under occlusion on the volar forearm of human<br />

volunteers at UCSF. When the application areas were sequentially stripped<br />

with adhesive tape and the strips then analyzed for metal content by inductively<br />

coupled plasma-mass spectroscopy, a concentration gradient became<br />

evident from the skin surface to the subcutis, characteristic of a passive diffusion<br />

process. The results indicate that in contact with skin compounds are<br />

formed, which can penetrate the intact stratum corneum to the level of live<br />

epidermis (unpublished study).<br />

Release of Copper in the Human Organism and<br />

Its Contraceptive Effects<br />

In the presence of saline, amino acids (e.g., glycine or histidine) or proteins<br />

that complex with cupric ions accelerate the release of copper by removing free<br />

copper ions from equilibrium. Copper incubated in a saline medium will cleave<br />

the S–S bond, and serum albumin will undergo a conformational change (11).<br />

Incubation with copper metal was seen to be spermatocidal at cupric ion concentrations<br />

below 10 3 mol (53), and enzymes important in the implantation<br />

process were also inactivated on incubation in the presence of copper metal<br />

in a saline medium. Copper metal incubated in saline in the presence of oxygen<br />

produces free radicals (9), and it appears probable that free radicals thus<br />

formed are responsible for the contraceptive action of copper in IUDs.<br />

The ‘‘copper IUD,’’ a plastic T-shaped device with copper wire wound<br />

around its stem, has an increased contraceptive effect in women as compared<br />

to the T insert without copper, and it continues to be a popular method of<br />

temporary contraception. With the aim of obtaining insight into the mode<br />

of action of these devices, studies were conducted to investigate the effect of<br />

copper IUDs on the uterine milieu in women. The chemical process involved<br />

on the surface of the metal in the uterus appears responsible for the contraceptive<br />

activity (i.e., inhibition of implantation) (54).<br />

In solutions of saline and serum albumin, a strip of 200 mm 2 copper<br />

dissolved at a rate comparable to that of copper IUDs when measured<br />

in vivo (23). Such losses of copper metal were determined on a number of


Corrosion Chemistry of Copper 15<br />

copper IUDs in the actual use environment, by weighing before their insertion<br />

into the uterus and after their removal a year later. Following one<br />

year’s use, the loss from two sets of IUDs (120 and 135 mm 2 surface area)<br />

amounted to 10.0 2.2 and 11.0 2.8 mg, respectively, the average release<br />

per day being calculated at 28.7 mg (55).<br />

Increases in systemic copper levels via parenteral exposure from a copper<br />

IUD can also lead to adverse effects, even though the amounts liberated<br />

from such a device are relatively low. Frentz and Teilum (54) traced induction<br />

of copper hypersensitivity in patients to the action of a copper IUD.<br />

They measured copper released from the device at 90 mg/day.<br />

A fertility-related phenomenon due to the local increase in systemic<br />

copper ion was described by Vesce et al., who noted that the use of the copper<br />

intrauterine device was effective in the management of secondary amenorrhea.<br />

In 40 of 48 volunteers with functional secondary amenorrhea, regular<br />

menses were restored after insertion of an IUD, and normal menses were<br />

maintained as long as the IUD remained in place. After removal of the<br />

device, the effect persisted for one year. The authors of that study ascribe<br />

the mechanism of action to the copper ion-mediated release of prostaglandins<br />

from the endometrium (56).<br />

CONCLUSIONS<br />

Copper and its alloys are subject to chemical reactions on exposure to<br />

environmental or physiological factors, whereby products are potentially<br />

generated, which become diffusible through mammalian skin. The chemistry<br />

of oxidation is reviewed as well as the factors contributing to corrosion. Skin<br />

exudates (sweat and sebum) can react with metal surfaces that they come in<br />

contact with, but even in the healthy organism their composition is variable,<br />

in function of physical, pharmacological and environmental conditions,<br />

gender, age, sweat rate, or body site. This overview addresses sweat and<br />

sebum composition, and discusses components that determine the skin’s<br />

corrosive action: chloride ion, low molecular weight acids and amino acids<br />

(AAs) in sweat, and fatty acids in sebum, which hold the potential to solubilize<br />

copper-containing metal objects. These components can form copper<br />

salts and soaps whose molecular characteristics (size and polarity) will<br />

determine rate and route of cutaneous penetration.<br />

Rate and degree of copper corrosion is subject to a number of variable<br />

environmental and biological factors that make predictions unlikely. Oxygen<br />

and saline solutions are the main corrosive factors.<br />

Elemental copper in the mammalian organism is highly reactive and<br />

the resulting ions have biological effects.<br />

In contact with the skin, metallic copper is measurably oxidized by<br />

exudates under formation of derivatives that, at least in animals, penetrate<br />

the integument as measured by the resulting anti-inflammatory activity.


16 Hostynek<br />

In the living organism, corrosion becomes manifest through biological<br />

evidence of contraceptive and spermatocidal action, resulting from the oxidation<br />

process and products of copper IUDs. Conversely, evidence was presented<br />

that in amenorrheic women the copper IUD can restore regular menses.<br />

GLOSSARY<br />

Corrosion Electrochemical process involving movement of<br />

electrons, through a metal from anodic to<br />

cathodic areas, and corresponding movement of<br />

ions in the electrolyte medium.<br />

Electromotive force Difference in driving force toward electron loss<br />

between two metals causing a flow of current,<br />

expressed in volt.<br />

Ionization potential Energy (electron volts, ev) required to remove an<br />

electron from its atomic orbit, with the value for<br />

the standard hydrogen electrode set at 0.00 ev as<br />

an arbitrarily selected standard.<br />

Metallic elements Characterized by luster, malleability, conductivity<br />

(thermal and electrical), and ability to form<br />

positive ions.<br />

Oxidation Process of electron removal from an atom or ion,<br />

or the increase in the proportion of oxygen in<br />

a compound.<br />

Oxidation potential Electrical driving force toward electron loss,<br />

expressed as a potential value (in electron<br />

volts, ev).<br />

Penetration Process of an exogenous agent entering one<br />

skin layer.<br />

ABBREVIATIONS<br />

AA amino acids<br />

L ligand<br />

mEq/L milliequivalents per liter<br />

SC stratum corneum<br />

REFERENCES<br />

1. Sorenson JRJ. In: Siegel A, ed. The anti-inflammatory activities of copper<br />

complexes. New York: Marcel Dekker, 1982.<br />

2. Yano T, Nakagawa A, Tsuji M, Noda K. Skin permeability of various nonsteroidal<br />

anti-inflammatory drugs in man. Life Sci 1986; 39:1043–1050.


Corrosion Chemistry of Copper 17<br />

3. Hadgraft J, du Plessis J, Goosen C. The selection of non-steroidal antiinflammatory<br />

agents for dermal delivery. Int J Pharm 2000; 207:31–37.<br />

4. Walker WR, Keats DM. An investigation of the therapeutic value of the ‘‘copper<br />

bracelet’’—dermal assimilation of copper in arthritic/rheumatoid conditions.<br />

Agents Actions 1976; 6:454–458.<br />

5. Cousins RJ. Absorption, transport, and hepatic metabolism of copper and zinc:<br />

special reference to metallothionein and ceruloplasmin. Physiol Rev 1985; 65:<br />

238–309.<br />

6. Powanda MC. Systemic alterations in metal metabolism during inflammation as<br />

part of an integrated response to inflammation. Agents Actions 1981(suppl 8):<br />

121–136.<br />

7. Linder MC, Hazegh-Azam M. Copper biochemistry and molecular biology. Am J<br />

Clin Nutr 1996; 63:797S–811S.<br />

8. Vinci C, Caltabiano V, Santoro AM, et al. Copper addition prevents the inhibitory<br />

effects of interleukin 1-b on rat pancreatic islets. Diabetologia 1995;<br />

38:39–45.<br />

9. Oster G, Oster GK. Free radical production by metallic copper. Contraception<br />

1974; 10:273–280.<br />

10. Leidheiser HJ. The corrosion of copper, tin and their alloys. New York: John<br />

Wiley and Sons, 1971.<br />

11. Oster GK. Reaction of metallic copper with biological substrates. Nature 1971;<br />

234:153–154.<br />

12. Li S, Miao X, Zhu D, Ni L, Sun C, Wang L. Copper release from copper tableware.<br />

Bull Environ Contam Toxicol 2003; 70:905–912.<br />

13. Broo AE, Berghult B, Hedberg T. Copper corrosion in water distribution<br />

systems—the influence of natural organic matter (nom) on the solubility of<br />

copper corrosion products. Corrosion Sci 1998; 40:1479–1489.<br />

14. Boulay N, Edwards M. Role of temperature, chlorine, and organic matter<br />

in copper corrosion by-product release in soft water. Water Res 2001; 35:<br />

683–690.<br />

15. Reese KM. Verdigris. Chem Eng News 2002; 80:72.<br />

16. Cavelier C, Foussereau J, Massin M. Nickel allergy: analysis of metal clothing<br />

objects and patch testing to metal samples. Contact Dermatitis 1985; 12:<br />

65–75.<br />

17. Lidén C, Carter S. Nickel release from coins. Contact Dermatitis 2001; 44:160–165.<br />

18. Aberer W. Platitudes in allergy-based on the example of the Euro. Contact<br />

Dermatitis 2001; 45:254–255.<br />

19. Aberer W, Kranke B. The new EURO releases nickel and elicits contact eczema.<br />

Br J Dermatol 2002; 146:155–6.<br />

20. Nestle FO, Speidel H, Speidel MO. High nickel release from 1- and 2-Euro coins.<br />

Nature 2002; 419:419.<br />

21. Fournier P-G, Govers TR. Contamination by nickel, copper and zinc during the<br />

handling of Euro coins. Contact Dermatitis 2003; 48:181–8.<br />

22. Lachapelle J-M, Marot L. High nickel release from 1- and 2-Euro coins: are<br />

there practical implications? Dermatology 2004; 209:288–290.<br />

23. Oster GK. Chemical reactions of the copper intrauterine device. Fertil Steril<br />

1972; 23:18–23.


18 Hostynek<br />

24. Sekelj P, Rasmussen K, McDougall D, Baggs J. Survey of electrolytes of unstimulated<br />

sweat from the hand in normal and diseased adults. Am Rev Respir Dis<br />

1973; 108:603–609.<br />

25. Grice K, Sattar H, Casey T, Baker H. An evaluation of Na, Cl and pH ionspecific<br />

electrodes in the study of the electrolyte contents of epidermal<br />

transudate and sweat. Br J Dermatol 1975; 92:511–518.<br />

26. Yousef MK, Dill DB. Sweat rate and concentration of chloride in hand and<br />

body sweat in desert walks: male and female. J Appl Physiol 1974; 36:82–85.<br />

27. Sato K, Feibleman C, Dobson RL. The electrolyte composition of pharmacologically<br />

and thermally stimulated sweat: a comparative study. J Invest Dermatol<br />

1970; 55:433–438.<br />

28. Willing SK, Gamlen TR. Sweat osmolality values in normal adults. Clin Chem<br />

1987; 33:612–613.<br />

29. Jensen O. ‘‘Rusters’’. The corrosive action of palmar sweat: II. Physical and chemical<br />

factors in palmar hyperhidrosis. Acta Derm Venereol (Stockh) 1979;<br />

59:139–143.<br />

30. Hier SW, Cornbleet T, Bergheim O. The amino acids of human sweat. J Biol Chem<br />

1946; 166:327.<br />

31. Coltman CA, Rowe NJ, Atwell RJ. The amino acid content of sweat in normal<br />

adults. J Clin Nutr 1966; 18:373.<br />

32. Gitlitz PH, Sunderman FW, Hohnadel DC. Ion-exchange chromatography of<br />

amino acids in sweat collected from healthy subjects during sauna bathing. Clin<br />

Chem 1974; 20:1305–1312.<br />

33. Liappis N, Kelderbacher S-D, Kesseler K, Bantzer P. Quantitative study of free<br />

amino acids in human eccrine sweat excreted from the forearms of healthy<br />

trained and untried men during exercise. Eur J Appl Physiol 1979; 42:227–234.<br />

34. Fellmann N, Grizard G, Coudert J. Human frontal sweat rate and lactate concentration<br />

during heat exposure and exercise. J Appl Physiol 1983; 54:355–360.<br />

35. Pilardeau PA, Lavie F, Vayasse J, et al. Effect of different work-loads on<br />

sweat production and composition in man. J Sports Med Phys Fitness 1988;<br />

28:247–252.<br />

36. Öhman H, Vahlquist A. The pH gradient over the stratum corneum differs in<br />

X-linked recessive and autosomal dominant ichthyosis: a clue to the molecular<br />

origin of the ‘‘acid skin mantle’’. J Invest Dermatol 1998; 111:674–677.<br />

37. Elias P. Epidermal lipids, barrier function, and desquamation. J Invest Dermatol<br />

1983; 80:44s–49s.<br />

38. Lampe MA, Burlingame AL, Whitney J, et al. Human stratum corneum lipids:<br />

characterization and regional variations. J Lipid Res 1983; 24:120–130.<br />

39. Wertz PW, Swartzendruber DC, Kathi C, Downing DT. Composition and<br />

morphology of epidermal cyst lipids. J Invest Dermatol 1987; 89:419–425.<br />

40. Schurer NY, Elias PM. The biochemistry and function of stratum corneum<br />

lipids. Adv Lipid Res 1991; 24:27–56.<br />

41. Stillman MA, Maibach HI, Shalita AR. Relative irritancy of free fatty acids of<br />

different chain length. Contact Dermatitis 1975; 1:65–69.<br />

42. Feingold KR. The regulation of epidermal lipid synthesis by permeability barrier<br />

requirements. Crit Rev Ther Drug Carrier Syst 1991; 193–210.


Corrosion Chemistry of Copper 19<br />

43. Cage GW, Dobson RL. Sodium secretion and reabsorption in the human eccrine<br />

sweat gland. J Clin Invest 1965; 44:1270–1276.<br />

44. Guyton AC. Textbook of Medical Physiology. Philadelphia: W.B. Saunders Co.,<br />

1991.<br />

45. Boman A, Karlberg AT, Einarsson O, Wahlberg JE. Dissolving of copper by<br />

synthetic sweat. Contact Dermatitis 1983; 9:159–160.<br />

46. Walker WR, Griffin BJ. The solubility of copper in human sweat. Search 1976;<br />

7:100–101.<br />

47. Walker WR, Beveridge SJ, Whitehouse MW. Dermal copper drugs: the copper<br />

bracelet and Cu(II) salicylate complexes. Agents Actions 1981(suppl 8):<br />

359–367.<br />

48. Schmid R, Winkler J. Über die Kutane Kupferresorption aus einer Kupfer<br />

Enthaltenden Salbe. Klin Wochenschr 1938; 17:559–561.<br />

49. Walker WR, Beveridge SJ, Whitehouse MW. Antiinflammatory activity of a dermally<br />

applied copper salicylate preparation (Alcusal). Agents Actions 1980;<br />

10:1–10.<br />

50. Beveridge SJ, Whitehouse MW, Walker WR. Lipophilic copper(II) formulations:<br />

some correlations between their composition and anti-inflammatory/<br />

anti-arthritic activity when applied to the skin of rats. Agents Actions 1982; 12:<br />

225–231.<br />

51. Odintsova NA. Permeability of human skin to potassium and copper ions and<br />

their ultrastructural localization. Chem Abs 1978; 89:360.<br />

52. Walker WR, Reeves RR, Brosnan M, Coleman GD. Perfusion of intact skin by<br />

a saline solution of bis(glycinato) copper(II). Bioinorg Chem 1977; 7:271–276.<br />

53. Jecht EW, Bernstein GS. The influence of copper on the motility of human spermatozoa.<br />

Contraception 1973; 7:381–401.<br />

54. Frentz G, Teilum D. Cutaneous eruptions and intrauterine contraceptive<br />

copper device. Acta Derm Venereol (Stockh) 1980; 60:69–71.<br />

55. Hagenfeldt K. Studies on the mode of action of the copper-T device. Acta Endocrinol<br />

Suppl (Copenh) 1972; 169:3–37.<br />

56. Vesce F, Jorizzo G, Bianciotto A, Gotti G. Use of the copper intrauterine device<br />

in the management of secondary amenorrhea. Fertil Steril 2000; 73:<br />

162–165.


3<br />

Basics of Metal Skin Penetration:<br />

Scope and Limitations<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

INTRODUCTION<br />

The skin as target organ presents imponderable and wide margins of variability.<br />

In vivo, permeability is subject to homeostasis regulating the overall<br />

organism; in vitro, the sections of skin used for diffusion experiments are<br />

likely to present artifacts. To further complicate the matter, diffusion of<br />

metals appears to defy laws empirically derived for passive diffusion across<br />

biological membranes. Endeavors to define rules governing skin penetration<br />

by metals toward derivation of predictive quantitative structure–diffusion<br />

relationships for risk assessment, thus, have been unsuccessful, and penetration<br />

of the skin still needs to be determined separately for each metal compound,<br />

by in vitro or in vivo assays. Because diffusion through biological membranes<br />

is highly metal specific, and, in addition, metal ions’ valence is mutable during<br />

the process of diffusion, molecular physicochemical parameters alone do not<br />

suffice to model migration of electrolytes into and through the strata of the<br />

skin. Certain factors are closely interrelated, and their combined effects are<br />

neither entirely understood nor predictable. For example, unless the dynamics<br />

This chapter, in part, was reprinted from Hosty´nek JJ, Maibach HI. Skin penetration by metal<br />

compounds with special reference to copper. Tox Mech Meth 2006; 16:245–265, with permission<br />

of Informa Healthcare.<br />

21


22 Hosty´nek and Maibach<br />

of in situ changes in speciation (oxidation state), or electrophilic reactivity,<br />

among others, can be factored in, metal diffusivity will elude modeling.<br />

Experimental data available so far from in vitro and in vivo experimentation<br />

have been acquired under disparate conditions, and the base is too thin to<br />

allow development of a predictive algorithm considering the number of metals<br />

and metalloids of variable valence existing as free ions or forming chelates,<br />

coordination compounds, or complexes with electron donors, such as oxygen,<br />

sulfur, or phosphorus active in biological systems. The number of metals<br />

discussed in this review may appear limited; this is due to the circumstance<br />

that most scientists and government agencies so far have given priority to<br />

the limited number of industrial materials, which comport special risks from<br />

work place exposure in their investigations.<br />

Healthy and intact skin was formerly considered to be an impermeable<br />

barrier designed to shield the living organism from environmental<br />

injury, also affording protection from chemical agents. More recently,<br />

however, the skin is being recognized as a membrane, which can act as<br />

filter, impenetrable to microorganisms but selectively pervious to chemicals<br />

within limits of size and polarity, allowing molecular passage in as<br />

well as out of the skin, and acting as a permanent depot or as dynamic<br />

reservoir for essential trace elements (ETEs) and xenobiotics. Only larger<br />

structures (e.g., microorganisms, proteins, and polymers) or highly polar<br />

compounds, such as sugars, will not diffuse measurably through that barrier.<br />

Although exposure to exogenous agents due to skin absorption is<br />

usually less than through inhalation or ingestion, the large surface area<br />

of the body is now recognized as a pharmacologically relevant port of<br />

entry for both nutrients and xenobiotics. Dermal absorption can present<br />

substantial risks in case of exposure to toxins, be they of natural or of<br />

anthropogenic origin. Dermal exposure presenting the potential for<br />

percutaneous absorption has become an integral part of toxicological risk<br />

assessment, especially after incidents of significant morbidity and mortality<br />

following exposure to agents of facile skin diffusivity. Thus, 36 infants<br />

died in France upon exposure to baby talcum powder erroneously formulated<br />

with a high dose of the neurotoxic antimicrobial hexachlorophene<br />

(1–3) or, more recently, to dimethyl mercury, which proved lethal upon<br />

skin contact in trace amounts (4).<br />

Conversely, the skin is also an important secretory organ, which plays<br />

a vital part in the process of detoxification and the maintenance of homeostatic<br />

balance. By that token, on the one hand it is an important factor in the<br />

organism’s clearing mechanism, because it allows elimination of toxics, such<br />

as certain heavy metals, but, on the other hand, it can also lead to significant<br />

elimination of ETEs, such as the detrimental loss of copper through excessive<br />

sweating due to elevated temperature or physical strain. Excretion levels<br />

of six trace metals examined in exercise-induced sweat placed copper in first<br />

place (5,6).


Basics of Metal Skin Penetration: Scope and Limitations 23<br />

Assessment of cutaneous absorption of metal compounds by the use of<br />

new, refined analytical tools is reviewed here. This route of absorption may<br />

be of pharmacological interest in order to compensate for shortages of<br />

ETEs, due to nutritionally caused shortages or genetically caused defects<br />

in intestinal absorption of metals, such as copper. Such shortages in nutrients<br />

may be corrected through cutaneous dosing, as could be shown in<br />

the case of zinc in humans (7–9) and in the rat (10).<br />

It is the objective of this review to identify and describe the numerous<br />

factors that need to be weighed critically when evaluating in vivo or in vitro<br />

data stemming from various sources, and also in planning new such experiments.<br />

Here the term ‘‘metal’’ refers to metals in all its forms of occurrence:<br />

metallic state, electrolytic (ionized) form, and organometallics, complexes,<br />

coordination compounds, or chelates. The term absorption is used to describe<br />

metal penetration of one or several skin strata; penetration signifies metal<br />

becoming available systemically.<br />

This review addresses skin physiology and the various aspects of metals’<br />

skin penetration according to the current scientific knowledge. This includes<br />

discussion of:<br />

1. Structure of the skin and its function as diffusion barrier<br />

2. Descriptors of dermal absorption<br />

3. Permeant categories and paths for their diffusion<br />

4. Metals derivatives formed in contact with skin<br />

5. Variables in skin diffusion by metal compounds<br />

6. Methods for measuring skin permeation<br />

7. Analytical methods for metal detection<br />

STRUCTURE OF SKIN AND ITS FUNCTION<br />

AS DIFFUSION BARRIER<br />

Skin is a major organ involved in the maintenance of the body’s homeostasis:<br />

its composition, heat regulation, blood pressure control, and excretory functions.<br />

It is a multilayered organ, which is a living envelope consisting of<br />

dermis and epidermis, with the stratum corneum (SC) as the outermost layer.<br />

In men, the latter is a 10- to 40-mm thick layer of keratinized epithelial cells,<br />

variable by anatomical site, held together in a ‘‘tongue-and-groove’’ arrangement.<br />

It represents the rate-limiting diffusion barrier for most chemical<br />

compounds (11,12), particularly limiting the loss of water (13). SC thickness,<br />

seemingly a simple measurement, remains at best an estimate. Schwindt<br />

et al. summarize previous efforts, adding indirect measurements in vivo in<br />

men, utilizing transepidermal water loss (TEWL) measurements based on<br />

Kalia’s diffusion equation (14,15). The SC is composed of layers of proteinaceous<br />

cells separated by layers of lipid material. In humans it consists of<br />

about 40% protein, 15% to 20% lipids, and 40% water (16). Likened to a


24 Hosty´nek and Maibach<br />

brick-and-mortar structure, the SC consists of corneocytes (bricks) embedded<br />

in an intercellular matrix of lipids (mortar), rendering the membrane poorly<br />

permeable to water and other polar compounds (17). Interspersed in this<br />

envelope are appendageal shunts, openings for hair follicles, and sweat<br />

ducts. Underlying the SC are the strata of the avascular viable epidermis,<br />

consisting of keratinocytes, the source of corneocytes. The epidermis is also<br />

transversed by shunts: the hair follicles and sebaceous and sweat glands. In<br />

ascending order, the human epidermis consists of the stratum basale,<br />

responsible for the maintenance of epidermal–dermal adhesion. As cells<br />

move outward from the basement membrane, in the process of differentiation<br />

they undergo keratinization, transition to the stratum spinosum, then<br />

further to the stratum granulosum. The latter consists of electron-dense<br />

keratohyalin granules, which are irregularly shaped, nonmembrane-bounded,<br />

electron-dense granules that contain profilaggrin, a structural protein, and a<br />

precursor of filaggrin that plays a role in keratinization and barrier function.<br />

The uppermost layers of the stratum spinosum and stratum granulosum<br />

contain small membrane-bounded organelles known as lamellar granules,<br />

Odland bodies, lamellated bodies, or membrane-coating granules. These<br />

are the granules that contain lipids that can fuse to the cell membrane to<br />

release its lipid content that will fill the intercellular space (18). The stratum<br />

granulosum is also source of corneocytes, which, keratinized, form the SC,<br />

the outermost layer of dead cells constituting an efficient barrier against<br />

transcutaneous water loss. Besides generating the SC, other important functions<br />

of the epidermis are metabolism of xenobiotics, synthesis of melanin by<br />

melanocytes, and provision of a first-line immune defense by means of dendritic<br />

Langerhans’ cells. In its entirety, the epidermis with the intercellular<br />

lipids between the SC layers constitutes the rate-limiting barrier to the<br />

absorption of both hydrophilic and lipophilic xenobiotics. Transition from<br />

the lipophilic SC to the hydrophilic medium of the epidermis constitutes a<br />

discontinuity that forms an important second barrier, largely inhibiting<br />

further passage of lipophilic compounds once they have transited through<br />

the SC. In its entirety, from skin surface to circulatory system the epidermis<br />

consists of aqueous and lipid barriers. Removal of the hydrophilic epidermal<br />

layer in in vitro diffusion experiments enhanced the percutaneous absorption<br />

of lipophilic compounds across the SC, because it no longer functions as that<br />

rate-limiting secondary barrier for the diffusion of lipophilic compounds<br />

across the skin (19).<br />

The support system for the epidermis is the underlying dermis, carrier<br />

of nutrition through its vascular network; it is also involved in the immune<br />

function through the lymphatic system, macrophages, and the mast cells<br />

responsible for immune and inflammatory responses. It produces elastic<br />

connective tissue consisting of collagen, laminin, and fibronectin, among<br />

other components, and is the origin of sebaceous glands, eccrine and apocrine<br />

sweat glands, and hair follicles.


Basics of Metal Skin Penetration: Scope and Limitations 25<br />

DESCRIPTORS OF DERMAL ABSORPTION<br />

Mostly, but not exclusively, description of dermal absorption is based on<br />

two approaches:<br />

The permeability coefficient, Kp, usually obtained through in vitro<br />

experiments for compounds from water where it can be measured<br />

directly<br />

Percent absorbed as fraction of dose applied in vivo<br />

The Permeability Coefficient K p<br />

The permeability coefficient Kp—a flux value, normalized for concentration—<br />

is a widely applied benchmark, representing the rate at which chemicals<br />

penetrate the skin. It is derived by Fick’s law of diffusion (20) from flux measurements<br />

through a biological barrier under conditions of steady state (J ss)<br />

from an infinite reservoir of the compound, through a biological barrier,<br />

and normalized for the concentration (C) applied: Jss ¼ Kp DC, e.g.,<br />

expressed as cm/hr. Formulated to characterize passive diffusion of compounds<br />

across membranes in general, Fick’s law also applies to passive<br />

diffusion of xenobiotics through the SC of the skin. The law states that<br />

if the permeation process reaches the point of steady-state equilibrium (i.e.,<br />

the concentrations in the donor and receptor phases remain constant over<br />

time), the steady-state penetration flux J ss per unit path length (here in cm)<br />

is proportional to the concentration gradient dC and to the penetrant’s<br />

permeation constant.<br />

In practice, in a typical experiment the donor solution is scaled in such<br />

a fashion that over the term of the experiment the concentration remains<br />

‘‘practically’’ constant (a.k.a. ‘‘infinite’’ dose); because the receptor phase<br />

is constantly removed, receptor concentration equals zero. Thereby, the<br />

expression DC becomes equal to C, the concentration in the donor solution,<br />

and the equation is simplified as<br />

Jss ¼ Kp C or Kp ¼ Jss=C<br />

The technique, easily standardized, allows the determination of Kp<br />

through skin or other membranes as long as the barrier properties are not<br />

affected by either permeant or carrier solvents. This implies that the permeant<br />

may not react with the barrier material (i.e., change barrier permeability with<br />

time). Because several heavy metals react with epidermal protein to some<br />

degree, however this somewhat prejudices the general validity of K p values<br />

(see section on variables).<br />

Expressing depth of diffusion per unit of time, the Kp provides a basis for<br />

comparison of the relative absorption rates of diverse chemicals. When the<br />

permeant is applied from an aqueous solution, as is the case for most metal


26 Hosty´nek and Maibach<br />

compounds, it also represents the method by which most compound-specific<br />

penetration data currently available have been generated, for drugs as well as<br />

hazardous pollutants (21,22).<br />

The Kp, here expressed in cm/hr, is the most convenient parameter for<br />

comparison of percutaneous penetration for purposes of dermatopharmacokinetics<br />

and dermatotoxicology. Values for metal derivatives range from<br />

7 10 7 cm/hr for nickel dichloride (23) to 2.1 10 3 cm/hr for sodium<br />

dichromate (24) through human skin.<br />

Discrepancies in Kp values reported by different investigators often<br />

depend on whether mass balance was part of the study (i.e., whether the results<br />

included material remaining in the barrier material at the end of the study).<br />

For both dermatotoxicological and dermatopharmacological purposes, such<br />

materials should be considered part of overall absorption unless it is<br />

ascertained that the permeant does not diffuse further to reach the systemic<br />

circulation. Transition metals especially tend to react with SC protein and<br />

are retained in the outer strata, sometime to a degree significantly exceeding<br />

amounts reaching the receptor phase.<br />

Percent of Dose Absorbed<br />

Relevant for purposes of risk assessment is the total amount of chemical systemically<br />

absorbed. By that token it appears more important to determine<br />

the amount which has disappeared from the site of application (surface<br />

of the skin) and which eventually will flux through the skin and into the<br />

organism and not the amount which is collected in the receptor phase in vitro<br />

over the (necessarily) limited amount of time of the experiment. It thus<br />

appears important that, whenever possible, absorption data be determined<br />

in vivo, in men or primates, for as accurate a risk assessment as possible,<br />

avoiding the uncertainties of cadaver skin or adjustments for species variation.<br />

One principal in vivo methodology, using human volunteers or monkeys, is<br />

the one developed by Feldmann and Maibach (25–27) and is the most applied<br />

in vivo method for the determination of skin absorption, yielding much of<br />

the data so far available on skin penetration by drugs and pesticides.<br />

In human studies, following topical application, plasma levels of test<br />

compounds are low, often falling below assay detection, and so it becomes<br />

necessary to use radiolabeled chemicals. The compound labeled with<br />

carbon-14 or tritium, or a metal isotope, is applied to the skin in a minimal<br />

volume of a volatile solvent that is left to evaporate, and the total amount of<br />

radioactivity (RA) excreted is then determined. The amount retained in the<br />

body is corrected for by determining the amount of RA excreted following<br />

parenteral administration. The resulting RA value is then expressed as the<br />

percent of the applied dose absorbed. Fick’s postulates for membrane diffusion<br />

are not met there because a concentration of the penetrant cannot be<br />

defined and neither is a steady-state equilibrium reached with this method;


Basics of Metal Skin Penetration: Scope and Limitations 27<br />

thus a permeability coefficient characterizing the compound can only be<br />

calculated through prior conversion as given below, yielding an ‘‘apparent’’<br />

Kp. When diffusion experiments are reported as the percent of the applied<br />

dose, disappearance over a time interval t is given either as [% lost/(time)],<br />

or as a first-order disappearance constant, k (min 1 ), for that time interval.<br />

Assuming steady-state conditions, either of these parameters can then be<br />

converted to the disappearance over a time interval t and is given either<br />

as [% lost/(time)], or as a first-order disappearance constant, k (min 1 ),<br />

for that time interval. Assuming steady-state conditions, either of these<br />

parameters can then be converted to the permeability coefficient, Kp. That is<br />

Kp ¼ J ð% lossÞ<br />

¼<br />

DC ½timeðhrÞŠ<br />

CA VA<br />

¼<br />

100 A CA<br />

ð% lossÞ<br />

½timeŠ<br />

VA<br />

100 A<br />

where J (mol/cm 2 /hr) is the chemical flux, C (mol/L) is the chemical concentration<br />

gradient across the skin (which is reasonably assumed to be equal<br />

to the applied concentration C A), V A (mL) is the volume of chemical solution<br />

applied, and A (cm 2 ) is the area of application.<br />

It follows, therefore, that, for a five-hour application of 1 mL of solution<br />

to a 3.14 cm 2 area of skin<br />

Kp;5 ¼ ð% lossÞ 5<br />

1570<br />

Kp ¼ J ðfraction lostÞ<br />

¼<br />

DC ½timeðhrÞŠ<br />

CA VA<br />

A CA<br />

where, assuming first-order disappearance kinetics (characterized by rate<br />

constant, k, min 1 ),<br />

ðfraction lostÞ ¼1 expf 60 k ½timeðhrÞŠg<br />

Thus, for a one-hour application of 1 mL of solution to 3.14 cm 2 of<br />

skin, Kp is calculated as<br />

Kp ¼ð1 expð 60 kÞÞ=3:14<br />

Together with the toxicity of a chemical the percent of a substance<br />

absorbed is the second most critical factor for risk assessment; it gives a measure<br />

for human exposure and an indication of the amount potentially<br />

absorbed in (the worst) case of total body immersion. For a chemical applied<br />

at 100 mg/cm 2 , at 1% absorption, for instance, the extent of systemic exposure<br />

to the compound applied on the entire body area of an average human<br />

adult (18,000 cm 2 ) would be in the milligram range in the course of 24 hours.<br />

ð1Þ<br />

ð2Þ


28 Hosty´nek and Maibach<br />

PERMEANT CATEGORIES AND PATHS OF DIFFUSION<br />

Several routes are available for the diffusion of compounds into and through<br />

the matrix of the skin. In order to facilitate a closer discussion of the alternatives,<br />

it is useful to associate them with the different categories of<br />

permeants as they transit alternate domains of the skin.<br />

Organic Compounds (Nonelectrolytes)<br />

The lipid matrix, which cements the corneocytes of the SC, is believed to be<br />

the principal route of transport for nonelectrolytes and lipophilic (organic)<br />

compounds into the deeper layers of the integument. Most extensively investigated<br />

so far is the process of intercellular diffusion by compounds from the<br />

categories of drugs, pesticides, organometals, and cosmetic materials,<br />

including fragrances. On first approximation, rate of penetration appears<br />

commensurate with compound polarity, measured as the octanol/water<br />

partition coefficient Poct, or its logarithm, log P (vide infra). Molecular<br />

transport of compounds has been described by mathematical models derived<br />

through multiple regression analysis of percutaneous absorption data and<br />

physicochemical constants of a large number of structures. Several mathematical<br />

models descriptive of molecular transport through human skin,<br />

in particular, have been developed more recently (21,28–32). Scheuplein<br />

et al. were among the first to describe an anatomically accurate and physicochemically<br />

reasonable model of the skin, and went on to demonstrate<br />

the correlation between lipophilicity (polarity) and experimental permeability<br />

coefficient values Kp of permeants in vitro (12,18,33,34). Polarity is most<br />

often expressed as the compound’s partition coefficient in equilibrium<br />

between n-octanol and water, Poct. Thanks to these modeling efforts, it is<br />

now possible to predict diffusion of nonelectrolytes through the skin as a<br />

model membrane with some accuracy, simply based on a few physicochemical<br />

parameters that characterize the permeant. Such models are useful in<br />

predicting the activity of chemicals in the absence of experimental data on<br />

a particular compound, without the need for its actual synthesis. Set in<br />

relation to the corresponding in vivo data, the predictive power of these<br />

models, known as quantitative structure–activity relationships (or QSAR),<br />

demonstrates that (i) three descriptors (size, polarity, and hydrogen bonding)<br />

are the principal determinants for cutaneous transport, and (ii) the<br />

properties of the intercellular SC lipids alone are sufficient to characterize<br />

the barrier properties of the skin as the route of permeation for that category<br />

of chemical structures (32).<br />

Rougier et al. developed a brief experimental method to predict skin<br />

absorption of lipophilic compounds. By that method, the systemic uptake<br />

of permeants can be derived from the amount that diffuses into the SC in<br />

vivo after a limited time of exposure (35). The chemical is dosed on the skin<br />

of animals or human volunteers, and after 30-minute-contact the surface of


Basics of Metal Skin Penetration: Scope and Limitations 29<br />

the SC wiped clean of residual compound, and the SC removed by successive<br />

application of adhesive tape and stripping. The strippings are analyzed<br />

for chemical adhering to tissue removed, and from that value the amount<br />

may be estimated that eventually will be systemically absorbed over a longer<br />

period of time.<br />

Certain metal compounds (organometallics) also represent a lipophilic<br />

category (36) that can penetrate the SC with relative ease. Such derivatives<br />

of the more toxic heavy metals, therefore, represent a major toxicological<br />

risk, particularly in the work place, due to their ready skin penetration.<br />

Representative Kps are 2.9 10 3 cm/hr for methyl 203 mercury dicyanamide<br />

50 and 2.3 10 3 cm/hr for lead naphthenate 51 .<br />

Electrolytes<br />

In our present understanding of structure and function of the skin, its permeation<br />

is determined by the physicochemical parameters of permeants.<br />

Intuitively, and also based on the therapeutic action of dermatologicals<br />

and cosmeceuticals, or the toxic effect of skin exposure to pesticides, we<br />

anticipate easy penetration by lipophilic compounds there, as is achieved<br />

in the process of inunction. But for polar structures, such as water or electrolytes<br />

(e.g., salts in aqueous solution), skin penetration to any significant<br />

degree is often dismissed because the skin appears designed as a barrier<br />

against external disturbance of a natural state of hydration. Still, for water<br />

and a number of hydrophilic-charged molecules, including a number of<br />

metal complexes, diffusion is also demonstrated, albeit the process proceeds<br />

at an average of two to three orders of magnitude more slowly than is the<br />

penetration of small-molecular-weight, lipophilic nonelectrolytes, where<br />

Kp values are on the order of 1 cm/hr (e.g., for the solvent toluene).<br />

Skin appendages or shunts (the hair follicles and sweat ducts which<br />

transit through all those layers) are considered the predominant pathway<br />

for the diffusion of large polar molecules or electrolytes across the skin as<br />

an early stage event. They constitute relatively large openings through which<br />

diffusion can occur if two principal conditions are met:<br />

1. Sweat is present to serve as an aqueous diffusion medium<br />

2. the outflow of sweat is significant (18,37)<br />

Measuring electric resistance to analyze for the diffusion pathways<br />

taken by charged molecules, Mali et al. calculated that 70% of the<br />

strong electrolytes permeate through sweat ducts (38). High current flow<br />

in iontophoresis confirmed that observation (12,30,39–43), as did use of<br />

autoradiography or microparticle-induced X-ray emission (PIXE) analysis<br />

(44,45). Considerable experimental data have indicated that the depth of<br />

sweat duct penetration is significantly dependent on ionic mobility (i.e., size)<br />

and electronic charge of the metal (46,47).


30 Hosty´nek and Maibach<br />

Direct or indirect observations speak to the ease of such electrolyte diffusion<br />

through skin shunts. Penetration through sweat ducts can occur<br />

within one to five minutes following exposure, with no comparable transport<br />

occurring via the transcellular path in that time span (48–50). Poral<br />

transit by nickel salts, for instance, can also become manifest clinically;<br />

follicular inflammation or punctate erythema were observed in the context<br />

of skin patch testing with nickel salts (51).<br />

The relatively rapid flux across the appendageal pathway is followed<br />

by slower but continuous, potentially more important intercellular diffusion.<br />

It is reasonable to postulate that metal ion pairs (soaps) formed with<br />

fatty acids on the skin surface will preferably partition into the lipophilic<br />

environment. The intercellular lipid domains in the SC seem to present a<br />

ready pathway for diffusion of such compounds, because the relatively<br />

slower, transcellular rate of penetration does not explain such phenomena<br />

as provocation of allergic reactions due to casual contact with metallic<br />

objects, such as coins.<br />

Occupational health statistics on untoward health effects are a prime<br />

indication for the skin diffusivity of noxious electrolytes. With respect<br />

to specific (allergic) and nonspecific (irritant) contact dermatitis that may<br />

constitute up to 90% of workers’ compensation claims for skin diseases,<br />

percutaneous absorption of potential allergens and irritants is a key determinant<br />

of the risk of skin sensitization and irritation, and may be enhanced<br />

if protective clothing entraps or occludes the irritant against the skin, by<br />

increased hydration of the SC, and by contact with anatomical sites where<br />

skin permeability is greater. Thus, exposure to nickel, chromium, cobalt,<br />

and mercury compounds are premier causes for allergic and irritant reactions<br />

in industrialized countries (52).<br />

The route of transcellular diffusion may be of marginal importance,<br />

especially for electrophilic transition metals that tend to form permanent<br />

deposits on the skin surface. Absorption would be limited to the outermost<br />

layers of the SC, and, possibly, the epithelium of appendages. Such adsorption<br />

may also be terminal, resulting in the depot formation repeatedly<br />

described in the literature for a number of electrophilic metals (53,54). Such<br />

non-Fickian behavior clearly puts that class of compounds beyond the scope<br />

of theoretical algorithms predictive of percutaneous penetration, and the<br />

fate of chemicals thus retained in the SC remains uncertain, prompting<br />

the question: Will the compounds continue to diffuse slowly to be ultimately<br />

absorbed systemically, or will they be shed with the corneocytes in the<br />

process of desquamation? Studies with lipophilic compounds in vivo and in<br />

vitro show that within weeks most of the xenobiotics initially retained in the<br />

SC have been absorbed systemically or have reached the receptor fluid,<br />

respectively. Rougier’s predictive method is applicable only to compounds<br />

of Fickian behavior, but not to electrophilic metals that react with barrier<br />

tissue, rendering in-depth diffusion unpredictable (55). In addition, certain


Basics of Metal Skin Penetration: Scope and Limitations 31<br />

metals, such as copper, zinc, and chromium, are essential nutrients subject<br />

to homeostatic controls, which appear to determine deposition or mobilization<br />

in response to fluctuating body burdens.<br />

Highly protein-reactive, electrophilic metals, such as mercury, to a<br />

large extent were seen to remain at the site of initial absorption. Hursh et al.<br />

measured the uptake of mercury vapor by the skin on human arms in vivo<br />

(56). The rate determined corresponded to a high permeability coefficient<br />

(K p of 1–2 cm/hr). As much as half of the absorbed mercury, however,<br />

appeared to have reacted with SC protein and was eliminated through the<br />

process of desquamation in the following weeks. In that experiment,<br />

the forearm was exposed to mercury vapor enclosed within a saran bag.<br />

On average, 6.8% (range: 3.0–10.6%) of the Hg vapor originally in the exposure<br />

chamber was absorbed by the arm. Up to half the mercury initially<br />

in the forearm was shed by desquamation of epidermal cells during several<br />

weeks. The remainder diffused into the general circulation and could be<br />

measured as systemic mercury. For two subjects on the day following exposure,<br />

the SC was collected from a 35 cm 2 area of the forearm by stripping<br />

off the superficial layers with adhesive cellophane tape. The 203 Hg measured<br />

on the tape, normalized for the total exposed area, corresponded to only<br />

0.3% and 1.3% of the Hg on the arm at that time. When the process was<br />

repeated for one subject 14 days later, and again 23 days after exposure,<br />

the additional recoveries normalized to the entire exposed area were 24%<br />

and 10.7%, respectively, reflecting gradual outward migration of keratinocyte<br />

and corneocyte carriers of the irreversibly bound mercury. The authors<br />

concluded that, due to protein reactivity, absorption of mercury vapor by<br />

the skin poses a minor occupational hazard when compared with inhalation.<br />

The three modes of diffusion available to xenobiotics also illustrate the<br />

characteristics of the skin, which can function to varying degrees as a barrier,<br />

a reservoir, and/or a filter, depending on the polarity and chemical reactivity<br />

of the solute. Topically applied compounds can penetrate the SC along more<br />

than one pathway simultaneously, albeit at different rates. A molecule can<br />

follow any of these routes—follicular, transcorneal, or intercellular—but it<br />

is difficult to assess the relative importance of each, as this will vary with<br />

the physicochemical nature of the molecule.<br />

COMPOUNDS FORMED BY METALS IN CONTACT<br />

WITH THE SKIN<br />

Direct and prolonged contact of metals and alloys with the skin may result<br />

in electrochemical reactions that release metal ions. Indicative of the process<br />

of skin penetration by certain metals is the onset of skin reactions, mostly<br />

immunological: contact with coinage, tools, jewelry, or articles of daily<br />

use. The majority of allergic reactions to metals involve nickel (57–64)<br />

and chromium (65–73).


32 Hosty´nek and Maibach<br />

Leaching or release of metals from alloys in contact with body fluids,<br />

such as sweat or sebum, is difficult to predict with accuracy. Aside from<br />

immediate environmental factors, the microenvironment within the particular<br />

alloy is a principal determinant, due to the action of electromotive forces<br />

generated by the presence of other metals (37). Review of published data on<br />

corrosion shows that release rates, as determined through leaching tests, do<br />

not correlate with metal content in an alloy (74). Associated metals in<br />

immediate proximity form a galvanic element (or pile), whereby an electron<br />

current flows from the more electronegative (e.g., nickel) to the nobler, more<br />

electropositive one (e.g., copper), resulting in oxidation or solution (‘‘corrosion’’)<br />

of the more electronegative metal. Corrosion is an electrochemical<br />

process involving movement of electrons (e ) through the metal from anodic<br />

to cathodic areas and related movement of ions in the electrolyte (sweat). At<br />

metal potentials and pH values that occur in sweat, anodic reactions for<br />

some metal constituents in alloys include<br />

Ni ! Ni 2þ þ 2e ; Cu ! Cu 2þ þ 2e ; Fe ! Fe 2þ þ 2e ; Cr ! Cr 3þ þ 3e<br />

The electrons generated are then consumed at the cathode, most<br />

commonly by reaction with oxygen:<br />

O þ H2O þ 2e ! 2ðOHÞ<br />

Whether and how much nickel is mobilized will depend on the immediate<br />

surrounding material (e.g., copper) present in the alloy. The products of<br />

corrosion may then enter the organism, taken up through the skin, through<br />

the oral mucosa, or by the gastrointestinal (GI) tract (75). Transported by<br />

blood throughout the organism, copper, in particular, will be sequestered<br />

by metallothionein (MT) or coeruloplasmin, and deposited in various<br />

organs and tissues (76).<br />

In order to discuss the phenomenon of skin penetration by metal compounds<br />

as ionized salts, complexes, or organometallic compounds, and to<br />

correctly interpret the meaning of experimental results and clinical observations<br />

attributed to the action of metals, it is important to visualize the<br />

structure and function of the skin as a membrane, which can act as barrier,<br />

filter, reservoir, or an outright port of entry, and the microenvironment prevailing<br />

on its surface. Most direct evidence for formation of skin-diffusible<br />

oxidation products on skin contact is the facile elicitation of allergic reactions<br />

by elements, such as nickel (77), and detection of that metal deep in<br />

the SC upon skin contact (78).<br />

The chemical environment encountered on the skin surface explains<br />

the process of solubilization and, subsequently, diffusion upon contact of<br />

metals in their elemental state. By the action of salts and acids present in<br />

sweat and sebum, metals can be converted to a hydrophilic or lipophilic<br />

derivative, respectively. Only then do they become diffusible via the transcellular,<br />

intercellular, and transappendageal route. Skin exudates are secretions


Basics of Metal Skin Penetration: Scope and Limitations 33<br />

of varying composition, covering the epidermis with a film that complements<br />

the skin’s barrier function. That film consists of two components of<br />

endogenous origin, sweat and sebum, secreted by the respective glands,<br />

which have the ability to corrode (oxidize and dissolve) metal surfaces on<br />

contact. Their composition varies in function of physical, pharmacological<br />

and environmental conditions, gender, age, sweat rate, body site, and<br />

method of collection. An abundance of eccrine sweat glands occurs widely<br />

distributed over all exposed skin areas, to dissipate body heat through evaporation.<br />

Besides these occur sebaceous sweat glands, closely associated with<br />

hair follicles, which produce an oily secretion, the sebum (79). The pathways<br />

that oxidation products follow in the process of diffusion through the skin will<br />

depend on the polarity of the salts formed with exudates, and can be predicted<br />

in light of earlier investigations of skin penetration by xenobiotics. Metal<br />

salts in dissociated ionic form do not penetrate the skin as readily as their<br />

unionized (e.g., organometallic) form. Being unionized, they are more lipid<br />

soluble and that seems to be the decisive factor for intercellular, relatively<br />

facile diffusion. Organic salts may penetrate in the form of ion pairs, and also<br />

exhibit relatively high fluxes. For a molecule that is dissociable in water, the<br />

dissociation constant and pH of the immediate environment will determine<br />

degree of ionization and, thus, permeability. Some organometallic compounds,<br />

such as derivatives of lead (80), mercury (81), or nickel (unpublished<br />

data), have actually been characterized as relatively good skin penetrants.<br />

Copper metal reacting with fatty acids may thus also form lipophilic compounds<br />

of marked diffusivity.<br />

Sweat<br />

Values for the main components of sweat have repeatedly been investigated<br />

over time, yielding increasingly accurate data reflecting improvements<br />

in analytical techniques. The main categories of solutes are discussed here.<br />

Listed in Table 1 are the prevalent ranges of eccrine sweat; they are approximations,<br />

as values are unavoidably subject to variation even in normal<br />

Table 1 Mean Levels of Eccrine Sweat Components<br />

Sodium Men 51.9 mEq/L<br />

Women 36.5 mEq/L<br />

Potassium Men 7.5 mEq/L<br />

Women 10.0 mEq/L<br />

Chloride 29.7 mEq/L<br />

Urea 260–1220 mg/L<br />

Lactic acid 360–3600 mg/L<br />

Amino acids 270–2590 mg/L<br />

Ammonia 60–110 mg/L<br />

Source: From Ref. 82.


34 Hosty´nek and Maibach<br />

subjects, due to the type of stress applied to sweat stimulation, ambient and<br />

body temperature, environmental humidity, diet and nutritional status, age,<br />

gender, sweat rate, skin area of collection, local skin temperature, muscular<br />

activity, etc.<br />

The validity of these values in adults is often questioned, however, due<br />

to differences in methods of analysis and sweat stimulation (physiological,<br />

physical, or pharmacological). The main cause for corrosion of metal surfaces<br />

from skin contact in individuals referred to as ‘‘rusters’’ is not due to<br />

elevated electrolyte concentration, as generally assumed, but rather seems<br />

to coincide with palmar hyperhydrosis in those individuals. ‘‘Rusters’’ exude<br />

palmar sweat at inordinately high rate and a low pH, with the pronounced<br />

corrosive action on metal surfaces observed. When the sodium concentration<br />

measured in normal subjects was compared to that of ‘‘rusters,’’ no<br />

significant difference could be observed (mean values of 49.6 mEq/L vs.<br />

49.1 mEq/L, respectively) (83). However, the pH of sweat was measured<br />

to range between 2.1 and 6.9 (57).<br />

Both proteins and amino acids are normal components of mammalian<br />

sweat (84). Quantitative analysis of amino acids has become routinely possible,<br />

thanks to ion-exchange chromatography; however, the data documented<br />

for men by authors differ significantly, probably due to differences in the<br />

stimulation methods applied, to regional differences in anatomical site, or<br />

to the methods used for sampling. Substantial variations were observed in<br />

relative concentrations of amino acids in sweat collected from various body<br />

parts. Amino acids excreted in sweat are independent of dietary intake (85),<br />

although their concentrations increase markedly in blood and urine on oral<br />

protein intake. No differences in amino acid patterns were seen between<br />

young and middle-aged adults, or between men and women (86).<br />

Sebum<br />

Acid components making up the sebum also play an important role in solubilizing<br />

(‘‘corroding’’) metal surfaces. Human skin features an acid mantle of<br />

pH 4 to 6 at the surface of the SC, which increases with depth to pH 7 at its<br />

juncture with live tissue (87). Determinants of this pH are protons that originate<br />

in the epidermis or as products of sebaceous gland activity, gradually<br />

reaching the surface of the skin. They stem from three classes of compounds:<br />

Amino acids (e.g., urocanic acid and pyrrolidone carboxylic acid)<br />

Alpha-hydroxy acids (e.g., lactic and butyric acid), also present in<br />

sweat<br />

Acidic lipids (e.g., cholesteryl sulfate and free fatty acids, primarily<br />

oleic and linoleic) (17,58,88)<br />

At its point of origin in the live epidermis, sebum as secreted by the<br />

sebaceous glands is a complex mixture of lipids consisting of glycerides,<br />

but no free fatty acids.


Basics of Metal Skin Penetration: Scope and Limitations 35<br />

The occurrence of free acids in the SC and on the skin surface is the<br />

result of hydrolysis of phospholipids and glycerides by lipolytic enzymes<br />

occurring in the sebaceous ducts and on the skin surface, and of bacterial<br />

decomposition. On the skin surface, lipids of epidermal origin contain up<br />

to 20% free fatty acids, with 16% originating in the pilosebaceous glands<br />

(58,88). They consist, to the greater part, of C16 and C18 acids, but their<br />

full range reaches from C5 to C22, with an average length of C16. Such<br />

an acid environment plays a regulating role for SC homeostasis with<br />

relevance to the integrity of the barrier function and regeneration of the<br />

SC barrier (89). It is now accepted that the acid environment on the skin<br />

surface supports a number of SC functions (90), some of which are<br />

control of moisture loss from the epidermis and permeability<br />

barrier homeostasis,<br />

providing resistance to fungal and bacterial infection,<br />

SC integrity and cohesion, and<br />

enzymatic processes regulating corneocyte desquamation.<br />

VARIABLES DETERMINING SKIN DIFFUSION OF<br />

METAL COMPOUNDS<br />

Skin penetration by metals is a multifactorial process, which is not fully<br />

understood, and even less predictable. Contributing to the uncertainty are the<br />

effects of chemical speciation of metallic elements, particularly that of<br />

the transition metals. Also, the skin as target organ presents imponderable<br />

and wide variability. Particularly with ETEs, numerous factors come to bear<br />

(e.g., in vivo, permeability can be subject to homeostasis regulating the overall<br />

organism; in vitro, the sections of skin used for diffusion experiments can<br />

present artifacts). Mathematically derived (mechanistic) models predictive<br />

of percutaneous penetration of organic molecules have been developed<br />

based on in vitro and in vivo data through regression analysis of experimental<br />

results, making the estimation of diffusivity of untested structures<br />

possible. However, similar reduction to a few molecular physicochemical<br />

parameters is not feasible when modeling electrolytes, because movement<br />

through biological membranes is highly element specific. A number of factors<br />

are closely interrelated, and their combined effects are not predictable.<br />

A detailed, in-depth review of parameters determining skin diffusivity of<br />

a large number of metals is given by Potts and Guy (32).<br />

Exogenous Factors<br />

Dose<br />

Rate of diffusion of certain transition metals is not commensurate with<br />

applied concentration. With increasing doses, absorption rates of certain<br />

transition metals can reach a plateau value, then decrease with a further


36 Hosty´nek and Maibach<br />

increase in applied concentration. For others, absorption steadily decreases<br />

with increasing dose. This is likely due to the buildup of a secondary diffusion<br />

barrier as a consequence of electrophilic metals forming stable bonds<br />

with proteins of the skin. Thereby, a depot accumulates in the SC, retarding<br />

further penetration in inverse proportion to metal concentration. In vitro,<br />

this leads to lag times that can be as long as days before any permeant is<br />

collected in the receptor compartment, an observation made mainly with<br />

salts of nickel, chromium, and mercury (54,91,92).<br />

An example of non-Fickian behavior is the diffusivity of nickel salts.<br />

Fluxes of NiCl2 and NiSO4 through full-thickness human skin were compared<br />

by Fullerton et al. (93). After lag times of about 50 hours, in experiments<br />

lasting 144 to 239 hours, occluded NiCl2 entered the receptor fluid about<br />

5 to 40 times more rapidly than (i) NiSO4, (ii) NiCl2 with added Na2SO4,<br />

or (iii) NiSO4 with added NaCl. Without occlusion, the permeation of nickel<br />

was reduced by more than 90%, an indication of permeability increasing<br />

with hydration.<br />

Application of chromate with increasing concentrations does not<br />

consistently lead to higher penetration rates. With chromate doses at concentrations<br />

of between 0.00048 and 0.73 M in guinea pig skin in vivo, absorption<br />

of chromate increased to a maximum Kp of 2.64 10 3 at 0.26 M dose,<br />

reached a plateau, and then gradually decreased below detectable levels (94).<br />

Determined with human abdominal epidermis in vitro, the diffusion<br />

coefficient for chromate ion consistently decreased with increasing donor<br />

concentration (95).<br />

With human full-thickness abdominal skin in vitro, permeability coefficients<br />

for dichromate were also inversely proportional to the concentrations<br />

applied (96).<br />

In guinea pigs, absorption of mercuric chloride applied in vivo reached<br />

a maximum at 16 mg Hg/mL, then decreased to nondetectable levels with<br />

increasing concentrations (59).<br />

Vehicle<br />

The solvent can have a profound effect on permeant solubility and on the<br />

skin membrane, and thus on its barrier properties. Petrolatum, for instance,<br />

is a poor solvent for metal salts, where the permeant remains suspended<br />

in fine particles, affording less-than-ideal uniformity in skin contact; on<br />

the other hand, it has an occlusive effect, which increases skin hydration<br />

and thus promotes diffusion of a hydrophilic compound. Quantitative<br />

absorption of zinc in human skin in vitro showed a distinct dependence<br />

on vehicle. The permeability coefficients comparing fluxes from petrolatum<br />

and those from a hydrogel containing zinc chloride were based upon<br />

72-hour periods. The permeability coefficients for 2.4% zinc formulated in<br />

petrolatum was 0.082 10 4 cm/hr. From the hydrogel containing the chloride<br />

salt, the permeability coefficient was 0.29 10 4 cm/hr (i.e., more than


Basics of Metal Skin Penetration: Scope and Limitations 37<br />

three times higher) (60,61). Zinc oxide was applied on intact human skin<br />

in gum rosin versus a hydrocolloid carrier; after 48 hours penetration of zinc in<br />

human epidermis was more than twice the level as compared to the hydrocolloid<br />

vehicle (62). Dimethyl sulfoxide is a solvent that enhances skin<br />

penetration, as it causes swelling of basal SC cells and disrupts the normal<br />

keratin pattern. Dimethyl formamide and dimethyl acetamide applied in<br />

that solvent were seen to diffuse at a significantly accelerated rate (63).<br />

Of practical importance also is the choice of vehicle in standard<br />

diagnostic skin patch testing for sensitization, with the aim of optimum<br />

release of allergen into the viable epidermis while avoiding allergic or irritant<br />

contact dermatitis, leading to false-positive reactions caused by the vehicle<br />

itself. A method for determining the optimal solvent for diagnostic skin<br />

patch testing of allergens, with the intent of minimizing false-negative reactions,<br />

is their application in different solvents on hypersensitive patients. By<br />

recording the ratio of positive elicitation, the solvent can be identified that<br />

better promotes skin diffusion of the xenobiotic. The reaction threshold<br />

to nickel sulfate was tested in 53 sensitized patients in both water and<br />

petrolatum at equal concentrations (390 ppm) (64). The mean reaction<br />

threshold for nickel sulfate in water was significantly lower (0.43%) than<br />

in petrolatum (0.51%). Also the irritation potential of a chemical can be<br />

assessed by applying it in different solvents in dermatological diagnostics,<br />

aiming to minimize the chemical’s potential for irritation and, thereby,<br />

false-positive reactions. Thus, the irritant reactivity of nickel salts in petrolatum<br />

was greater than in water (97).<br />

Such differences in skin reaction in functions of solvent serve to<br />

demonstrate the effect of the solvent carrier on dermal structures and, thus,<br />

the diffusivity of permeants.<br />

Counterion and Molecular Volume<br />

Diffusion of a cation is necessarily tied to the diffusion of its counterion (i.e.,<br />

diffusion of a metal will also proceed as an ion pair); otherwise, an electrical<br />

potential builds up, which will inhibit further diffusion. Thus, the overall<br />

volume is composed of two factors: the ionic radius of the elemental species<br />

[for example, chromium(III) as cations or chromium(IV) as chromate, the<br />

oxo-complex] and the variable size of the counterion (e.g., chloride versus<br />

sulfate versus nitrate, etc).<br />

Barrier diffusion of charged ions as ion pairs has been characterized as<br />

one criterion in skin permeation (98). Their obvious effect on permeant size,<br />

polarizability, and polarity ultimately find expression in respective diffusion<br />

constants (99,100). As the water content of the SC progressively increases<br />

in the deeper layers, degree of ion (complex) hydration increases further,<br />

to become fully hydrated toward the deeper regions of the membrane.<br />

The resulting increased steric hindrance also leads to a further decrease in<br />

ion mobility.


38 Hosty´nek and Maibach<br />

Experimental data also suggest that diffusivity of metal compounds<br />

through skin appears to correlate with their size and, for a given metal<br />

species, the counterion, thus, represents a determining variable. Own experimental<br />

data from depth analysis of nickel levels in the SC following in vivo<br />

application of a number of its salts confirm such effects of counterion size on<br />

their diffusion (101), an effect further amplified by hydration spheres.<br />

Polarity<br />

The polarity of nickel salts as measured by their solubility in the nonpolar<br />

solvent n-octanol at 22 C increases in highly significant intervals (p < 0.0005)<br />

in the following sequence: nitrate < chloride < acetate < sulfate (101). This<br />

increase in polarity determines the same sequential hierarchy for skin diffusivity<br />

and irritancy (93,97,102).<br />

Nature of Chemical Bond<br />

Going from inorganic, mostly water-soluble ionic mineral salts to lipophilic,<br />

organometallic compounds, bonds increasingly assume covalent character,<br />

and their penetration characteristics approach those of nonelectrolytes. With<br />

lead as an example, the role of ionic bond versus covalent bond is an obvious<br />

codeterminant for diffusion rates, the Kp ranging over four orders of magnitude,<br />

from 10 7 for the acetate to 10 3 for the lipophilic naphthenate (103).<br />

The range of penetration constants determined for a set of lead compounds<br />

illustrates the point of polarity as a determinant for skin penetration. That<br />

ranking of skin absorption in function of polarity was obtained for lead<br />

compounds by Bress and Bidanset, in vitro on human skin at equal concentrations,<br />

expressed as quantity absorbed (Table 2) (103).<br />

The lipophilic category, mainly alkyl and aryl derivatives of the more<br />

toxic metals, thus may represent a major health risk in chemical manufacture<br />

and industrial use, due to their ease of skin penetration. As an<br />

example, minute quantities of the highly toxic organometal dimethylmercury<br />

of previously unknown skin diffusivity were apparently absorbed<br />

Table 2 Skin Absorption of Lead Compounds Through Human Skin In Vitro in<br />

Function of Polarity<br />

Compound (as 10 mg Pb) Amount absorbed (mg)<br />

Percentage of dose<br />

absorbed<br />

Tetrabutyl lead 632 56 6.3<br />

Lead nuolate 130 15 1.3<br />

Lead naphthenate 30 3 0.30<br />

Lead acetate 5.0 0.9 0.05<br />

Lead oxide


Basics of Metal Skin Penetration: Scope and Limitations 39<br />

transdermally that led to the death of a university scientist having handled<br />

the chemical in the laboratory (81,104).<br />

Valence<br />

Ionic radii change with the number of outer electrons (i.e., with their<br />

valence), and thereby also the ion’s electrophilicity and reactivity toward<br />

proteins. These variables also determine the rate of metal diffusion. The<br />

negatively charged chromate and dichromate ions as oxo-complexes of<br />

hexavalent chromium do not bind to organic substances, whereas the electrophilic<br />

chromic ion [Cr(III)] shows a strong affinity for epithelial and<br />

dermal tissues, forming stable complexes that slow the rate of diffusion<br />

(73) or prevent it altogether (96).<br />

Occurrence (induction or elicitation) of allergic reactions in the skin due<br />

to allergenic agents may be considered to be an indicator of skin diffusivity<br />

(i.e., does an allergen penetrate the SC to reach the Langerhans’ cells in<br />

the viable epidermis). In animals, epicutaneous sensitization with trivalent<br />

chromium could be achieved only with difficulty. A 20-fold increase in the<br />

concentration of chromic sulfate [Cr(III)] was required to achieve the same<br />

degree of sensitization as with potassium chromate [Cr(VI)]. On the other<br />

hand, the same degree of contact sensitivity resulted from the intradermal<br />

application of either potassium dichromate or chromic sulfate, demonstrating<br />

that the SC is the barrier inhibiting percutaneous diffusion of trivalent chromium<br />

(105). In a study in which chromium sensitive volunteers were patch<br />

tested with a Cr(III)-containing detergent as well as with dichromate [Cr(VI)],<br />

those identified as chromium sensitive showed a positive response to dichromate,<br />

but none were sensitized to the detergent bar containing trivalent<br />

chromium (106). Differences in diffusivity due to protein reactivity between<br />

dichromate and chromic ions were also noted in vitro with human fullthickness<br />

abdominal skin. The permeability constant for hexavalent<br />

chromium as dichromate was an order of magnitude greater than that of<br />

trivalent chromic salts, overriding the retarding effect of dichromate’s larger<br />

molecular volume (106).<br />

Iron occurs mainly in its trivalent state (ferric ion) in nature, which is<br />

biologically unavailable. As a consequence, several mechanisms have<br />

evolved in the course of evolution to make its absorption by living organisms<br />

possible, and in mammals, iron is primarily absorbed in the duodenum<br />

as the divalent ferrous ion. Nutritionally most important is Fe 2þ bound<br />

in hemoglobin, occurring in organs and muscle tissues, and absorbable<br />

through gastrointestinal membranes. Once it reaches its target organ, iron<br />

is enzymatically oxidized back to Fe 3þ (107).<br />

pH and Solubility<br />

Changes in pH can have an effect on the skin penetration by electrolytes.<br />

From a dermatotoxicological perspective chromic [Cr(III)] salts are the least


40 Hosty´nek and Maibach<br />

problematic because of their poor aqueous solubility, which also decreases<br />

further with rising pH. Diffusivity of chromium as the oxo-complex dichromate<br />

can vary widely, ranging over more than an order of magnitude.<br />

Depending on pH, Cr(VI) exists in either the chromate (pH > 6) or dichromate<br />

form (pH ¼ 2–6). In either form, Cr(VI) is the greater hazard for<br />

human health, due to its heightened membrane diffusivity. The permeability<br />

constants measured for chromate–dichromate were higher in the pH range<br />

8 to 12.7 than in the lower range of pH 1.4 to 5.6. Chromium in the higher<br />

pH range exists as the smaller CrO4 2<br />

ion, whereas in the lower pH range,<br />

the larger, less diffusible dichromate ion Cr2O7 2 predominates (108).<br />

The pH dependence of zinc-oxide absorption was demonstrated by<br />

A˚ gren, where at a pH of 5.4 in vitro, uptake of the compound through<br />

human skin was 21 times greater than at pH 7.4 (8).<br />

Protein Reactivity and Depot Formation<br />

The electrophilic nature of many metals creates pronounced protein reactivity<br />

that can result in depot formation in the SC. Such protein–metal<br />

binding typically occurs with aluminum(III), silver(I), mercury(II), chromium(III),<br />

nickel(II), and the metalloid arsenic(III); it can take place in<br />

all strata of the skin to the extent of building up a secondary barrier and<br />

inhibiting further diffusion (109,110). Such deposits can also be reversible,<br />

however, as is the case for essential elements subject to homeostatic control<br />

(e.g., zinc or copper), which are retained by specific storage proteins,<br />

such as MT or coeruloplasmin, and released again upon physiological<br />

demand (111).<br />

Chromium is an example of the metals that, through nonspecific binding,<br />

causes protein denaturation with formation of permanent depots in the<br />

epidermis; these depots appear to be subject to the countercurrent effect of<br />

continuous sloughing of the outer SC layers, with release of significant<br />

amounts of the metal originally absorbed.<br />

Traces of chromium(III) are tenaciously retained, following exposure<br />

to chromium-containing dermatologicals. Elevated levels of the metal were<br />

seen in the skin up to three weeks, following a single inunction of 0.5%<br />

potassium dichromate in petrolatum (112).<br />

In an in vitro diffusion experiment by Gammelgaard, no chromium<br />

was detected in the recipient phase following the application of Cr(III) as<br />

the chloride or nitrate on full-thickness skin for 168 hours, or following<br />

application of dichromate over 48 hours (105).<br />

The avidity with which aluminum(III) forms complexes with skin and<br />

shunt protein means that there is only superficial penetration into the epidermis.<br />

This is the basis of the antiperspirant activity of certain aluminum<br />

salts, such as aluminum chlorohydrate, which, at the pH of the skin (or<br />

sweat), form insoluble salts and precipitates to effectively form a diffusion<br />

barrier (113).


Basics of Metal Skin Penetration: Scope and Limitations 41<br />

Dermal or systemic long-term exposure to a number of metals through<br />

occupation, therapy, lifestyle, or diet can result in their accumulation in the<br />

body, and their preferential deposition in dermal tissues can become visible<br />

as a characteristic discoloration of the skin. As these metals are not subject<br />

to significant natural processes of elimination, they accumulate over a lifetime<br />

and form permanent deposits in their elemental state, visualized histologically<br />

as metal particles. With the exception of arsenic, however, that phenomenon<br />

does not seem to have any untoward effects other than to alter the natural<br />

coloring of the skin.<br />

Time Postapplication<br />

Deposits formed by electrophilic metals appear as the cause for considerable<br />

lag times of several hours or days as observed in in vitro diffusion experiments.<br />

Before any permeant appears in the receptor compartment, in some<br />

cases, such as with chromium as observed by Gammelgaard in the previous<br />

section, the metal fails to appear at all. According to other observations, diffusion<br />

rates will increase with increases in donor concentration to a point,<br />

level off, and ultimately decrease.<br />

With mercuric chloride in vitro on whole-thickness skin, postapplication<br />

time was the most influential variable for absorption rate, indicating secondary<br />

barrier buildup. The percutaneous absorption of mercury was greatest in<br />

the first period (zero to five hours); it decreased in each successive period to the<br />

lowest rate, usually in the final period (36–48 hours), or sometimes in the nextto-the<br />

last period (24–36 hours). By the end of the experiments, the absorption<br />

rate was 40% of the initial rate, often 25% to 35% and sometimes as little as 1%.<br />

This trend was seen with both guinea pig skin and human skin (114).<br />

Commercial products (emulsions and ointments) containing copper have<br />

been studied over a period of 72 hours (9,61). Whether formulated with copper<br />

sulfate or the organic salt copper 2-pyrrolidone 5 carboxylate, apparent<br />

permeability coefficients over 72 hours were in the range 0.015 10 4 to<br />

0.13 10 4 cm/hr. Noteworthy differences in permeability coefficients were<br />

associated with the period of exposure; the values decreased over successive time<br />

periods from near 1 10 4 cm/hr (zero to two hours) to 0.1 10 4 cm/hr or<br />

less (25–48 hours) and finally to undetectable values in two cases (49–72 hours).<br />

As for percutaneous zinc, absorption rates from commercial emulsions<br />

and ointments, zinc 2-pyrrolidone 5-carboxylate, ZnO and ZnSO4, measured<br />

in vitro with human skin were highest in the first two hours after<br />

application, then decreased to less than a 10th of the initial rates (9,61).<br />

Endogenous Factors<br />

Regional Variation<br />

Skin absorption by chemicals, investigated with steroids and pesticides, is<br />

subject to variation among different sites of the body, decreasing in the


42 Hosty´nek and Maibach<br />

order of scrotum > forehead > postauricular > abdomen > forearm > leg ><br />

back (115–117). Apart from regional SC thickness and shunt density, this<br />

appears to be mainly due to intercellular lipid weight percentage and<br />

composition (118).<br />

Because penetration of electrolytes appears to occur mainly through the<br />

skin’s appendages (12,18,30,42), diffusion in hairy areas may be enhanced,<br />

although absorption was also observed through the palm of the hands devoid<br />

of hair follicles; the route of diffusion there was probably via the sweat<br />

ducts (119).<br />

When investigating site dependence of percutaneous absorption for<br />

a number of organic compounds in vivo, Lotte et al. noted a linear relationship<br />

between penetration and TEWL (i.e., the relationship between<br />

the permeability of the skin to the outward movement of water and inward<br />

movement of permeants) (117).<br />

Following application of nickel chloride on the arm and back of<br />

human volunteers, based on tape strip analysis of the SC, the depth/concentrations<br />

of the two sites were steep though at differing gradients, declining<br />

toward deeper SC layers, with significantly different areas under the curve:<br />

30.8 for skin on the back versus 62.4 on the arm (p < 0.0005), an indication<br />

of heightened diffusivity in arm skin (101).<br />

Age of Skin<br />

The incomplete barrier function observed in infants and young children gradually<br />

increases to values seen in the skin of mature individuals. Neonatal or<br />

infant skin has been found to be more permeable to lipophilic compounds<br />

than is adult skin. Feldmann and Maibach studied percutaneous penetration<br />

of taurocholic acid in vitro through male thigh skin as related to age;<br />

after 24 hours, while skin from a 32-year-old absorbed 8.2%, the skin of a<br />

53-year-old absorbed only 0.3% of the dose (119). One theory associates<br />

such decreased permeability with age as function of diminishing blood<br />

supply (120).<br />

Our own in vitro investigation of permeability to a nickel salt (chloride)<br />

and a nickel soap (di-octanoate), using dermatomed skin under identical<br />

experimental conditions, confirms the decreasing trend in skin diffusivity<br />

with age, specifically between skin from a young (age 16 years) versus an<br />

older source (age 64 years) (unpublished data). While the ratios of diffusivity<br />

values for salt and soap remained the same, advanced age brought a reduction<br />

in rates in excess of two orders of magnitude (121).<br />

Homeostatic Controls<br />

Essential elements such as sodium, potassium, calcium, copper, or zinc present<br />

in the SC, epidermis, and dermis are kept in equilibrium by homeostatic<br />

control mechanisms. These mechanisms play a part in the overall physiological<br />

dynamics of hydration, and also appear to keep the body burden of


Basics of Metal Skin Penetration: Scope and Limitations 43<br />

certain xenobiotics under control. Homeostasis ascertains maintenance of<br />

equilibria necessary for optimal functioning of the organism (e.g., by preventing<br />

undue loss due to perspiration or desquamation through a process of<br />

reabsorption, as has been described for sodium and calcium, in particular)<br />

(122,123).<br />

An important part of such controls are metal-binding MTs, single-chain<br />

polypeptides, present in several organs, including the skin. They have an unusual<br />

amino acid composition consisting of one-third cysteine (20 cysteines in<br />

mammalian MTs) and no histidine, aromatic, or heterocyclic components.<br />

This abundance of thiol groups imparts both reversible metal-binding capacity<br />

and the ability to scavenge free radicals. At least three functions have been<br />

attributed to MTs: metal sequestration, temporary (reversible) binding as well<br />

as long-term storage, and both intra- and extra-cellular metal transport. They<br />

bind the metabolically essential zinc and copper, as well as cytotoxic metals<br />

such as mercury, lead, or nickel.<br />

Levels of free ionic copper, a relatively toxic metal, are moderated<br />

to the minimum levels sufficient for physiologic needs; 10 19 mol/L are<br />

estimated in blood plasma via binding to MT as well as the copper carrier<br />

protein ceruloplasmin (124). An excess of free ionic copper in cells would<br />

lead to oxidative damage, and the affinity of MTs for copper is second only<br />

to that of mercury. The dynamic equilibrium between ceruloplasmin and<br />

MT contribute to the prevention of both toxic accumulation and deficiency<br />

of copper in mammals (125).<br />

When present above specific threshold levels, even ETEs will exhibit<br />

acute toxicity; and by immobilizing excess amounts, MTs fulfill a critical<br />

detoxifying role. MTs thus remove, store, or release metals on environmental<br />

exposure or physiological demand, and are an important factor in homeostasis,<br />

regulating their uptake (diffusion) and release.<br />

Measurements of the rate of skin penetration by zinc have been contradictory,<br />

possibly due to such endogenous controls. Apparently zinc<br />

absorption does not occur by simple diffusion, but seems to be regulated<br />

by MTs (126). MTs thus provide a buffering capacity maintaining intracellular<br />

steady-state kinetics for both copper and zinc, and ensuring a supply of<br />

these metals for other metabolic functions.<br />

As homeostasis is responsible for a rapid exchange between skinapplied<br />

zinc and the large pool of endogenous metal, dermal absorption<br />

experiments conducted in vivo thus should account for natural processes<br />

that may counteract passive diffusion, adding a degree of uncertainty to permeability<br />

constants measured.<br />

Skin Sections<br />

Data are limited on the effect of individual skin layers (SC, epidermis, and<br />

dermis) on percutaneous penetration by metals, and on metal uptake in the<br />

individual strata. An example of the variability observed is the different


44 Hosty´nek and Maibach<br />

penetration rates measured in vitro for nickel chloride. Comparison of Kps<br />

determined through different skin strata by different authors shows that<br />

penetration by nickel chloride is slowest through the SC (37,53,91,93). From<br />

a Kp of 10 7 cm/hr in the SC, the values progressively increase toward fullthickness<br />

skin, with a maximum of 9.8 10 3 cm/hr in dermatomed skin.<br />

Skin sections prepared for in vitro experiments, other than dermatomed<br />

skin, are likely to introduce artifacts to varying degrees. One possible<br />

explanation for low diffusion values through the SC is that shunts in isolated<br />

SC (and to some degree in epidermis) swell shut upon hydration (127). In<br />

vitro diffusion data, which come closest to in vivo conditions, are likely to<br />

be those measured through split-thickness skin. That tissue can be standardized<br />

using the dermatome to a desired thickness (200–400 mm). Unlike other<br />

methods of skin sectioning, use of the dermatome is the best way of preparing<br />

skin for percutaneous absorption studies. A dermatome can be used with<br />

hairless or haired skin, without adversely affecting the viability of the membrane.<br />

Dermatomed tissue includes the layer of dermis where the permeant<br />

is taken up in the capillary system, without adding the variable thickness<br />

of dermal and subdermal tissue and fat.<br />

Skin Metabolism (Red/Ox)<br />

There is evidence of cutaneous metabolic effects on metals of exogenous or<br />

endogenous sources; both oxidation and reduction can occur, manifest in<br />

changes of oxidation state in situ during the process of permeation. The<br />

result can be altered immunogenicity of the metals; examples are known<br />

where oxidation can lead to enhanced immunogenicity of certain metals<br />

[e.g., of Ni(II) to Ni(III) or Ni(IV)] (128), or reduction to lesser immunogenicity<br />

[e.g., Cr(VI) to Cr(III)] (129).<br />

Animal experiments conducted by Artik et al. showed that biooxidation<br />

of Ni(II) to Ni(III) or Ni(IV) occurs through endogenous-reactive<br />

oxygen in the form of hydrogen peroxide or hypochlorite present in inflamed<br />

skin. In animal and cell line tests Artik did find that Ni(II) sensitizes only<br />

na€ve T cells following bio-oxidation to its higher valence, but not the form<br />

of Ni(II) itself (128).<br />

Chromium applied on the skin as chromate or dichromate [Cr(VI)] is<br />

reduced to chromic ion (Cr 3þ ) by tissue proteins containing sulfhydryl<br />

groups (130). In in vitro diffusion experiments with Cr(VI) salts, only chromic<br />

ion is found initially in the receptor phase. On sustained application,<br />

however, Cr(VI) as chromate or dichromate is seen to pass through the skin<br />

unchanged, an indication that a given tissue mass has only a limited capacity<br />

to reduce the chromate ion (105). For some metals, reduction can lead to<br />

discoloration of the skin due to accumulation of the element in the metallic<br />

state (e.g., systemic silver preferentially accumulates in the skin, where it is<br />

reduced to the metallic state). Such impregnation of dermal tissues with<br />

silver deposits results in permanent graying of the skin, a condition termed


Basics of Metal Skin Penetration: Scope and Limitations 45<br />

argyria (131). Chronic dermal application of mercurials used as skin<br />

bleaches (mercurous chloride, ammoniated mercury, and mercurous oxide)<br />

can lead to tissue accumulation of metallic mercury, characterized by slategray<br />

pigmentation or hydrargyrosis cutis (132). The skin is a target organ<br />

for arsenic and is critically sensitive to arsenic toxicity, regardless of the<br />

route of exposure. This is due to the attraction of arsenic to the skin’s sulfhydryl-group<br />

containing proteins. Such accumulation of arsenic in the skin<br />

is characterized by hyperpigmentation, keratoses of the palms of the hands<br />

and soles of the feet, and diffuse macular pigmentation with the characteristic<br />

appearance of ‘‘raindrops’’ or diffuse darkening of the skin on the limbs<br />

and trunk, attributed to the reduction and deposition of the element in the<br />

metallic state (127).<br />

METHODS FOR MEASURING PERCUTANEOUS ABSORPTION<br />

In this context, the terms absorption, diffusion, and penetration are used<br />

interchangeably as they apply to the process of penetrating the outermost<br />

skin layer, the SC, and to all the associated and subsequent events, including<br />

distribution to the different strata and appendages of the skin.<br />

In Vitro Methods<br />

Preferably skin diffusivity of metals is measured in vitro using excised<br />

(animal or human) skin, especially in the investigation of materials of<br />

unknown or obvious toxicity. Reasons for such preference are:<br />

1. Formation of depots in the SC by electrophilic metal ions, establishing<br />

a secondary barrier to further diffusion, which results in<br />

substantial lag times or failure to penetrate the epidermis altogether<br />

(93,105)<br />

2. The need to use radiolabeled materials for the detection of exceedingly<br />

low levels of permeant<br />

3. The pronounced toxicity of some (transition) metals, too hazardous<br />

to apply on the human organism in vivo<br />

Widely used methods for the exploration of percutaneous absorption,<br />

in vitro designs allow a preliminary and early evaluation of safety for<br />

chemicals in the developmental stage, as well as of those too toxic to test<br />

in living models. These methods offer the advantage of yielding data that<br />

reflect the process occurring in selected domains of the skin (SC, epidermis,<br />

and dermis) without involvement of other factors, such as secondary absorption,<br />

deposition, or metabolism in the body’s tissues.<br />

The most commonly used in vitro technique for measuring percutaneous<br />

absorption involves placing a piece of excised skin in a two-chamber<br />

diffusion cell. It consists of a top chamber to receive an adequate volume of


46 Hosty´nek and Maibach<br />

penetrant in solution, an O-ring to secure the skin in place, a temperaturecontrolled<br />

bottom chamber with continually circulating solution removing<br />

the penetrating amounts on the receptor side of the membrane, and a sampling<br />

port to withdraw fractions at specific time intervals for analysis (133).<br />

The solute diffuses from the fixed higher concentration medium in the donor<br />

chamber into the less concentrated solution in the receptor chamber. The<br />

solutions in both chambers are stirred continuously to maintain uniform<br />

concentrations. The advantage of diffusion experiments thus conducted lies<br />

in the applicability of Fick’s first law of diffusion.<br />

In the two-chambered system the two chambers are separated by the<br />

skin membrane. To satisfy the requirements of the Fickian diffusion, useful<br />

for the study of diffusion through biological membranes, an (ideally) infinite<br />

dose is placed in the donor chamber, and appearance of the permeant continually<br />

monitored in the receptor chamber. The solutions in both chambers<br />

are stirred to maintain uniform concentration. Experimental details are<br />

given with an example of steady-state flux measured through the SC (23).<br />

In the one-chambered system, the skin is placed on the chamber and is<br />

open to the environment above, simulating conditions prevailing in real-life<br />

application of drugs and cosmetic products to the skin. As permeation proceeds,<br />

steady-state (Fickian) conditions are not attained (20). The chamber<br />

beneath the skin serves as a container for the receptor fluid that is continually<br />

stirred. Sampling occurs through a side arm for analysis to determine rates of<br />

absorption (134,135). Automatic sample collection is also possible from a<br />

one-chambered cell (136). The receptor volume is small (0.13–0.26 mL) and<br />

allows complete and rapid flushing of the receptor chamber. Special provisions<br />

are necessary to avoid evaporation of volatile compounds from<br />

the surface of the skin. They can be collected through appropriate cell<br />

design (137,138).<br />

Limitations and Problems in Measuring Percutaneous<br />

Absorption of Metal Compounds In Vitro<br />

A limitation in the validity of percutaneous absorption values determined<br />

for electrophilic metals in vitro is the formation of depots in the skin. Such<br />

protein reactivity can result in depot formation in the SC before permeant<br />

diffusion continues into the deeper layers of the skin, and in vitro it can cause<br />

considerable delays (lag times) before the compound emerges in the receptor<br />

phase. For some metals, lag times of several hours or days have been recorded<br />

before any permeant appears in the receptor compartment, or as in some<br />

cases such as chromium, fails to emerge at all. This phenomenon has been<br />

reported for the more extensively investigated metals, such as nickel, cadmium,<br />

and chromium (93,105,139). Due to the electrophilicity of transition<br />

metals, in particular, it can be safely assumed that latency or retardation in<br />

the diffusion process is a predictable phenomenon. As permeability constants<br />

and bioavailability are calculated only from amounts of permeant collected


Basics of Metal Skin Penetration: Scope and Limitations 47<br />

in the receptor phase at steady-state rates, the formation of depots is<br />

rarely part of in vitro diffusion studies, although it should be considered as<br />

part of the total-dose absorbed. The permeant retained in the different levels<br />

of the skin tissues may then eventually become available systemically by<br />

slow diffusion.<br />

For those metals where skin penetration was investigated, the data<br />

rarely are adequate for the calculation of flux or permeability coefficient,<br />

and values can only be derived through a number of assumptions.<br />

Results that are amenable to quantitative analysis often followed<br />

diverse experimental protocols that lead to results that are not necessarily<br />

comparable.<br />

While a permeability coefficient is ideally determined under steadystate<br />

conditions, the percutaneous absorption of metals rarely meets this<br />

criterion.<br />

A problem inherent in the in vitro approach is the lack of total-dose<br />

accountability, as results in the literature are often based on the quantity<br />

of permeant found in the receptor phase only. The amount of chemical collected<br />

in the receptor phase, expressed as percentage of dose, is then taken to<br />

be the amount becoming available systemically, and material retained in the<br />

diffusion membrane (SC, epidermis, and dermatomed or full-thickness skin)<br />

that may subsequently diffuse further, and thus become a significant factor<br />

for risk assessment purposes, is not determined.<br />

In Vivo Methods<br />

The purpose of measuring the percutaneous absorption in vivo is to study<br />

bioavailability of chemicals applied on the skin by analysis of dose moving<br />

into the skin, further into the systemic circulation, and beyond. Such<br />

kinetics measure diffusion by skin tape stripping, bioengineering techniques<br />

such the TEWL, or by traditional analysis of blood and excreta levels.<br />

Taken together, these components can define the dermatopharmacokinetics<br />

of a chemical. After topical application, the levels of xenobiotic in tissues<br />

and body fluids as a rule are below assay detection levels, and it is necessary<br />

to use tracer methods.<br />

The most relevant in vivo data on percutaneous absorption will be<br />

obtained from studies in humans themselves. However, animal models are<br />

needed for the development of basic pharmacokinetic principles and for<br />

the investigation of pharmacological mechanisms. On the basis of currently<br />

available data, the only animals in which permeation is consistent qualitatively<br />

and qualitatively with human permeation data are the weanling pig<br />

(138), the rhesus monkey (136), and the hairless rat (137). Extra body fat<br />

in the pig may affect drug distribution compared to humans, however,<br />

and must be considered. In the rhesus monkey, skin application should be<br />

limited to the nonhairy regions on the ventral surfaces of the animal.


48 Hosty´nek and Maibach<br />

Overall, correlation of animal skin penetration with human absorption data<br />

made by different researchers can vary depending on the compound nature<br />

involved (22). Skin absorption rates determined in animals as a rule result<br />

in a conservative estimate as they overestimate the dose percutaneously<br />

absorbed in humans. Penetration and absorption studies must address toxicologic<br />

aspects and toxicokinetic aspects, as well as developmental and<br />

clinical issues. Also the benefits of using radiolabeled versus nonradiolabeled<br />

analytical methodology have to be considered.<br />

Studies can focus on drug in the skin, in venous blood draining<br />

the application site, in the systemic circulation, or in the excreta. For pharmacokinetics,<br />

drug concentrations are followed over time. For topical<br />

dermatological products, toxicokinetic and safety studies are preferably<br />

performed in the same species.<br />

An indirect method is the quantification of RA in excreta, where percutaneous<br />

absorption is determined by measuring the appearance of RA following<br />

topical application of a labeled compound. Measured is the total RA<br />

of parent compound and any labeled metabolites, which may occur in transit<br />

through skin and body. Label retained in the organism or excreted by<br />

another route (e.g., exhaled air) will not be counted by that method. Therefore,<br />

Feldmann and Maibach used the following expression to correct for<br />

RA unaccounted for:<br />

Percent absorbed ¼ 100 total RA from topical administration/total RA<br />

following parenteral administration<br />

Percutaneous absorption can be measured directly by including material<br />

exhaled and that remaining in tissues at the end of the experiment,<br />

besides excreted material in urine and feces. This sum then gives a direct<br />

measure of absorption (140,141). Because absorption is expressed as the<br />

percent of dose applied, no Kp becomes available using these methods.<br />

In the disappearance method the amount of drug absorbed is assessed<br />

as the difference between the amount applied and that recovered at a given<br />

interval. The method relies on the assumption that the difference between the<br />

quantity applied and that recovered corresponds to the amount absorbed; it<br />

can follow different approaches:<br />

1. Following drug application for a fixed time, the residual formulation<br />

is washed from the skin surface, and the amount removed is<br />

analyzed.<br />

2. The formulation is applied, and the drug content in the outer skin<br />

layers is followed in function of time by spectroscopic or radioisotopic<br />

monitoring techniques.<br />

3. Disappearance (loss) of a radioactive compound from the surface<br />

of the skin and the appearance of RA in the excreta following the<br />

topical application of a labeled compound are followed over time.


Basics of Metal Skin Penetration: Scope and Limitations 49<br />

The total RA in the excreta is a mixture of the parent compound<br />

and any labeled metabolites that may result from metabolism of<br />

the parent in the skin and in the body. The method can involve<br />

single-point measurement, or continuous or periodic monitoring<br />

of compound uptake.<br />

One limitation in this approach is the fact that once the applied<br />

compound has penetrated, its disposition in the organism eludes detection,<br />

especially because the application of radioactive material on men is limited<br />

for ethical reasons. Another problem inherent in this approach, particularly<br />

when involving electrophilic, protein-reactive ions, is that the rate measured<br />

may be a combination of absorption into the systemic circulation and the<br />

dose which is retained in the SC. Frederickson described in detail the problems<br />

arising when measuring absorption of compounds that permeate the<br />

skin slowly (142). As seen in in vitro experiments conducted with reservoirforming<br />

compounds, the portion retained in the surface strata of the SC<br />

is far more important than material reaching the receptor phase, if any<br />

(93,105,143).<br />

Also, depth of penetration through the epidermis remains unknown.<br />

Copper is subject to homeostatic control, and is known to be dynamically<br />

bound to MTs in the skin (93). Thereby, even recovery in excreta may not<br />

afford a relevant mass balance. Advantages of this method are that it<br />

requires small amounts of active formulation (at pharmacologically insignificant<br />

concentrations), it is inexpensive, relatively rapid, and applicable in<br />

clinical studies.<br />

The difficulties inherent in skin recovery, volatility of penetrant, and<br />

errors associated with using the difference between the amount of compound<br />

applied and amount remaining make this an inexact method for the quantitative<br />

determination of absorption rates. The disappearance method has<br />

been used extensively with radiolabeled metal salts, primarily by Wahlberg<br />

et al. To measure the loss of material from the skin surface over time, the<br />

guinea pig is the model mostly used in vitro and in vivo, with a limited number<br />

of experiments conducted on human skin in vitro (144). Consistently<br />

using that technique, Wahlberg and coworkers studied a dozen metal salts<br />

for their skin diffusivity. Although the data on skin disappearance were<br />

obtained over relatively short exposure times, the results are valuable as<br />

benchmarks, internally consistent yielding a scale of relative diffusivities<br />

allowing at least partial validation of other measurements for those metals.<br />

Zinc that is reversibly bound to sulfhydryl storage sites becomes available<br />

for immediate systemic absorption when deficiency develops (145).<br />

Skin conditions due to nutritional zinc deficiency, collectively described<br />

as zinc deficiency dermatoses, have been found to respond promptly to<br />

treatment, and dramatic improvement was seen within days of initiating<br />

transdermal zinc therapy (130,146). Occlusive dermal application of a


50 Hosty´nek and Maibach<br />

concentrated zinc ointment to healthy human volunteers on a normal diet<br />

on the other hand did not lead to a significant increase in serum zinc concentration.<br />

When a similar application was made to the skin of patients<br />

on total parenteral nutrition, a dietary routine that typically results in zinc<br />

deficiency, serum zinc levels were nevertheless maintained at surprisingly<br />

normal and constant values. Based on such observations the percutaneous<br />

absoption of zinc appears to depend on actual zinc status in the overall<br />

organism, and thereby is variable. The close association and interrelationship<br />

between zinc and copper, therefore, would indicate similar variability<br />

in trasdermal diffusion rates measured for the latter, making Fick’s law of<br />

diffusion inapplicable.<br />

These caveats notwithstanding, determined under observance of steady<br />

state and normalized for concentration, by default the Kp is still the most<br />

widely used descriptor of chemical diffusion through the skin. Based on a<br />

number of necessary assumptions, literature data can be transformed so as<br />

to put metal absorption values on a common scale, adequate for purposes<br />

of risk assessment.<br />

Skin Stripping—A Semiquantitative In Vivo Method<br />

Use of adhesive tape to sequentially remove layers of the SC in vivo following<br />

topical application of a compound was proven useful in dermato-pharmacokinetic<br />

and -toxicological research to investigate reservoir formation by drugs<br />

(147). Based on an absorption study of selected drugs in vivo, a relationship<br />

was defined between SC concentration and their eventual total (systemic)<br />

absorption (35). By partial stripping (six iterations) of the SC, the amount<br />

of chemical residing in the SC after 30 minutes exposure time highly correlates<br />

with the total amount of chemical penetration within four days, as determined<br />

by the standard urinary excretion method. It thus becomes possible to make a<br />

predictive assessment of total drug uptake from the analysis of superficial SC<br />

levels only.<br />

Although skin stripping can also be used in vitro, its use in vivo most of<br />

all permits drawing a more authentic profile of the SC, as it avoids problems<br />

otherwise encountered, such as the creation of artifacts and damage potentially<br />

associated with sectioning cadaver skin, storing conditions, or duration<br />

of the experiment (tissue integrity). Rougier’s tape stripping protocol obviates<br />

the need for urinary and fecal excretion analysis, and is applicable to nonradiolabeled<br />

determination of percutaneous absorption, because strippings<br />

remove permeant in quantities adequate for nonlabel assay, such as high pressure<br />

liquid chromatography (HPLC).<br />

Rougier’s correlation, however, cannot be expected to be applicable to<br />

investigation of highly electrophilic, protein-reactive compounds, such as transition<br />

metals, some of which appear to form permanent depots in the SC (e.g.,<br />

nickel, lead, or mercury). For those metal compounds, the direct skin stripping


Basics of Metal Skin Penetration: Scope and Limitations 51<br />

technique followed by inductively coupled plasma–mass spectrometry analysis<br />

was useful in visualizing SC penetration by nickel, in particular (78,101).<br />

The outstanding biological characteristic of nickel, from a public<br />

health perspective, is its immunotoxicity. Nickel started out as a prime occupational<br />

hazard reported from the metalworking and refining industry in the<br />

late 19th century and the first part of the 20th century due to its allergenicity<br />

(139,148,149), but since the Second World War it has become a consumers’<br />

affliction as well, because the metal is now part of most alloys used in the<br />

manufacture of common materials in tools and articles of daily contact.<br />

In a number of dermatotoxicity studies, the incidence of nickel hypersensitivity<br />

noted for women formerly ranged from 10% to 20%, that for men<br />

from 1% to 4%. In more recent studies from a number of countries, a striking<br />

increase in these numbers has been recorded, particularly among women<br />

and children of school age: to 31.9% among schoolgirls in Italy (150), to<br />

43.7% among dermatological clinic patients in Poland (151), and to 30%<br />

among an unselected group of schoolgirls in Finland (152).<br />

To gain insight into the dermatopharmacokinetics of nickel as allergen,<br />

the protocol of sequential adhesive tape stripping was implemented to<br />

examine the penetration characteristics of both the nickel salts and nickel in<br />

its metallic state in human SC and its potential to reach the viable epidermis,<br />

following their application on the skin volunteers. Inductively coupled<br />

plasma-mass spectroscopy (ICP-MS) was used to analyze tape strips (78,101).<br />

For nickel chloride, sulfate, nitrate, and acetate, the depth-penetration<br />

profiles obtained by tape stripping and analysis led to a number of conclusions:<br />

1. Up to 24 hours, most of the nickel dose applied remained on the<br />

SC surface<br />

2. Nickel adsorbed accumulated in the uppermost SC layers, and the<br />

concentration gradient between the superficial and deeper layers<br />

increased commensurate with exposure time and concentration.<br />

3. In a number of experiments, mass balance based on amounts<br />

retrieved from the skin surface and cumulative nickel analysis to<br />

the level of the glistening layer showed a deficit. Within 24 hours<br />

nickel salts thus appeared to penetrate beyond the SC to a minor<br />

degree, possibly via the skin shunts, to become systemically available.<br />

4. While the concentration gradients of nickel adsorbed varied with<br />

counterion, anatomical site, dose, and exposure time, toward the<br />

level of the glistening layer for all variables tested, the depth profiles<br />

converged toward nondetectable levels (


52 Hosty´nek and Maibach<br />

distribution profiles increased proportionally with occlusion time but leveled<br />

off with increasing depth after the 10th strip, to continue at constant levels<br />

to the 20th strip. From application (of a large excess) of metal powder on a<br />

surface of 1.15 cm 2 , the total nickel removed with 20 SC strips after maximum<br />

occlusion of 96 hours was 41.6 mg/cm 2 (101).<br />

The method of SC stripping as applied in nickel research could be used<br />

to investigate skin diffusivity of other metals, such as copper. It would bring<br />

insight into the interaction of that metal brought in contact with the microenvironment<br />

on live skin.<br />

In Vivo–In Vitro Correlation<br />

In validation studies comparing absorption values, good agreement between<br />

in vivo and in vitro methods have been obtained if the studies were conducted<br />

along generally accepted guidelines. The application of a permeant<br />

to the skin in vivo may be more physiologically relevant than the use of<br />

in vitro methods, and in case of disagreement, precedence goes to in vivo<br />

data, although in vivo execution may be subject to error. The results of<br />

in vivo studies are reported as the percent of dose absorbed, whereby appropriate<br />

transformation becomes necessary if a Kp is required.<br />

In vitro absorption values are generally good predictors of the rate or<br />

extent of percutaneous absorption in the intact animal. However, the factors<br />

described may affect the predictive accuracy of in vitro absorption methods,<br />

especially for compounds of high hydrophilicity or lipophilicity. Failure to<br />

account for these variables will lead to poor correlation between in vitro<br />

and in vivo percutaneous absorption. If Kp data are used to estimate percutaneous<br />

absorption, it is necessary to examine the experimental conditions<br />

used if in vitro Kps are to correlate with in vivo data. An example is given<br />

with the skin absorption experiment conducted with boron compounds.<br />

Parallel in vitro and in vivo studies conducted by Wester et al. on the skin<br />

absorption of a number of boron compounds identified substantial and<br />

unexpected discrepancies between the results obtained (153). In the in vitro<br />

study, dosing corresponded to infinite (1 mL/cm) and finite (2 mL/cm)<br />

amounts. These doses correspond to infinite amounts available for absorption<br />

as a theoretical setting to define diffusivity, versus a dose remaining on the<br />

skin to convey realistic in vivo short-term exposure. The latter dose was also<br />

applied on the skin in vivo, which was then allowed to dry. The Kp values<br />

calculated from the infinite dose exposure was 1000-fold higher than the<br />

one from the in vivo study, while the finite in vitro dosing mode was only<br />

10-fold higher. The authors concluded that the in vivo and the in vitro finite<br />

dose data appeared more relevant for a real-life exposure scenario, reflecting<br />

that under infinite dosing over 24 hours in vitro the skin membrane was progressively<br />

becoming more permeable. The infinite in vitro dosing protocol<br />

would only be relevant for a full-body exposure over an extended period.


Basics of Metal Skin Penetration: Scope and Limitations 53<br />

ANALYTICAL METHODS FOR METAL DETECTION<br />

The toxicology of metal compounds is becoming increasingly important in<br />

the fields of environmental, occupational, and clinical medicine. The basis<br />

for studying metal toxicity lies in the application of sensitive and accurate<br />

analytical techniques, and tissue localization of metals in tissue (skin) contributes<br />

significantly to the understanding of toxic mechanisms. Going beyond<br />

conventional chemical analysis, physical methods now afford sensitivities in<br />

metal detection that are lower by several orders of magnitude, often reaching<br />

below the ppb level, and the quantitative tools of atomic spectroscopy have<br />

brought the most significant advances. Adapted to monitoring of toxic elements<br />

in biological substrates, such as tissues or body fluids, such sensitivity<br />

is essential for the assessment of risk to toxic metals without the need to<br />

resort to radionuclides. Thus, monitoring of metal levels in skin and body<br />

fluids by physical methods can provide adequate estimates of dose absorbed,<br />

indicating the need for corrective intervention in cases of potential exposure<br />

hazard, especially in the work environment.<br />

Inductively Coupled Plasma–Atomic Emission Spectroscopy<br />

ICP-atomic emission spectroscopy (ICP-AES) permits detection of metals at<br />

the trace amount level, obviating the use of radioisotopes. For the detection<br />

of copper, the current quantitation limit falls in the 5 to 10 ppb (mg/L) range,<br />

a factor of 5 above the true instrumental detection limit as defined by the U.S.<br />

Environmental Protection Agency (EPA) (22). Quantitative detection of<br />

metals is accomplished by ionization of elements in inductively coupled<br />

argon plasma maintained by the interaction of a radiofrequency field and<br />

ionized argon gas. In a sample aerosol (e.g., a vaporized metal salt solution)<br />

atoms and ions are activated at 6000 C to an unstable energy state, and as<br />

they revert to their ground state again, they emit light of characteristic wavelength<br />

and intensity that can be measured.<br />

Inductively Coupled Plasma–Mass Spectroscopy<br />

ICP-MS is a technique applicable to mg/L (ppb) concentrations of many<br />

elements in aqueous medium upon appropriate sample preparation of biological<br />

materials. Reliability of the method for elemental analysis is based upon<br />

multilaboratory performance compared with that of either furnace atomic<br />

absorption spectroscopy or ICP-AES. Normal instrumental detection limit<br />

for copper, as for a number of other transition metals, falls in the 0.5 ppb range.<br />

The method measures ions produced by radiofrequency inductively<br />

coupled plasma. Compound to be analyzed, present in liquid form, is nebulized<br />

and the resulting aerosol transported by argon gas into the plasma<br />

torch. The ions produced are entrained in the plasma gas and introduced,<br />

by means of a water-cooled interface, into a quadrupole mass spectrometer.


54 Hosty´nek and Maibach<br />

The ions produced in the plasma are sorted according to their mass-tocharge<br />

ratios and quantified with a channel electron multiplier (142).<br />

Stable-Isotope Inductively Coupled Plasma–Mass<br />

Spectroscopy Analysis<br />

Assessment of the relevance of metal absorption data can be difficult and is<br />

rendered ambiguous by the intake of naturally occurring elements in the<br />

normal diet, reflecting in the skin and in the overall organism.<br />

Thanks to recent developments in analytical techniques and the availability<br />

of artificially generated stable metal isotopes, it is now possible to<br />

overcome those earlier difficulties. By dosing stable isotopes in vivo, which<br />

pose no risk to volunteers, it is now possible to differentiate between the<br />

two kinds.<br />

Using ICP-MS for the analysis of the stable isotope 10 B, the percutaneous<br />

absorption of boric acid, borax, and disodium octaborate over a<br />

24-hour exposure period was determined in human volunteers, unambiguously<br />

differentiating between the amount absorbed through the skin and<br />

that absorbed with the diet (153).<br />

The stable-isotope approach was used to investigate lead metabolism<br />

in normal humans by replacing part of the dietary lead with lead-204 tracer.<br />

Kinetic and metabolic balance studies of the volunteers kept on controlled<br />

diets for six months indicated that approximately two-thirds of assimilated<br />

lead was dietary in origin; the remainder was inhaled (143).<br />

By that approach, absorption of copper and other metals in the skin<br />

now becomes measurable, independent from endogenous natural isotopes<br />

present in the organism.<br />

Atomic Absorption Spectrophotometry<br />

Atomic Absorption Spectrophotometry (AAS) with Zeeman background<br />

correction is the reference method accepted by the International Union of<br />

Pure and Applied Chemistry and the International Agency for Research<br />

on Cancer for trace element analysis (144). The method is based on absorption<br />

of radiation energy by free atoms, characteristic for each element. In an<br />

atomizer, thermal energy (air–acetylene flame) converts the analyte to free<br />

atoms. In the ground state they absorb resonance radiation from a light<br />

source that emits characteristic radiation (i.e., the spectrum of the analyte<br />

element) whereby an electron transitions from the ground state to one of<br />

the empty orbitals at a higher energy level. A portion of this light is thus<br />

attenuated by such resonance absorption in the probe. A photomultiplier<br />

detector measures the change in radiation intensity and converts it into an<br />

absorbance signal. There is a linear relationship between absorbance and<br />

the concentration of the analyte. The technique is intrinsically specific,


Basics of Metal Skin Penetration: Scope and Limitations 55<br />

because the atoms of a particular element absorb only radiation of their<br />

own characteristic wavelength (154).<br />

AAS is the most common technique for nickel analysis in biological<br />

fluids. The sample to be analyzed is digested in acid, the nickel then chelated<br />

with ammonium tetramethylene dithiocarbamate, the chelate extracted with<br />

4-methyl-2-pentanone, and the nickel in the extract measured by AAS, or the<br />

initial acid solution analyzed as such by AAS directly (155). Currently,<br />

the detection range for nickel by AAS in body fluids (urine and serum) is<br />

0.4 to 0.05 ppb (156).<br />

Electron Spin Resonance<br />

Electron spin resonance (ESR) applies to transitions between electronic spin<br />

states in a magnetic field. The field of a probe is swept by a magnet,<br />

and resonance indicates presence of unpaired d-electrons in paramagnetic<br />

transition metals, the triplet state of two unpaired electrons in orthogonal<br />

orbitals, such as in elemental oxygen, or in isotopes, such as 13 Cor 17 O.<br />

If an unpaired electron registers proximity of magnetic nuclei with a nuclear<br />

spin 1/2, the single absorption is split into hyperfine structures, identifying<br />

interacting nuclei and their number in the vicinity of the unpaired electron.<br />

ESR is applied in in vitro analysis of biological materials, and, most<br />

recently, in analogy with nuclear magnetic resonance (NMR), instruments<br />

have been developed that allow in vivo analysis of live tissue. ESR detects<br />

the presence of stable organic species, such as nitroxides; organometallic<br />

and inorganic species with unpaired electrons can be detected in liquid as well<br />

as solid materials. Transient, short-lived species, such as organic free radicals,<br />

must be continuously generated in the ESR probe to maintain a sufficient<br />

steady-state concentration for detection. Low temperature (freezing) and<br />

spin trapping techniques are used to visualize transient radicals in the biological<br />

matrices. To stabilize short-lived radicals, these are trapped with alkyl<br />

nitroso compounds leading to long-lived adducts, making them amenable<br />

to ESR analysis.<br />

Identity and oxidation state of paramagnetic transition metals with<br />

unpaired electrons in their d-shell are detectable by ESR (e.g., Mn 4þ and<br />

Mn 2þ ,Cu 2þ ,Ni 1þ and Ni 3þ ,Ti 3þ , and Fe 3þ ). The nuclear spin of the transition<br />

metal and the natural abundance of the magnetic isotopes assist in this<br />

intent by causing different signal splitting and patterns.<br />

Changes in the structure and properties of biological membranes (e.g.,<br />

skin) under the influence of changing physical parameters or under the influence<br />

of xenobiotic penetrants have been studied by ESR. Perturbation and<br />

conformational changes in natural constituents in the skin, due to experimental<br />

penetration enhancers applied, have been described by incorporating<br />

various (ESR-detectable) stearic acids as spin labels in the lipid bilayers of<br />

human skin (157).


56 Hosty´nek and Maibach<br />

Using skin biopsies, the effects of ultraviolet (UV) irradiation on<br />

epidermal tissue have been studied by ESR spectroscopy and ESR imaging,<br />

providing evidence for the generation of hydroxyl and lipid radicals that<br />

potentially result in oxidative injury in the SC and epidermis (158).<br />

Particle-Induced X-Ray Emission<br />

PIXE analysis with a proton microprobe allows the determination of trace<br />

elements in epidermal strata prepared by cryosection (159). In PIXE a proton<br />

beam activates an atomic electron, lifting it into a higher orbital. When<br />

an outer shell electron falls back to fill the vacancy created, the transition is<br />

measured as the emission of an X-ray photon, characteristic of the excited<br />

atom. The method with an elemental sensitivity approaching 0.1 mg/kg<br />

(0.1 ppm) is also useful in bulk analysis of alloys with multielement detection<br />

capability, and in spatial analysis to localize elements present in a sample<br />

(160). Using this analytical method in their research into skin penetration<br />

by nickel, Forslind et al. and Malmqvist et al. have been able to localize<br />

nickel in the superficial strata of human skin, with a spacial resolution of<br />

3 mm by15mm and a detection limit of about 30 ppm. The concentration<br />

of nickel following exposure to a nickel solution was found to be highest<br />

in the outer parts of the epidermis, mainly in the outermost SC (161,162),<br />

an indication of the metal ion’s reactivity.<br />

SUMMARY AND CONCLUSIONS<br />

While there is a considerable volume of medical and toxicological literature<br />

addressing the topical and systemic effects of metal absorption, the data available<br />

on the rate of their skin diffusivity are few, vary considerably, and are<br />

fragmented and hard to reconcile. In reviewing the determinants for the<br />

diffusion of metals, both exogenous and endogenous, the intent here was to<br />

delineate the numerous interrelated factors involved in that process; a closer<br />

examination of the multitude of variables may serve to explain the difficulties<br />

encountered by investigators in this line of research, and underscores the need<br />

for continued endeavors in that field and for the development of reliable methods<br />

by which to determine exposure toward toxicological risk assessment.<br />

The diffusion process of metals, particularly of the transition metals, is<br />

different from that of small-molecular-weight, lipophilic organic compounds,<br />

such as drugs, solvents, perfumes, etc., which follow Fick’s law of<br />

diffusion and whose Kps can be predicted applying structure–activity relationships.<br />

Development of a mathematical algorithm with which to assess<br />

dermal absorption of metals by a unified approach on the other hand has<br />

not been possible, due to the myriad of variables described here.<br />

For a small number of metals, diffusion has been thoroughly investigated,<br />

primarily motivated by morbidity and mortality, as a consequence


Basics of Metal Skin Penetration: Scope and Limitations 57<br />

of skin exposure occurring intentionally for the purpose of personal embellishment,<br />

or accidentally in the work environment. The process of diffusion<br />

examined from the anatomical viewpoint of the skin barrier and the metallurgical<br />

and physicochemical properties of the permeants are nature given<br />

but, as documented in this review, they present only part of the difficulties<br />

to be overcome when investigating metal diffusion. The other set of difficulties<br />

stems from the experimental approach in examining the process, either<br />

in vitro or in vivo.<br />

In determining skin diffusivity of metals in vitro for toxicological risk<br />

assessment or therapeutic benefit, for instance, it would appear important<br />

not to limit the focus solely on permeant reaching the receptor phase. Analysis<br />

should also include permeant retained in the strata of the skin for topical<br />

bioavailability purposes. In the case of some metals, the more electrophilic<br />

ions in particular, that value may be the preponderant quantity, and a potentially<br />

significant factor for consideration in exposure risk analysis, as it may<br />

be mobilized subsequently through homeostasis. Some metals, such as copper,<br />

zinc, or calcium, form a reservoir that can be mobilized on homeostatic<br />

demand; others again penetrate into the strata by passive diffusion and can<br />

albeit slowly, reach the systemic compartment.<br />

On the other hand, metals of slow diffusivity, such as transition metals,<br />

also encounter a countercurrent effect in the strata of the skin. As that barrier<br />

is characterized by continuous desquamation, with a total turnover over two<br />

to three weeks (corresponding to an approximate ‘‘inverse’’ K p of 10 6 ), bioavailability<br />

of the metal may be less than indicated by absorption in skin tissue,<br />

because epidermal turnover can significantly reduce further diffusion<br />

into the systemic circulation of highly electrophilic compounds.<br />

As illustrated here, the process of skin barrier diffusion by metal ions<br />

is multifactorial, and the rate of their absorption, both in vitro and in vivo,<br />

remains imponderable a priori due to a number of potentially promoting<br />

and retarding processes acting simultaneously.<br />

ABBREVIATIONS<br />

EPA Environmental Protection Agency<br />

ESR electron spin resonance<br />

GI gastrointestinal<br />

HPLC high pressure liquid chromatography<br />

MT metallothionein<br />

NMR nuclear magnetic resonance<br />

QSAR quantitative structure–activity relationships<br />

RA rheumatoid arthritis<br />

SC stratum corneum<br />

UV ultraviolet


58 Hosty´nek and Maibach<br />

REFERENCES<br />

1. Curley A, et al. Dermal absorption of hexachlorophene in infants. Lancet 1971;<br />

2:296–297.<br />

2. Alder VG, et al. Absorption of hexachlorophene from infants’ skin. Lancet<br />

1972; 2:384.<br />

3. Shuman RM, Leech RW, Alvord EC Jr. Neurotoxicity of hexachlorophene in<br />

humans. II. A clinicopathologic study of 46 premature infants. Arch Neurol<br />

1975; 32:320.<br />

4. Siegler RW, Nierenberg DW, Hickey WF. Fatal poisoning from liquid dimethylmercury:<br />

a neuropathologic study. Hum Pathol 1999; 30:720.<br />

5. Cohn JR, Emmett EA. The excretion of trace metals in human sweat. Ann Clin<br />

Lab Sci 1978; 8:270.<br />

6. Aruoma OI, et al. Iron, copper and zinc concentrations in human sweat and<br />

plasma; the effect of exercise. Clin Chim Acta 1988; 177:81.<br />

7. A ˚ gren MS. Studies on zinc in wound healing. Acta Derm Venereol (suppl)<br />

1990; 154:1.<br />

8. A ˚ gren MS. Percutaneous absorption of zinc from zinc oxide applied topically to<br />

intact skin in man. Dermatologica 1990; 180:36.<br />

9. Vinson JA, Proch J. Age-related differences in topical zinc absorption from different<br />

formulations: implications for health and disease. J Geriat Dermatol<br />

1997; 5:276.<br />

10. Keen CL, Hurley LS. Zinc absorption through skin: correction of zinc deficiency<br />

in the rat. Am J Clin Nutr 1977; 30:528.<br />

11. Marzulli FN. Barriers to skin penetration. J Invest Dermatol 1962; 39:387.<br />

12. Scheuplein RJ, Blank IH. Permeability of the skin. Physiol Rev 1971; 51:707.<br />

13. Blank IH, et al. The diffusion of water across the stratum corneum as a function<br />

of its water content. J Invest Dermato 1984; 82:188.<br />

14. Schwindt DA, Wilhelm KP, Maibach HI. Water diffusion characteristics of<br />

human stratum corneum at different anatomical sites in vivo. J Invest Dermatol<br />

1998; 111:385.<br />

15. Kalia YN, Pirot F, Guy RH. Homogenous transport in a heterogeneous membrane:<br />

water diffusion across stratum corneum in vivo. Biophys J 1996; 71:2692.<br />

16. Michaels AS, Chandrasekaran SK, Shaw JE. Drug permeation through human<br />

skin: theory and in vitro experimental measurements. Am Inst Chem Eng J<br />

1975; 21:985.<br />

17. Elias P. Epidermal lipids, barrier function, and desquamation. J Invest Dermatol<br />

1983; 80:S44.<br />

18. Scheuplein RJ. Mechanism of percutaneous absorption. I. Routes of penetration<br />

and the influence of solubility. J Invest Dermatol 1965; 45:334.<br />

19. Hawkins GS, Reifenrath WG. Influence of skin source, penetration cell fluid,<br />

and partition coefficient on in vitro skin penetration. J Pharm Sci 1986; 75:378.<br />

20. Fick AE. On liquid diffusion. Phil Mag 1855; 10:30.<br />

21. Flynn GL. Physicochemical determinants of skin absorption. In: Gerrity TR,<br />

Henry CJ, eds. Principles of Route-to-Route Extrapolation for Risk Assessment.<br />

New York: Elsevier Science Publishing Co., 1990:93.<br />

22. EPA. Dermal exposure assessment: principles and applications. Washington<br />

D.C.: Office of Research and Development, 1992.


Basics of Metal Skin Penetration: Scope and Limitations 59<br />

23. Tanojo H, et al. In vitro permeation of nickel salts through human stratum corneum.<br />

Acta Derm Venereol 2001; 212(suppl):19.<br />

24. Baranowska-Dutkiewicz B. Absorption of hexavalent chromium by skin<br />

in man. Arch Toxicol 1981; 47:47–50.<br />

25. Feldmann RJ, Maibach HI. Percutaneous penetration of steroids in man.<br />

J Invest Dermatol 1969; 52:89.<br />

26. Feldmann RJ, Maibach HI. Absorption of some organic compounds through<br />

the skin of man. J Invest Dermatol 1970; 54:399.<br />

27. Feldmann RJ, Maibach HI. Percutaneous penetration of some pesticides and<br />

herbicides in man. Toxicol Appl Pharmacol 1974; 28:126.<br />

28. Kasting GB, Smith RL, Cooper ER. Effect of lipid solubility and molecular size<br />

on percutaneous absorption. In: Shroot B, Schaefer H, eds. Skin Pharmacokinetics.<br />

Basel: Karger, 1987:138.<br />

29. El Tayar N, et al. Percutaneous penetration of drugs: a quantitative structurepermeability<br />

relationship study. J Pharm Sci 1991; 80:744.<br />

30. Potts RO, Guy RH. Predicting skin permeability. Pharm Res 1992; 9:663.<br />

31. Pugh WJ, Hadgraft J. Ab initio prediction of human skin permeability coefficients.<br />

Int J Pharm 1994; 103:163.<br />

32. Potts RO, Guy RH. A predictive algorithm for skin permeability: the effects of<br />

molecular size and hydrogen bond activity. Pharm Res 1995; 12:1628.<br />

33. Blank IH. Mechanism of percutaneous adsorption: the effect of temperature on<br />

the transport of non-electrolytes across the skin. J Soc Cosmet Chem 1960;<br />

11:57.<br />

34. Scheuplein RJ, Blank IH. Molecular structure and diffusional processes<br />

across intact skin. In: Report to the US Army Chemical R&D Laboratories.<br />

Maryland: Edgewood Arsenal, 1967.<br />

35. Rougier A, Lotte C, Dupuis D. An original predictive method for in vivo<br />

percutaneous absorption studies. J Soc Cosmet Chem 1987; 38:397.<br />

36. Sanderson RT. Chemical Periodicity. New York: Reinhold Publishing Corp.,<br />

1960.<br />

37. Cavelier C, Foussereau J, Massin M. Nickel allergy: analysis of metal clothing<br />

objects and patch testing to metal samples. Contact Dermat 1985; 12:65.<br />

38. Mali JWH, Spruit D, Seutter E. Chelation in human sweat. Clin Chim Acta<br />

1964; 9:187.<br />

39. Guy RH, Maibach HI. Correction factors for determining body exposure from<br />

forearm percutaneous absorption data. J Appl Toxicol 1984; 4:26.<br />

40. Burnette RR, Bagniefski TM. Influence of constant current iontophoresis on the<br />

impedance and passive Naþ permeability of excised nude mouse skin. J Pharm<br />

Sci 1988; 77:492.<br />

41. Burnette RR, Ongpipattanakul B. Location of sites of increased transport<br />

during iontophoresis. J Pharm Sci 1988; 77:132.<br />

42. Cullander C, Guy RH. Sites of iontophoretic current flow into the skin:<br />

identification and characterization with the vibrating probe electrode. J Invest<br />

Dermatol 1991; 97:55.<br />

43. Cullander C, Guy RH. Visualization of iontophoretic pathways with confocal<br />

microscopy and the vibrating probe electrode. Solid State Ionics 1992;<br />

53:197.


60 Hosty´nek and Maibach<br />

44. Odintsova NA. Permeability of the epidermis for lead acetate according to<br />

fluorescence and electron-microscopic studies. Vestn Dermatol Venerol 1976;<br />

9:19.<br />

45. Bos AJ, et al. Incorporation routes of elements into human hair: implications<br />

for hair analysis used for monitoring. Sci Tot Environ 1985; 42:157.<br />

46. Lyon I, Klotz IM. The interaction of epidermal protein with aluminum salts.<br />

J Am Pharm Assn 1958; 47:509.<br />

47. Fiedler HP. Der Schweiss. 2nd ed. Aulendorf: Cantor, 1968.<br />

48. Abramson HA, Gorin MH. The electrophoretic demonstration of the patent<br />

pores of the living human skin in relation to the charge of the skin. J Phys Chem<br />

1940; 44:1094.<br />

49. MacKee GM, et al. Histologic studies on percutaneous penetration with special<br />

reference to the effect of vehicles. J Invest Dermatol 1945; 6:43.<br />

50. Shelley WB, Melton FM. Factors accelerating the penetration of histamine<br />

through normal intact skin. J Invest Dermatol 1949; 13:61.<br />

51. Kanerva L, Estlander T. Occupational allergic contact dermatitis associated<br />

with curious pubic nickel dermatitis from minimal exposure. Contact Dermat<br />

1995; 32:309.<br />

52. Schnuch A, et al. National rates and regional differences in sensitization to<br />

allergens of the standard series. Population-adjusted frequencies of sensitization<br />

(PAFS) in 40,000 patients from a multicenter study (IVDK). Contact Dermat<br />

1997; 37:200.<br />

53. Samitz MH, Katz SA. Nickel–epidermal interactions: diffusion and binding.<br />

Environ Res 1976; 11:34.<br />

54. Fullerton A, Hoelgaard A. Binding of nickel to human epidermis in vitro. Br J<br />

Dermatol 1988; 119:675.<br />

55. Hosty´nek JJ. Factors determining percutaneous metal absorption. Food Chem<br />

Toxicol 2003; 41:327.<br />

56. Hursh JB, et al. Percutaneous absorption of mercury vapor by man. Arch<br />

Environ Health 1989; 44:120.<br />

57. Haudrechy P, et al. Nickel release from stainless steels. Contact Dermat 1997;<br />

37:113.<br />

58. Lampe MA, et al. Human stratum corneum lipids: characterization and regional<br />

variations. J Lipid Res 1983; 24:120.<br />

59. Friberg L, Skog E, Wahlberg JE. Resorption of mercuric chloride and methyl<br />

mercury dicyandiamide in guinea-pigs through normal skin and through skin<br />

pre-treated with acetone, alkylaryl-sulphonate and soap. Acta Derm Venereol<br />

1961; 41:40.<br />

60. Pirot F, et al. In vitro study of percutaneous absorption, cutaneous bioavailability<br />

and bioequivalence of zinc and copper from five topical formulations.<br />

Skin Pharmacol 1996; 9:259.<br />

61. Pirot F, et al. Simultaneous absorption of Cu and Zn through human skin in<br />

vitro. Skin Pharmacol 1996; 9:43.<br />

62. A ˚ gren MS. Influence of two vehicles for zinc oxide on zinc absorption through<br />

intact skin and wounds. Acta Derm Venereol Suppl (Stockh) 1991; 71:153.<br />

63. Sharata HH, Burnette RR. Effect of dipolar aprotic permeability enhancers on<br />

the basal stratum corneum. J Pharm Sci 1988; 77:27.


Basics of Metal Skin Penetration: Scope and Limitations 61<br />

64. Wahlberg JE, Skog E. Nickel allergy and atopy. Threshold of nickel sensitivity<br />

and immunoglobulin E determinations. Br J Dermatol 1971; 85:97.<br />

65. Rothman S. Überempfindlichkeit gegen hartgeld. Dermatol Wochenschr 1930;<br />

90:98.<br />

66. Preininger T. Überempfindlichkeit gegen nickelgeld. Dermatol Wochenschr<br />

1934; 99:1082.<br />

67. Samitz MH, Pomerantz H. Studies of the effects on the skin of nickel and chromium<br />

salts. AMA Arch Ind Health 1958; 18:473.<br />

68. Gollhausen R, Ring J. Allergy to coined money: nickel contact dermatitis in<br />

cashiers. J Am Acad Dermatol 1991; 25:365.<br />

69. Lidén C, et al. Nickel release from tools on the Swedish market. Contact Dermat<br />

1998; 39:127.<br />

70. Lidén C, Carter S. Nickel release from coins. Contact Dermat 2001; 44:160.<br />

71. Fisher AA. Allergic contact urticaria from various metals. 3rd ed. Philadelphia:<br />

Lea and Febiger, 1986.<br />

72. Fregert S. Occupational dermatitis in a 10-year material. Contact Dermat 1975;<br />

1:96.<br />

73. Wass U, Wahlberg JE. Chromated steel and contact allergy. Contact Dermat<br />

1991; 24:114.<br />

74. Flint GN. A metallurgical approach to metal contact dermatitis. Contact Dermat<br />

1998; 39:213.<br />

75. Bergman M, Bergman B, Söremark R. Tissue accumulation of nickel released<br />

due to electrochemical corrosion of non-precious dental casting alloys. J Oral<br />

Rehab 1980; 7:325.<br />

76. Hosty´nek JJ. Nickel-induced hypersensitivity: etiology, immune reactions,<br />

prevention, and therapy. Arch Dermatol Res 2002; 294:249.<br />

77. Menné T, et al. Patch test reactivity to nickel alloys. Contact Dermat 1987;<br />

16:255.<br />

78. Hosty´nek JJ, et al. Human stratum corneum penetration by nickel: in vivo<br />

study of depth distribution after occlusive application of the metal as powder.<br />

Acta Derm Venereol 2001; 212(suppl):5.<br />

79. Sens DA, Simmons MA, Spicer SS. The analysis of human sweat proteins by<br />

isoelectric focusing. I. Sweat collection utilizing the macroduct system demonstrates<br />

the presence of previously unrecognized sex-related proteins. Pediat Res<br />

1985; 19:873.<br />

80. Rasetti L, Cappellaro F, Gaido P. Contribution to the study of saturnism by<br />

inhibited oils. Rass Med Ind Ig Lav 1961; 30:71.<br />

81. Toribara TY, Clarkson TW, Nierenberg DW. Chemical safety: more on working<br />

with dimethylmercury. Chem Eng News 1997; 75:6.<br />

82. Geigy Scientific Tables. 7th ed. Geigy Pharmaceutical, New York: Ardsley, 1975.<br />

83. Jensen O. ‘‘Rusters’’. The corrosive action of palmar sweat: II. Physical and<br />

chemical factors in palmar hyperhidrosis. Acta Derm Venereol (Stockh) 1979;<br />

59:139.<br />

84. Lind PO, et al. Amalgam-related oral lichenoid reaction. Scand J Dental Res<br />

1986; 94:448.<br />

85. Hier SW, Cornbleet T, Bergheim O. The amino acids of human sweat. J Biol<br />

Chem 1946; 166:327.


62 Hosty´nek and Maibach<br />

86. Coltman CA, Rowe NJ, Atwell RJ. The amino acid content of sweat in normal<br />

adults. J Clin Nutr 1966; 18:373.<br />

87. Öhman H, Vahlquist A. The pH gradient over the stratum corneum differs in<br />

X-linked recessive and autosomal dominant ichthyosis: a clue to the molecular<br />

origin of the ‘‘Acid Skin Mantle’’. J Invest Dermatol 1998; 111:674.<br />

88. Schurer NY, Elias PM. The biochemistry and function of stratum corneum<br />

lipids. Adv Lipid Res 1991; 24:27.<br />

89. Feingold KR. The regulation of epidermal lipid synthesis by permeability<br />

barrier requirements. Crit Rev Ther Drug Carrier Syst 1991; 193.<br />

90. Fluhr JW, et al. Impact of anatomical location on barrier recovery, surface pH<br />

and stratum corneum hydration after acute barrier disruption. Br J Dermatol<br />

2002; 146:770.<br />

91. Bennett BG. Environmental nickel pathways to man. In: Sunderman FW, Aitio<br />

A, eds. Nickel in the Human Environment: Proceedings of a Joint Symposium<br />

Held at the International Agency for Research on Cancer, Lyon, France, 8–11<br />

March 1983. Oxford University Press, New York, 1984:487.<br />

92. Ermolli M, et al. Nickel, cobalt and chromium-induced cytotoxicity and intracellular<br />

accumulation in human hacat keratinocytes. Toxicol 2001; 159:23.<br />

93. Fullerton A, et al. Permeation of nickel salts through human skin in vitro. Contact<br />

Dermat 1986; 15:173.<br />

94. Wahlberg JE. Percutaneous absorption of trivalent and hexavalent chromium.<br />

Arch Dermat 1965; 92:315.<br />

95. Fitzgerald JJ, Brooks T. A new cell for in vitro skin permeability studies—<br />

chromium (III)/(VI) human epidermis investigations. J Invest Dermatol 1979;<br />

72:198.<br />

96. Gammelgaard B, et al. Permeation of chromium salts through human skin in<br />

vitro. Contact Dermat 1992; 27:302.<br />

97. Wahlberg JE. Nickel: the search for alternative, optimal and non-irritant patch<br />

test preparations. Assessment based on laser Doppler flowmetry. Skin Res<br />

Technol 1996; 2:136.<br />

98. Hadgraft J, Green PG, Wotton PK. Facilitated percutaneous absorption of<br />

charged drugs. In: Bronaugh R, Maibach HI, eds. Percutaneous Absorption.<br />

New York: Marcel Dekker, 1989:555.<br />

99. Mazzenga GC, Berner B. The transdermal delivery of zwitterionic drugs I: the<br />

solubility of zwitterionic salts. J Contr Rel 1991; 6:77.<br />

100. Mazzenga GC, Berner B, Jordan F. The transdermal delivery of zwitterionic<br />

drugs II: the flux of zwitterionic salts. J Contr Rel 1992; 20:163.<br />

101. Hosty´nek JJ, et al. Human stratum corneum adsorption of nickel salts: investigation<br />

of depth profiles by tape stripping in vivo. Acta Derm Venereol 2001;<br />

212(suppl):11.<br />

102. Wahlberg JE. Nickel chloride or nickel sulfate? Irritation from patch-test<br />

preparations as assessed by laser Doppler flowmetry. Dermatol Clin 1990; 8:41.<br />

103. Bress WC, Bidanset JH. Percutaneous in vivo and in vitro absorption of lead.<br />

Vet Human Toxicol 1991; 33:212.<br />

104. Smith SL. An avoidable tragedy. Occup Hazards 1997; 59:32.<br />

105. Wahlberg JE. Percutaneous absorption from chromium ( 51 Cr) solutions of<br />

different pH, 1.4–12.8. Dermatologica 1968; 137:17.


Basics of Metal Skin Penetration: Scope and Limitations 63<br />

106. Dupuis D, et al. In vivo relationship between horny layer reservoir effect and<br />

percutaneous absorption in human and rat. J Invest Dermatol 1984; 82:353.<br />

107. Hallberg L. Bioavailability of iron in man. Annu Rev Nutr 1981; 123:1981.<br />

108. Wahlberg JE. Percutaneous toxicity of metal compounds. Arch Environ Health<br />

1965; 11:201–204.<br />

109. Alder JF, et al. Depth concentration profiles obtained by carbon furnace<br />

atomic absorption spectrometry for nickel and aluminium in human skin.<br />

J Anal Atomic Spectrom 1986; 1:365.<br />

110. Santucci B, et al. The influence exerted by cutaneous ligands in subjects reacting<br />

to nickel sulfate alone and in those reacting to more transition metals. Exp Dermatol<br />

1998; 7:162.<br />

111. Burch RE, Hahn HKJ, Sullivan JF. Newer aspects of the roles of zinc, manganese<br />

and copper in human nutrition. Clin Chem 1975; 21:501.<br />

112. Christie OHJ, et al. Spark source mass spectrographic study of metal allergenic<br />

substances on the skin. J Invest Dermatol 1976; 67:587.<br />

113. Quatrale RP, Thomas EL, Birnbaum JE. The site of antiperspirant action by<br />

aluminum salts in the eccrine sweat glands of the axilla. J Soc Cosm Chem<br />

1985; 36:435.<br />

114. Wahlberg JE. Percutaneous absorption of sodium chromate (51Cr) cobaltous<br />

(58Co), and mercuric (203Hg) chlorides through excised human and guinea<br />

pig skin. Acta Derm Venereol 1965; 45:415.<br />

115. Maibach HI, et al. Regional variation in percutaneous penetration in man.<br />

Arch Environ Health 1971; 23:208.<br />

116. Rougier A, et al. Regional variation in percutaneous absorption in man:<br />

measurement by the stripping method. Arch Dermatol Res 1986; 278:465.<br />

117. Lotte C, et al. In vivo relationship between transepidermal water loss and percutaneous<br />

penetration of some organic compounds in man: effect of anatomic<br />

site. Arch Dermatol Res 1987; 279:351.<br />

118. Loth H, et al. Statistical testing of drug accumulation in skin tissues by<br />

linear regression versus contents of stratum corneum lipids. Int J Pharm<br />

2000; 209:95.<br />

119. Feldmann RJ, Maibach HI. Regional variation in percutaneous penetration of<br />

C-14 cortisol in man. J Invest Dermatol 1967; 48:181.<br />

120. Kligman AM. Perspectives and problems in cutaneous gerontology. J Invest<br />

Dermatol 1979; 73:39.<br />

121. Hosty´nek JJ. Flux of nickel salts vs. a nickel soap across human skin. In:<br />

Brain KR, Walters KA, eds. Perspectives in Percutaneous Penetration.<br />

Cardiff: STS Publishing, 2002:99.<br />

122. Warner RR, Myers MC, Taylor DA. Electron probe analysis of human skin:<br />

element concentration profiles. J Invest Dermatol 1988; 90:78.<br />

123. Menon GK, Elias PM. Ultrastructural localization of calcium in psoriatic and<br />

normal human epidermis. Arch Dermatol (Chicago) 1991; 127:57.<br />

124. Cousins RJ. Absorption, transport, and hepatic metabolism of copper and zinc:<br />

special reference to metallothionein and ceruloplasmin. Physiol Rev 1985;<br />

65:238.<br />

125. Rugstad NE, Norseth T. Cadmium resistance and content of cadmiumbinding<br />

protein in cultured human cells. Nature 1975; 257:136.


64 Hosty´nek and Maibach<br />

126. Weismann K. Zinc Deficiency and Effects of Systemic Zinc Therapy. Dissertation,<br />

FADL’s Forlag, Kobenhavn, 1980.<br />

127. Holmqvist I. Occupational arsenical dermatitis. A study among employees at a<br />

copper ore smelting work including investigations of skin reactions to contact<br />

with arsenic compounds. Acta Derm Venereol 1951; 31(suppl 26):1–214.<br />

128. Artik S, et al. Nickel allergy in mice: enhanced sensitization capacity of nickel at<br />

higher oxidation states. J Immunol 1999; 163:1143.<br />

129. van Neer FCJ. Reacties op intracutane injecties van drie en zeswaardige chroomverbindgen<br />

bij gesen-sibiliseerde mensen, vackens en caviae. Nederlands Tijdschrift<br />

Geneeskunde 1965; 109:1684.<br />

130. Little MC, Gawkrodger DJ, Macneil S. Chromium- and nickel-induced cytotoxicity<br />

in normal and transformed human keratinocytes: an investigation of<br />

pharmacological approaches to the prevention of Cr(VI)-induced cytotoxicity.<br />

Br J Dermatol 1996; 134:199.<br />

131. Requena CL, Sanchez LM, Coca MS. Argiria generalizada. Estudio clinico-histologico,<br />

histoquimico y ultrastructural. Actas Derm Sif 1985; 76:533.<br />

132. Granstein RD, Sober AJ. Drug- and heavy metal-induced hyperpigmentation.<br />

J Am Acad Dermatol 1981; 5:1.<br />

133. Gummer CL, Hinz RS, Maibach HI. The skin penetration cell: an update. Int J<br />

Pharm 1987; 40:101.<br />

134. Franz TJ. Percutaneous absorption: on the relevance of in vitro data. J Invest<br />

Dermatol 1975; 64:190.<br />

135. Franz TJ. On the relevance of in vitro data. J Invest Dermatol 1975; 64:190.<br />

136. Bronaugh RL, Maibach HI. Percutaneous absorption of nitroaromatic<br />

compounds: in vivo and in vitro studies in the human and monkey. J Invest<br />

Dermatol 1985; 84:180.<br />

137. Spencer TS, et al. Evaporation of diethyltoluamide from human skin in vivo<br />

and in vitro. J Invest Dermatol 1979; 72:317.<br />

138. Reifenrath WG, Robinson PB. In vitro skin evaporation and penetration<br />

characteristics of mosquito repellents. J Pharm Sci 1982; 71:1014.<br />

139. Bulmer FMR, Mackenzie EA. Studies in the control and treatment of nickel<br />

rash. J Industr Hyg 1926; 8:517.<br />

140. Shah PV, Guthrie FE. Dermal absorption of benzidine derivatives in rats. Bull<br />

Environ Contam Toxicol 1983; 31:73.<br />

141. Yang JJ, Roy TA, Mackerer CR. Percutaneous absorption of benzo[a]pyrene in<br />

the rat: comparison of in vivo and in vitro results. Toxicol Ind Health 1986;<br />

2:409.<br />

142. EPAU. Inductively Coupled Plasma—Mass Spectrometry. Washington D.C.:<br />

US EPA, 1986.<br />

143. Rabinowitz MB, Wetherill GW, Kopple JD. Studies of human lead metabolism<br />

by use of stable isotope tracers. Environ Health Persp 1974; 7:145.<br />

144. Brown SS, et al. IUPAC reference method for analysis of nickel in serum and<br />

urine by electrothermal atomic absorption spectrophotometry. Clin Biochem<br />

1981; 14:295.<br />

145. Derry JE, McLean WM, Freeman JB. A study of the percutaneous absorption<br />

from topically applied zinc oxide ointment. J Parent Bulmer Ent Nutr 1983;<br />

7:131.


Basics of Metal Skin Penetration: Scope and Limitations 65<br />

146. Miller SJ. Nutritional deficiency and the skin. J Am Acad Dermatol 1989; 21:1.<br />

147. Tsai JC, et al. Drug and vehicle deposition from topical applications: use of in<br />

vitro mass balance technique with minoxidil solutions. J Pharm Sci 1992; 81:736.<br />

148. Blaschko A. Die Berufsdermatosen der arbeiter, ein beitrag zur gewerbehygiene<br />

(I). Deutsche Med Wochenschrift 1889; 15:925.<br />

149. Cronin E, ed. Nickel, Contact Dermatitis. Edinburgh: Churchill Livingstone,<br />

1980:338.<br />

150. Angelini G, Veña GA. Allergia da contatto al nickel. Considerazioni su vecchie e<br />

nuove acquisizioni. Bollettino Dermatol Allergol Professionale 1989; 4:5.<br />

151. Kiec-Swierczynska M. Allergy to chromate, cobalt and nickel in Lodz 1977–1988.<br />

Contact Dermat 1990; 22:229.<br />

152. Kanerva L, Jolanki R, Toikkanen J. Frequencies of occupational allergic diseases<br />

and gender differences in Finland. Int Arch Occup Environ Health<br />

1994; 66:111.<br />

153. Wester RC, et al. In vivo percutaneous absorption of boric acid, borax, and disodium<br />

octaborate tetrahydrate in humans compared to in vitro absorption in<br />

human skin from infinite and finite doses. Toxicol Sci 1998; 45:42.<br />

154. Reynolds RJ. Atomic Absorption Spectroscopy: A Practical Guide by<br />

R.J. Reynolds and K. Aldous, with a Chapter by K.C. Thompson. New York:<br />

Barnes & Noble, 1970.<br />

155. Sunderman FW Jr. Chemistry, analysis and monitoring. In: Maibach HI, Mennéed<br />

T, eds. Nickel and the Skin: Immunology and Toxicology. Boca Raton,<br />

Florida: CRC Press, Inc., 1989:2.<br />

156. Sunderman FW Jr. Biological monitoring of nickel in humans. Scand J Worker<br />

Environ Health 1993; 19(suppl 1):34.<br />

157. Quan D, Maibach HI. Electronic spin resonance. In: EW Smith, Maibach HI,<br />

eds. Percutaneous Penetration Enhancers. Boca Raton: CRC Press, 1995:427.<br />

158. Herrling T, et al. Electron spin resonance detection of UVA-induced free radicals.<br />

Skin Pharmacol Appl Skin Physiol 2002; 15:381.<br />

159. Cullander C, et al. A quantitative minimally invasive assay for the detection of<br />

metals in the stratum corneum. J Pharm Biomed Anal 2000; 22:265.<br />

160. Johansson SAE, Campbell JL. PIXE—a Novel Technique for Elemental<br />

Analysis. Chichester: Wiley, 1988.<br />

161. Forslind B, et al. Recent advances in X-ray microanalysis in <strong>dermatology</strong>. Scan<br />

Electron Micros 1985; (pt 2):687.<br />

162. Malmqvist KG, et al. The use of PIXE in experimental studies of the physiology<br />

of human skin epidermis. Biol Trace Element Res 1987; 12:297.


4<br />

Percutaneous Absorption of<br />

Copper Compounds<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

INTRODUCTION<br />

Published and unpublished qualitative, semi-quantitative, and quantitative<br />

data on cutaneous absorption of this essential trace element brought in contact<br />

with the skin in its metallic form and its organic and inorganic species<br />

are reviewed. The different approaches that have been used to investigate copper<br />

diffusion and analysis of results are dicussed, as well as problems associated<br />

with their interpretation. Diffusion constants Kp registered range<br />

from 10 7 to 10 3 cm/h. Recognized as a critical endogenous anti-inflammatory<br />

agent in the normal organism, inflammatory conditions induce an<br />

increased demand in the organism for copper; such demand could potentially<br />

be satisfied by dermal exposure. Role and importance of copper’s<br />

anti-inflammatory activity and the possibility of transdermal dosing is<br />

viewed in light of the metal’s skin diffusivity data now available.<br />

The largest part of the literature published on the biology of copper<br />

addresses its importance and function as an essential trace element (ETE) in<br />

living organisms, its biochemistry, and role in nutrition and clinical medicine.<br />

Little has been published on its potential for skin diffusivity. So far, the only<br />

quantitative cutaneous diffusion rates given in the literature refer to experiments<br />

performed with copper compounds on the cat in vitro and in vivo,<br />

and with human skin in vitro as part of dermatological formulations.<br />

67


68 Hosty´nek and Maibach<br />

Transformation of data from those earlier studies, based on certain assumptions,<br />

lead to estimated Kp values of 10 6 –10 5 cm/hr for the copper salts<br />

tested, values which lie at the lower end of skin diffusivity rates measured<br />

for transition metal salts. Interpretation of absorption data so far must rely<br />

on clinical observations.<br />

Quantitative data are difficult to interpret due to the facile and<br />

dynamic changes in speciation potentially occurring in the membrane environment.<br />

Copper can exert its biological role thanks to the lability of its<br />

outer electrons in the 3d and 4s orbital shells, energy levels which overlap,<br />

allowing the element to switch readily from the monovalent to the divalent<br />

oxidation state. The transition thus requires minimal activation energy, and<br />

the shift between Cu(I) and Cu(II) is rendered favorable. Such facile changes<br />

in valence, noted also for chromium and iron, play a decisive role for the<br />

ions’ rate of transfer across cell membranes and epithelia, including the skin.<br />

Copper absorbed from the gastrointestinal tract is bound to albumin as a<br />

Cu(II) ion or as a Cu(II)-amino acid-albumin complex, and circulates in<br />

plasma bound to ceruloplasmin, albumin, or transcuprein. At the cellular<br />

interphase, copper dissociates from the carrier and is reduced to Cu(I) by<br />

plasma membrane reductase; once it has crossed the plasma-membrane<br />

barrier, it continues in the Cu(I) state (1). Such changes in speciations are<br />

presumed to occur in transit through the skin also.<br />

QUALITATIVE DIFFUSION DATA<br />

Earlier data on skin penetration by copper are circumstantial and qualitative<br />

only, derived from clinical observations, pharmacological activity, reports<br />

from dermatological practice, and observations made through use of spectroscopic<br />

methods (Table 1). In those studies, parameters that would allow<br />

calculation of diffusion rates are not available.<br />

Reaction of Metallic Copper in Contact with the Skin<br />

and Absorption of Derivatives Formed There<br />

In prolonged contact with the skin, metallic copper is oxidized, forming copper<br />

salts with amino acids, or soaps with fatty acids naturally present on the<br />

skin surface. This becomes apparent through the discoloration of the skin<br />

and of the copper-releasing objects in the areas of contact. These copper<br />

complexes formed on sustained skin contact appear to penetrate the skin,<br />

and the anti-inflammatory (AI) activity attributed to wearing of copper<br />

metal is attributed to their action, as Cu(II) in these complexes is considered<br />

to be essential for such an effect (11,12).<br />

Corrosion of copper articles, worn as bracelets, in contact with the skin<br />

or on immersion in natural or synthetic sweat was investigated by Walker<br />

and Griffin; weight losses of 0.1% to 0.8% per month were recorded (4).


Percutaneous Absorption of Copper Compounds 69<br />

Table 1 Clinical Signs and Analytical Data Demonstrating Cutaneous<br />

Copper Absorption<br />

References Experiment Species Detection<br />

2 In vivo, copper oleate Human Urine levels<br />

3 In vivo, copper metal Human Serum level<br />

4 In vivo, copper metal Human Metal loss<br />

5 In vitro Human Electron<br />

microscopy<br />

Warner R.R.,<br />

personal<br />

communication,<br />

1993<br />

In vitro, Cu sulfate Human Spectral analysis<br />

6 In vivo, copper<br />

salicylate (Alcusal TM Rat Inflammation<br />

)<br />

arthritis<br />

7 In vivo, copper<br />

salicylate<br />

(Dermcusal TM )<br />

Rat Inflammation<br />

8 In vivo, lipophilic<br />

copper complexes<br />

Rat Inflammation<br />

9 In vivo Cu metal in Serum, scalp hair,<br />

ointment urine; AA, ICP-<br />

MS<br />

10 In vivo Cu metal Inflammation, urine<br />

level<br />

Abbreviations: ICP-MS, inductively coupled plasma-mass spectrometry; AA, atomic absorption.<br />

Absorption of copper as reflected in serum levels has been observed<br />

following the accidental lodging of the finely divided metal in the skin. From<br />

an initial value of 13.0 mmol/L, copper serum levels increased over four days<br />

to 19.4 mmol/L (average control: 17.34 mmol/L), then gradually decreased<br />

again, with an apparent half-life of over 100 days. The shape of the serum<br />

copper plot suggests prolonged, extensive absorption (3).<br />

Absorption of the metal was determined qualitatively on men and women<br />

after a four-week application of an ointment containing 20% elementary<br />

copper. Serum and urine concentrations were determined by atomic activation<br />

spectroscopy and concentrations in hair by inductively coupled plasmamass<br />

spectrometry (ICP-MS). Serum concentration was seen to have increased<br />

significantly, in urine it decreased, and in hair it remained constant (9).<br />

Diffusion of Copper Compounds<br />

Following in vivo application of copper oleate in a lanolin-petroleum base<br />

over 24 hours to human back skin, a significant increase in urinary copper


70 Hosty´nek and Maibach<br />

levels was observed over several days, to a maximum of 0.32 mg/L; determined<br />

for a normal control, that value was 0.11 mg/L (2).<br />

Alcusal TM (copper salicylate ethanolate) in ethanol-glycerin applied<br />

on rat skin in vivo showed AI and anti-arthritic activity (6). Dermcusal TM<br />

(copper salicylate) in dimethyl sulfoxide applied on the skin of rats with<br />

adjuvant-induced polyarthritis was three times as effective as a similar<br />

Alcusal formulation (7). AI activity for a number of lipophilic copper complexes<br />

and phenols in dimethyl sulfoxide was studied by Beveridge et al.;<br />

particularly miflumic acid applied dermally on rats was found to be effective<br />

in reducing experimental arthritis in those animals (8).<br />

Electron microscopy of skin treated topically with copper acetate<br />

revealed that copper initially was localized in the intercellular spaces.<br />

Subsequently, the membranes of viable cells were also penetrated, with accumulation<br />

of the metal in and around the cell nucleus (5).<br />

By electron probe analysis and analytical electron microscopy, the<br />

occurrence of copper was followed across human skin in vitro following<br />

application of copper sulfate using Franz-type diffusion cells. Copper was<br />

observed to enter the outer stratum corneum (SC) cells, then, due to an<br />

apparent barrier occurring approximately halfway within the SC, the level<br />

fell below the detection limit (0.1% by weight), only to reappear again at<br />

the granular interface. There, copper was seen to diffuse by the intercellular<br />

route through the stratum granulosum to reach the stratum spinosum<br />

(Warner R.R., personal communication, 1993). Analysis of coppercontaining<br />

organic compounds applied by those authors on human cadaver<br />

skin by iontophoresis showed high and uniform penetration into the viable<br />

epithelial cells, with approximately half that level present in the dermis.<br />

SEMIQUANTITATIVE DATA<br />

Semiquantitative evidence for the formation of copper complexes and their<br />

diffusion into the superficial skin strata (epidermis) was obtained by occlusion<br />

of finely divided copper powder on the arm of volunteers, followed by stripping<br />

of the exposed areas. Evidence was thus obtained that, over time, copper<br />

derivatives formed with skin exudates were absorbed beyond the SC and<br />

became available to systemic absorption (Hostynek, unpublished data).<br />

Under occlusion with exclusion of air, up to 72-hour exposure, copper<br />

values detected in the strippings were low, reverting to naturally present<br />

levels in those individuals. Under semiocclusion allowing access of air by<br />

covering the skin with ‘‘breathable’’ tape, however, after 72 hours, 0.4–<br />

1.4 mg/cm 2 was seen at the level of the stratum lucidum, and in individual<br />

volunteers cumulative amounts of up to 84 mg/cm 2 copper were present<br />

(area under the curve [AUC], determined by ICP-MS analysis; Table 2).<br />

These observations confirm the occurrence of chemical action of skin exudates<br />

(sweat and sebum) on contacting metals, such as copper, to form in situ


Percutaneous Absorption of Copper Compounds 71<br />

Table 2 Copper (AUC) Removed with Tape Strips After Semiocclusive<br />

Application of Copper Powder on Human Skin<br />

Occlusion time (hr) Mean AUC (ng/cm 2 )<br />

0 (control) 38033<br />

24 45845<br />

48 55960<br />

72 83974<br />

Abbreviation: AUC, area under the curve.<br />

diffusible salts, such as the chloride, pyruvate or lactate, and lipophilic derivatives<br />

(soaps) of likely diffusivity, as demonstrated earlier for nickel (13,14).<br />

QUANTITATIVE DATA<br />

Bis(glycinato) Cu(II) complex was the first copper compound for which quantitative<br />

absorption data became available (15). A radioactive ( 64 Cu) 0.05 M<br />

solution in physiological saline applied to excised cat skin revealed, after a<br />

lag time, a steady-state transport rate described as Kp ¼ 24 10 4 cm/hr.<br />

The percutaneous absorption of copper from copper chloride and copper<br />

sulfate has been investigated in vitro with human skin (16). Formulated<br />

for therapeutic purposes, these salts were administered together with zinc<br />

chloride and zinc sulfate, incorporated in the vehicles petrolatum and two<br />

aqueous gels. For CuSO4 from petrolatum or from Carbopol TM gel and for<br />

CuCl2 from Metolose TM gel, the apparent permeability coefficients were<br />

similar, 0.032–0.045 10 4 cm/hr (Table 3). For CuCl2 from petrolatum,<br />

the permeability coefficient was somewhat higher, 0.023–0.16 10 4 cm/hr.<br />

Skin penetration of commercial products (emulsions and ointments)<br />

containing copper and zinc was investigated in human, dermatomed abdominal<br />

skin (400 mm) in vitro for periods of 72 hours (16). Whether formulated<br />

with copper sulfate or the organic salt copper 2-pyrrolidone 5-carboxylate,<br />

Table 3 Diffusivity of Copper (with Zinc) Salts Through Human Skin In Vitro<br />

Salt Formulation 10 4 K p a (cm/hr) SD<br />

CuSO4 (þZnSO4) Petrolatum 0.032 0.031<br />

CuSO 4 (þZnSO 4) Carbopol 940 TM gel 0.045 0.057<br />

CuCl2 (þZnCl2) Petrolatum 0.16 0.12<br />

CuCl2 (þZnCl2) Metolose 60 SH TM gel 0.023 0.010<br />

a Permeability coefficient Kp calculated from copper in the receptor fluid.<br />

Source: From Ref. 16.


72 Hosty´nek and Maibach<br />

Table 4 Percutaneous Absorption of Copper Compounds from Various<br />

Vehicles Through Human Skin In Vitro<br />

Exposure (hr) 10 4 Kp a<br />

Formulation Compound<br />

Concentration<br />

(mg/cm 3 ) 0–2 24–48 48–72<br />

Experiment No. 1<br />

Emulsion A CuPC 1 0.57 0.02 0.06<br />

Emulsion B CuSO4 1.3 0.44 0.010 0.015<br />

Emulsion C CuSO4 0.5 1.2 0.03 0.08<br />

Experiment No. 2<br />

Emulsion A CuPC 1 1.2 0.06 0.13<br />

Ointment D CuSO4 0.9 0.8 0.05 0.09<br />

Ointment E CuSO 4 0.5 0.9 0.10 0.12<br />

a Permeability coefficient Kp (cm/hr).<br />

Source: From Ref. 17.<br />

apparent permeability coefficients, were in the range 0.015 10 4 –<br />

0.13 10 4 cm/hr (Table 4). Permeability coefficient values decreased over<br />

successive time periods from near 1 10 4 cm/hr (zero to two hours) to<br />

0.1 10 4 cm/hr or less (25–48 hours) and finally to undetectable values in<br />

two cases (49–72 hours). The vehicles were three emulsions (two commercial<br />

products and a custom-made variant of one of the former) and two commercial<br />

ointments. All contained both zinc and copper compounds. Each<br />

formulation was applied at the rate of 16 mg/cm 2 .<br />

1. Emulsion A: water/oil, containing ZnO, ZnPC, and CuPC<br />

2. Emulsion B: same as A except that CuSO4 and ZnSO4 replaced<br />

CuPC and ZnPC<br />

3. Emulsion C: water/oil, containing ZnSO4, ZnO, and CuSO4<br />

4. Ointment D: containing ZnSO4, ZnO, and CuSO4<br />

5. Ointment E: containing ZnO and CuSO4<br />

The absorption rate of copper generally decreased as the experiments<br />

progressed, and for formulations B, C, and E there was little or no absorption<br />

in the final 24 hours.<br />

After 72 hours exposure, there was a significant increase in the average<br />

copper concentrations (140–430% of control values) in the epidermis for<br />

all the formulations. In the dermis, where control concentrations averaged<br />

just 2% to 3% of the epidermal control values, the treatments significantly<br />

increased the copper contents.<br />

Using human abdominal epidermis, the diffusion of copper sulfate<br />

and copper acetate as 1% aqueous donor solutions was compared in vitro<br />

(Hosty´nek, unpublished data). The receptor fluid, high pressure liquid chromatography–pure<br />

water, was collected up to 96 hours after the application


Percutaneous Absorption of Copper Compounds 73<br />

Table 5 Copper a Diffusion Constants Kp Through Human Skin In Vitro<br />

Compound 10 4 K p (cm/hr) Comments References<br />

Sulfate (þZnSO4) 0.032–0.045 Split thickness 16<br />

Sulfate 8.84 Split thickness Unpublished b<br />

Sulfate 0.027 Epidermis Unpublished<br />

Chloride (þZnCl2) 0.023–0.16 Split thickness 16<br />

Acetate 0.019 Split thickness Unpublished<br />

Acetate 0.066 Epidermis Unpublished<br />

Histidinate 1.03 Split thickness Unpublished<br />

Gluconate 2.28 Split thickness Unpublished<br />

Glycinate 24 Full thickness c<br />

15<br />

Glycinate 2.43 Split thickness Unpublished<br />

GHK d<br />

0.76 e<br />

Split thickness Unpublished<br />

GHK 25.1 e<br />

Split thickness Unpublished<br />

GHK 0.002 Epidermis Unpublished<br />

a<br />

Cu(II) compounds.<br />

b<br />

Unpublished data, Hostynek et al.<br />

c<br />

Cat skin.<br />

d<br />

Cu-glycine-histidine-lysine.<br />

e<br />

Kps from separate sets of cells in the same diffusion experiment.<br />

of the donor solutions. The copper concentrations in the receptor fluid were<br />

analyzed using ICP-MS. The Kps measured at steady-state flux were 2.73<br />

0.29 10 6 cm/hr for the sulfate and 6.05 1.19 10 6 cm/hr for the acetate.<br />

Permeability coefficients measured for aqueous copper sulfate and<br />

acetate through human epidermis in vitro are of the order of 10 6 cm/hr.<br />

For copper compounds formulated together with zinc compounds for<br />

therapeutic purposes, applied on dermatomed human skin in vitro in<br />

various vehicles, the apparent penetration coefficients were in the range of<br />

3.2 10 6 –1.6 10 5 cm/hr (Table 5).<br />

DISCUSSION AND CONCLUSIONS<br />

Toxicological considerations and pharmacological evidence for the effects of<br />

exposure to exogenous copper: Aspects of safety of exogenous copper need<br />

to be put in proper perspective to allay concerns on the part of clinicians<br />

not accustomed with the action of copper compounds in the therapy of<br />

rheumatoid arthritis and other degenerative-inflammatory human diseases.<br />

Data available indicate that copper may have noxious effects only following<br />

chronic oral (or parenteral) exposure to high amounts of the metalloelement,<br />

particularly upon chronic oral ingestion with food (e.g., water) that<br />

supplies the organism with more than 5 mg/kg of copper per day (18) or in<br />

the case of prolonged hemodialysis with systems that introduce into the circulation<br />

the metal from copper-containing, semipermeable membranes or


74 Hosty´nek and Maibach<br />

Table 6 Acute Toxicity and Therapeutic Indices of Copper Complexes<br />

Copper complex LD50 (SC) mg/kg CFE PA<br />

Cupric acetate 350 70 –<br />

Cupric anthranilate 750 106 50 750<br />

Cupric aspirinate 760 100 150 760<br />

Abbreviations: TI, therapeutic index; CFE, carrageenan foot edema; PA, polyarthritis.<br />

Source: From Refs. 20 and 21.<br />

copper tubing (19). Thus, a real risk of copper toxicity may exist only if the<br />

administering amounts of the metallo-element certainly are far greater than<br />

those that can be actually used for therapeutic purposes, by transdermal<br />

exposure in particular.<br />

Sorenson investigated the acute toxicity of AI copper complexes<br />

injected subcutaneously in rats (partly listed in Table 6) (20,21). Comparison<br />

with the chelating agent demonstrated that the copper complexes were less<br />

toxic and damaging to the organism than the parent drug.<br />

Cupric sulfate has been used medicinally as emetic, astringent, and<br />

anthelmintic. The intestinal mucosa acts as an efficient barrier; however,<br />

excessive amounts of oral copper salts may lead to death. In the normal<br />

human organism, an efficient homeostatic mechanism controls copper<br />

activity, as most is firmly bound to alpha-ceruloplasmin, and heightened<br />

exposure does not result in disease.<br />

Investigating the cutaneous absorption of copper in the skin, applied<br />

as copper(I) oxide and in its elemental form in an ointment at 20% over a<br />

four-week period, the authors concluded that dermal exposure to copper<br />

at those levels does not present a risk of systemic toxicity, nor does it have<br />

any untoward effects in the skin exposed (9).<br />

LIMITATIONS IN MEASURING COPPER ABSORPTION IN VIVO<br />

Homeostasis: Formation of depots and thus of a secondary barrier, which<br />

inhibits further diffusion can be reversible or irreversible. Examples of the<br />

first kind are essential elements subject to homeostatic control, e.g., copper<br />

or zinc, which are retained by specific storage proteins, such as metallothionein<br />

or, for the former, ceruloplasmin. They are again released upon<br />

physiologic demand (22). Homeostasis may thus compromise the validity<br />

of experimentally determined Kp values for copper. Salts of copper or zinc<br />

present in the SC, epidermis, and dermis are kept in equilibrium by control<br />

mechanisms as part of the overall physiological dynamics of micronutrients,<br />

which regulate their status as free ions. Homeostasis maintains equilibria<br />

TI


Percutaneous Absorption of Copper Compounds 75<br />

necessary for the optimal functioning of the organism in general, and the skin<br />

in particular, as absorption is increased in the copper-deficient organism and<br />

decreases once optimal copper status is established. Dermal absorption<br />

experiments conducted in vivo thus are bound to reflect such controls, which<br />

may counteract passive diffusion and add a degree of uncertainty to permeability<br />

constants measured.<br />

INTERDEPENDENCE OF SYSTEMIC COPPER AND ZINC LEVELS<br />

Levels and activity of zinc in the mammalian organism are closely related to<br />

tissue levels of copper. By inference, it is likely that dermal copper absorption<br />

is also a variable in the function of prevailing zinc levels, and some<br />

observations made as to physiological dynamics of zinc by inference may<br />

reflect on copper also, based on such interrelationship of these two metals.<br />

Measurements of zinc absorption have been contradictory. Apparently its<br />

absorption does not take place by simple diffusion, being regulated by<br />

homeostasis, whereby uptake is inversely related to the individual’s nutritional<br />

status and the actual body burden of the metal. Because copper<br />

and zinc coexist in a competitive relationship, excessive levels of zinc as consequence<br />

of dietary supplementation for therapeutic purposes appear to<br />

determine deficiency in copper, resulting in untoward health effects. The<br />

normal serum copper/zinc ratio of 0.9 to 1.2, for example, decreased to<br />

0.5 in consequence of long-term dietary supplementation with the latter (23).<br />

As an electrophilic metal, copper(II) is highly reactive with electronrich<br />

biological tissues (24) and as applies other transition elements, its<br />

in-depth penetration beyond the superficial layers of the SC will be retarded<br />

by reactivity with sulfhydryl groups in cystein, glutathione, or thioglycolic<br />

acid, which abound in skin tissues.<br />

Limitations in the validity of percutaneous absorption data determined<br />

for copper in vivo are thus due to (i) depot formation, (ii) homeostasis, and<br />

(iii) the interdependence of copper and zinc.<br />

As with most transition metals, skin penetration by copper is subject<br />

to numerous interrelated factors, and its nature as an ETE makes diffusion<br />

experiments difficult and interpretation of results uncertain. This may be an<br />

additional reason why in-depth investigation of copper’s dermatopharmacokinetics<br />

is virtually nonexistent.<br />

Overall evidence on skin penetration by metals available so far points<br />

to a few commonalities, be it in respect to the individual xenobiotic or to<br />

characteristics of the skin barrier and the underlying organism; this makes<br />

a few generalizations and anticipatory statements possible, putting copper<br />

diffusivity in perspective with other transition metals. A challenging determinant<br />

in transition metals is the variability in their speciation, which is<br />

critical for their diffusivity through biological membranes. Mineral salts of<br />

copper (electrolytes) in their ionized form, however, traverse the skin barrier


76 Hosty´nek and Maibach<br />

with permeability coefficients of the order of 10 4 –10 6 cm/hr. For comparison,<br />

the Kp for water has been determined at 10 3 cm/hr.<br />

The present literature review covers only studies conducted by us and<br />

others on normal, healthy, and intact skin, not preconditioned in any way.<br />

The only quantitative data on skin penetration by copper or its compounds<br />

through human skin in vivo or in vitro so far is that acquired in our laboratory<br />

(Table 5).<br />

The information generated in our laboratory and by others earlier on<br />

diffusion characteristics permits the following conclusions.<br />

1. Copper compounds and copper applied as the metal do penetrate<br />

the stratum corneum; however, data on their penetration as measured<br />

so far differ widely and, thus, rate and degree of absorption<br />

remain unpredictable.<br />

2. Transepidermal (primarily intercellular) permeation pathways have<br />

been observed for copper compounds. Penetration via hair follicles,<br />

sebaceous glands, and sweat glands has also been established.<br />

3. Lipophilic organo-copper compounds are more easily absorbed<br />

when compared with coordination complexes and salts.<br />

4. Brought in contact with the skin in the elemental state, copper is<br />

oxidized by exudates present on the skin surface, and the resulting<br />

ion penetrates the SC (as the derivative salt or soap) in a timedependent<br />

fashion.<br />

5. In terms of biological functions, the degree and rate of copper compound<br />

penetration is influenced by: (i) homeostatic mechanisms,<br />

(ii) formation and/or pre-existence of a reservoir, and (iii) regulation<br />

by specific binding proteins (metallothionein or ceruloplasmin).<br />

RECOMMENDATIONS FOR RESEARCH<br />

TO FILL EXISTING DATA GAPS<br />

Evidence is well established for the various functions of copper as an ETE, AI<br />

activity among others. In consideration of the evidence concerning the benefits<br />

resulting from transdermal AI therapy with copper compounds, it would<br />

appear useful to establish a database for the skin penetration of such compounds<br />

using rigorous experimental protocols. That becomes possible by<br />

implementing in vitro diffusion experiments through human skin by accurately<br />

measuring the absorption of copper and its compounds under carefully controlled<br />

conditions of skin membrane selection and preparation, ‘‘permeant’’<br />

concentration, vehicle, area and time of exposure, donor and receptor medium,<br />

and also by analysis of diffusion membrane analysis for retained permeant.<br />

There appears to be a need for research to establish the potential for<br />

dermal absorption of copper through skin-diffusible forms of the metal.<br />

Supplementation in the copper-deficient organism may present much needed


Percutaneous Absorption of Copper Compounds 77<br />

relief for a number of conditions associated with the deficiency in this<br />

ETE: integumentary and skeletal abnormalities, defects in growth and<br />

development, abnormalities in sensory perception (22), the myriad of<br />

manifestations of inflammation and connective tissue disease, such as rheumatoid<br />

arthritis (25). While copper(II) has been administered by traditional<br />

drug delivery systems, intravenously as salts or by intramuscular injection of<br />

Cu(II) complexes, there still appears to be a need for a suitable Cu(II)<br />

complex of ready transdermal transport in order to deliver the element to<br />

intracellular sites where it is needed. Parenteral administration is hazardous<br />

because of local toxicity when long-term therapy is necessary (26). Oral<br />

administration of copper complexes in turn is problematic due to their breakdown<br />

at the low gastric pH, whereby copper ions released from the complex<br />

will be subject to excretion and the highly efficient homeostatic control<br />

mechanism designed to prevent much of the metal to be assimilated (27,28).<br />

While the scarce data reviewed here indicate that copper in the ionized<br />

form may be a permeant too poor to effectively reach the target tissue in<br />

adequate dose, results obtained by Pirot et al. and in our laboratory point<br />

to somewhat higher diffusion rates when skin is exposed to copper complexed<br />

with organic moieties. For a better understanding of the pharmacodynamics<br />

of copper skin penetration, further investigation of its diffusivity<br />

in its different chemical forms would appear useful. In analogy with results<br />

obtained for nickel applied as the metal (14), particularly when associated<br />

with amino acids or skin-identical ligands, topical application of copper as<br />

finely disperse powder for maximum surface activity, e.g., already points to<br />

therapeutically effective penetration rates of lipophilic oxidation products<br />

formed with skin exudates, although oxidation of the metal on the skin to<br />

yield diffusible derivatives may appear to be an ineffective dosage form.<br />

CONCLUSIONS<br />

Observations made in vivo serve to confirm that copper metal on prolonged<br />

contact with skin exudates (sweat and sebum), and in the presence of air forms<br />

in situ diffusible salts such as the chloride, pyruvate or lactate, and lipophilic<br />

derivatives (soaps) of likely diffusivity. The role of the skin as a toxicologically<br />

important route of exposure to environmental agents in general and metal<br />

compounds in particular is hereby underscored. It represents an important<br />

port of entry not only to hazardous materials such as readily oxidized metals<br />

which harbor the potential for serious health effects, but also to essential elements<br />

such as copper, whose release and diffusivity through skin contact has<br />

the potential for beneficial and therapeutic action in the inflamed organism.<br />

These findings may contribute to the acceptance of the long-held belief in<br />

the AI effects of copper metal in direct contact with the skin, in particular, and<br />

also may promote the concept of external AI therapy by patches as alternative<br />

to systemic dosing such as intravenous, intraarticular, or intraperitoneal.


78 Hosty´nek and Maibach<br />

Critical for AI activity may be the supplementation of endogenous copper<br />

with exogenous sources, irrespective of the agent’s nature (i.e., elemental, salt,<br />

complex, or as a covalent derivative), because endogenous formation of the<br />

chelate of maximum biological activity will be formed under homeostatic control.<br />

The mechanism of copper complexes acting as AI agents is not known in<br />

all its details, yet evidence so far points to the possible formation of a unique,<br />

as yet undefined metabolite responsible for the observed clinical AI effect.<br />

GLOSSARY<br />

Absorption Uptake into the organism.<br />

Adsorption Material adhering to the skin surface.<br />

Corrosion Electrochemical process involving redox<br />

reactions in the presence of electrolytes.<br />

Electromotive series Arrangement of metals by decreasing order in<br />

their ability to oxidize.<br />

Ionization potential Energy (electron volts, ev) required to remove<br />

an electron from its atomic orbit, with the<br />

value for the Standard Hydrogen Electrode<br />

set at 0.00 ev as an arbitrarily selected<br />

standard reference.<br />

Ligand Atom, ion or functional group bonded to<br />

a central atom, usually a metal, to form a<br />

complex.<br />

Oxidation Process of electron removal from an atom or<br />

ion (e.g., the increase in the proportion of<br />

oxygen in a compound).<br />

Oxidation potential Electrical driving force toward electron loss,<br />

expressed as a potential value (in electron<br />

volts, ev).<br />

Penetration Passive diffusion process of a solute<br />

through skin.<br />

Permeation Diffusion through one or several layers.<br />

Reduction Process of electron gain by an atom or an ion<br />

(e.g., the increase in the proportion of<br />

hydrogen in a compound).<br />

Speciation Valence state; organic vs. inorganic ligand<br />

bonds.<br />

ABBREVIATIONS<br />

AA atomic absorption<br />

AAS atomic activation spectroscopy<br />

AI anti-inflammatory


Percutaneous Absorption of Copper Compounds 79<br />

AUC area under the curve<br />

ETE essential trace element<br />

HPLC high pressure liquid chromatography<br />

NMR nuclear magnetic resonance<br />

SC stratum corneum<br />

UV ultraviolet<br />

REFERENCES<br />

1. Elam JS, et al. Copper chaperones. Adv Protein Chem 2002; 60:151.<br />

2. Schmid R, Winkler J. Über die Kutane Kupferresorption aus einer Kupfer<br />

Enthaltenden Salbe. Klin Wochenschr 1938; 17:559.<br />

3. Bentur Y, et al. An unusual skin exposure to copper; clinical and pharmacokinetic<br />

evaluation. J Toxicol Clin Toxicol 1988; 26:371.<br />

4. Walker WR, Griffin BJ. The solubility of copper in human sweat. Search 1976;<br />

7:100.<br />

5. Odintsova NA. Permeability of human skin to potassium and copper ions and<br />

their ultrastructural localization. Chem Abstr 1978; 89:360.<br />

6. Walker WR, Beveridge SJ, Whitehouse MW. Antiinflammatory activity of<br />

a dermally applied copper salicylate preparation (Alcusal). Agents Actions<br />

1980; 10:1.<br />

7. Beveridge SJ, Walker WR, Whitehouse MW. Anti-inflammatory activity of<br />

copper salicylates applied to rats percutaneously in dimethyl sulphoxide with<br />

glycerol. J Pharm Pharmacol 1980; 32:425.<br />

8. Beveridge SJ, Whitehouse MW, Walker WR. Lipophilic copper(II) formulations:<br />

some correlations between their composition and anti-inflammatory/anti-arthritic<br />

activity when applied to the skin of rats. Agents Actions 1982; 12:225.<br />

9. Gorter RW, Butorac M, Cobian EP. Examination of the cutaneous absorption<br />

of copper after the use of copper-containing ointments. Am J Ther 2004; 11:<br />

453–458.<br />

10. Fat L, Gyorffy L. Occupational dermatitis due to copper exposure. Proceedings<br />

of the OEESC conference, 2002:51–52.<br />

11. Walker WR, Beveridge SJ, Whitehouse MW. Dermal copper drugs: the copper<br />

bracelet and Cu(II) salicylate complexes. Agents Actions (Suppl) 1981; 8:359.<br />

12. Walker WR, Keats DM. An investigation of the therapeutic value of the<br />

‘‘Copper Bracelet’’—dermal assimilation of copper in arthritic/rheumatoid conditions.<br />

Agents Actions 1976; 6:454.<br />

13. Hostynek JJ. Flux of nickel (II) salts vs. a nickel (II) soap across human skin,<br />

in vitro. Exog Dermatol 2003; 2:216–222.<br />

14. Hostynek JJ, et al. Human stratum corneum penetration by nickel: in vivo study<br />

of depth distribution after occlusive application of the metal as powder. Acta<br />

Derm Venereol (Suppl) 2001; 212:5.<br />

15. Walker WR, et al. Perfusion of intact skin by a saline solution of bis(glycinato)<br />

copper(II). Bioinorg Chem 1977; 7:271.<br />

16. Pirot F, et al. Simultaneous absorption of Cu and Zn through human skin<br />

in vitro. Skin Pharmacol 1996; 9:43.


80 Hosty´nek and Maibach<br />

17. Pirot F, et al. In vitro study of percutaneous absorption, cutaneous bioavailability<br />

and bioequivalence of zinc and copper from five topical formulations. Skin<br />

Pharmacol 1996; 9:259.<br />

18. Aggett PJ, Fairweather-Tait S. Adaptation to high and low copper intakes: its<br />

relevance to estimate safe and adequate daily dietary intakes. Am J Clin Nutr<br />

1998; 6:1061S–1063S.<br />

19. Scheinberg HI, Sternlieb I. Copper toxicity and Wilson’s disease. Trace Elements<br />

in Human Health and Disease, Vol. 1; Zinc and Copper, New York: Academic<br />

Press, 1976:415–438.<br />

20. Sorenson RJ. Some copper coordination compounds and their antiinflammatory<br />

and antiulcer activities. Inflammation 1976; 1:317–331.<br />

21. Sorenson RJ. Copper chelates as possible active forms of the antiarthritic agents.<br />

J Med Chem 1976; 19:135–148.<br />

22. Burch RE, Hahn HKJ, Sullivan JF. Newer aspects of the roles of zinc, manganese<br />

and copper in human nutrition. Clin Chem 1975; 21:501.<br />

23. Abdulla M. Copper levels after oral zinc. Lancet 1979; 1:616.<br />

24. Oster G, Salgo MP. The copper intrauterine device and its mode of action.<br />

N Engl J Med 1975; 293:432–438.<br />

25. Rafter GW. Rheumatoid arthritis: a disturbance in copper homeostasis. Med<br />

Hypoth 1987; 22:245.<br />

26. Perrin DD, Whitehouse MW. Metal ion therapy: some fundamental considerations.<br />

In: Rainsford KD, Brune K, Whitehouse MW, eds. Trace Elements in the<br />

Pathogenesis and Treatment of Inflammation. Basel: Birkhauser, 1981.<br />

27. Underwood EJ. Trace Elements in Human and Animal Nutrition. 4th ed.<br />

New York: Academic Press, 1977.<br />

28. Williams DR, Furnival C, May PM. Computer analysis of low molecular weight<br />

copper complexes in biofluids. In: Sorenson JRJ, ed. Inflammatory Diseases and<br />

Copper. Clifton: Humana Press, 1982; 45:45–55.


5<br />

Diffusion of Copper Through<br />

Human Skin In Vivo<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

Frank Dreher<br />

Neocutis, Inc., San Francisco, California, U.S.A.<br />

INTRODUCTION<br />

Sequential tape stripping was implemented on healthy volunteers to examine<br />

the diffusion of copper through human stratum corneum in vivo following<br />

application of the metal as powder on the volar forearm for periods of up to<br />

72 hours.<br />

Exposure sites were stripped 20 times and the strips analyzed for metal<br />

content by inductively coupled plasma–mass spectroscopy with a detection<br />

limit for copper of 0.5 ppb.<br />

Untreated skin was stripped in the same fashion to determine baseline<br />

copper levels for comparison with exposure values resulting from exposure<br />

in respective volunteers.<br />

This chapter, in part, was reprinted from Hosty´nek JJ, Maibach HI, Dreher F. Human stratum<br />

corneum penetration by copper: in vivo study after occlusive and semiocclusive application of<br />

the metal as powder. Food Chem Toxicol (in press), with permission of Elsevier.<br />

81


82 Hosty´nek et al.<br />

Under occlusion with exclusion of air, up to 72 hours copper values<br />

decreased from the superficial to the deeper layers of the stratum corneum<br />

with gradients increasing commensurately with occlusion time, characteristic<br />

of passive diffusion processes. From the 10th strip on, however, levels reverted<br />

to background values.<br />

Under semiocclusion allowing access of air by covering the skin with<br />

‘‘breathable’’ tape, initial copper values lay significantly above baseline values<br />

and concentration gradients increased proportionally with occlusion time. At<br />

72 hours, from the 10th to the 20th strip reaching the glistening epidermal layer,<br />

copper values continued at constant levels, significantly above baseline values.<br />

The results indicate that, in contact with skin, copper will oxidize and<br />

may penetrate the stratum corneum after forming an ion pair with skin exudates.<br />

The rate of reaction seems to depend on contact time and availability<br />

of oxygen. A marked interindividual difference was observed in baseline<br />

values and amounts of copper absorbed.<br />

Arthritis patients have worn copper jewelry of various types for thousands<br />

of years for the relief of inflammation and musculoskeletal disorders.<br />

Their use continues today as folk remedy. Data documenting statistical significant<br />

amelioration of arthritic conditions in patients wearing copper<br />

objects in intimate contact with skin as compared with the effect of placebo<br />

objects, however, are missing to date.<br />

Indicative of the process of skin penetration by certain metals is the<br />

short-term onset of skin reactions, primarily immunologic or nonimmunologic<br />

irritation, resulting from contact with coinage, tools, jewelry, or other<br />

articles of daily use. Major skin reactions to metals involve nickel and chromium<br />

(1,2). Few reports suggest skin reactions to copper also (3–6).<br />

It remains to be demonstrated that copper metal in contact with the<br />

skin does diffuse through the SC to reach the nucleated epidermis, becoming<br />

locally or systemically available. The critical factor in determining if a metal<br />

will penetrate the SC is believed to be the formation of soluble compounds.<br />

In the presence of oxygen, skin exudates may lead to electrochemical reactions<br />

resulting in metal oxidation (corrosion) to form potentially skin-diffusible<br />

compounds with naturally present skin-identical anions such as chloride ion,<br />

with amino acids or fatty acids present on the skin surface (7,8). Such reaction<br />

usually becomes apparent through the discoloration of the skin of the wearer<br />

and of the copper-releasing objects in the areas of contact.<br />

Besides metallurgical properties of the metal (composition, surface<br />

structure, and finish), occurrence and rate of such corrosion will depend<br />

on individual variables: amount and composition of exudates, pH, temperature,<br />

and the availability of oxygen.<br />

So far, this phenomenon of copper metal dissolution, its uptake into the<br />

SC, and its clinical effects has been supported by a number of investigations.<br />

After immersion of copper turnings in human sweat over 24 hours, the metal<br />

concentration increased by an average of two orders of magnitude from the


Diffusion of Copper Through Human Skin In Vivo 83<br />

natural level: from 1–3.5 10 5 to 0.6–3.4 10 3 M (9). Copper complexes<br />

generated on sustained contact of the metal with animal skin appeared to<br />

exert anti-inflammatory (AI) activity in the animals; that was attributed<br />

to the action of copper compounds formed in situ and their cutaneous absorption<br />

(10). By electron microscopy, Odintsova (11) observed the presence of<br />

copper in intercellular spaces, and ultimately around the cell nucleus in<br />

human skin upon exposure to Cu(II) acetate. Absorption of copper has been<br />

observed indirectly following the accidental lodging of the finely divided<br />

metal in human skin dermatoglyphics. Serum levels were found to increase<br />

over four days, and then gradually decrease with an apparent half-life of over<br />

100 days (12). A strong argument in favor of the therapeutic activity of elemental<br />

copper brought in intimate contact with the organism was provided<br />

earlier in an animal study that demonstrated the AI properties of copper<br />

metal present in the organism: a clear prophylactic effect was achieved when<br />

experimental inflammation failed to be induced in rats in which copper metal<br />

had been implanted two months prior to the experiment (13).<br />

It remains to be demonstrated that copper penetrates the skin upon<br />

exposure to the metal in vivo, and thus potential AI effects could be achieved<br />

by dermal exposure also in humans. In order to arrive at a conclusive assessment<br />

of skin penetration by copper, it appears useful to obtain evidence of<br />

cutaneous penetration on contact by use of quantitative metal analysis.<br />

The purpose of the present investigation into the fate of metallic copper<br />

brought in intimate contact with the skin was (i) to obtain (semiquantitative)<br />

evidence for the formation of copper complexes that diffuse and are detectable<br />

by the presence of copper in the SC, (ii) to trace the path and kinetics of<br />

copper diffusion through the SC, (iii) to assess adsorption and reservoir<br />

formation by the penetrating metal in the SC, and (iv) obtain evidence that<br />

if copper absorbed over time it could penetrate beyond the SC and become<br />

available to systemic absorption. Credence could thus be conferred to the<br />

contended benefits of relief from musculoskeletal disorders attributed to<br />

the wearing of copper jewelry, as the potential for the metal’s activity as an<br />

AI agent by dermal exposure in humans so far is still subject of controversy.<br />

This report illustrates the process of copper diffusion following application<br />

of finely distributed (micronized) copper metal on the skin, through<br />

intimate contact under occlusion and semiocclusion. It thus becomes possible<br />

to estimate actual copper concentration profiles, to the level of the living epidermal<br />

tissue and presumed subsequent uptake by dermal microcirculation.<br />

Determining kinetics and penetration depth of permeants by tracing<br />

the concentration profiles in SC has been rendered facile by using the<br />

virtually noninvasive method of SC stripping with adhesive tape and by<br />

the possibility to detect minute quantities of metals using ICP-MS analysis<br />

(14–19). It thus becomes possible to analyze for the presence of elements<br />

such as copper in skin and other biological materials, with detection limits on<br />

the order of 0.5 ppb (20), making the use of radioisotopes unnecessary.


84 Hosty´nek et al.<br />

Investigated by Schwindt et al. (21), on average each tape strip removes<br />

one layer of corneocytes, of approximate 0.5 mm thickness, and after the first<br />

2–3 strips, for a given skin site and test subject each subsequent tape<br />

strip removes the same amount of SC, down to approximately the 20th strip<br />

(r 2 ¼ 0.99). Although furrows in the SC account for the uncertainty in singlelayer<br />

stripping (22), by progressive SC removal with 20 strips it nevertheless<br />

becomes possible to estimate actual copper concentration profiles to the<br />

level of the living epidermal tissue.<br />

EXPERIMENTAL<br />

Subjects<br />

Healthy Caucasian volunteers between the ages of 34 and 69 without evident<br />

dermatological disease participated after giving informed consent. The<br />

study was approved by the University of California Committee on Human<br />

Research. The volar forearm was selected as study site. Three replicates on<br />

adjacent sites were conducted on the same volunteer.<br />

Copper Application, Occlusion, Decontamination, and Stripping<br />

For the purpose of determining baseline copper values, the naturally occurring<br />

copper in the skin of the volunteers was determined by stripping<br />

untreated skin, with three iterations for each occlusion period, on the volar<br />

forearm of the participating volunteers. For occlusion of the SC, plastic<br />

chambers of 12 mm diameter (1.15 cm 2 ) were placed on the premarked area<br />

of the flexor surface on the arm, the chamber covered with the dressing and<br />

left for the predetermined length of time (24, 48, and 72 hours). The occlusive<br />

and semiocclusive application systems consisted of a plastic chamber<br />

(Hill-Top Res., Inc., Cincinnati, U.S.A.) and micropore semiocclusive tape<br />

(3M Health Care, St. Paul, Minnesota, U.S.A.), respectively.<br />

Copper powder [99.7% 3 mm particle size (Aldrich Chem. Co., Milwaukee,<br />

Wisconsin, U.S.A.)] was applied in triplicate on three volunteers. Prior to<br />

application, skin sites were cleansed with deionized water and dried using cotton<br />

swabs. Copper powder was applied on the volar forearm between wrist<br />

and antecubital fossa. The experiment was conducted during the months<br />

of September through April, whereby the exposed skin of the volunteers<br />

exhibited no noticeable tanning. Twenty-five milligrams of copper powder<br />

were placed on a plastic chamber; the chamber then placed on the premarked<br />

area of the flexor surface of the arm of the volunteer, and left<br />

undisturbed for the predetermined length of time. At the end of the exposure<br />

period, the occlusive materials were removed, the site carefully<br />

washed with a metal-complexing detergent containing a 5% solution of<br />

Extran 300 TM (EM Science, Gibbstown, New Jersey, U.S.A.), using cotton<br />

swabs to remove all traces of metal left on the skin surface, then rinsed


Diffusion of Copper Through Human Skin In Vivo 85<br />

with water. The skin site was dried by tapping with cotton balls and finally<br />

by passing an air stream over the surface for one minute.<br />

Before tape stripping, the area of metal contact was marked with a pen<br />

and a template (Transpore TM tape with a 1.2 cm diameter hole) was applied,<br />

to guarantee that SC was removed only from the application site. After<br />

increasing intervals of occlusion, the area of application was stripped sequentially<br />

20 times after covering the 1.15 cm 2 treated area with a 1 in (6.45 cm 2 )<br />

of the precut adhesive tape strips. Polypropylene tape with a backing of<br />

pressure-sensitive acrylate adhesive (Transpore, 3M Health Care, St. Paul,<br />

Minnesota, U.S.A.) of 2.54 cm width was used for stripping. Constant and<br />

uniform pressure (100 g/cm 2 ) was applied on the tape for five seconds, which<br />

was then gradually removed in one slow draw (17). The tape with adhering<br />

SC were placed individually in scintillation vials, 5 mL of concentrated nitric<br />

acid (70%; EM Science, Gibbstown, New Jersey, U.S.A.) was added and the<br />

vials agitated vigorously for three hours on a rotary shaker (Gyrotory TM<br />

water bath model G76; Edison, New Jersey, U.S.A.). The acid solution<br />

was then diluted 20-fold with water for ICP-MS analysis.<br />

Metal Analysis<br />

Samples were prepared by diluting the samples from the stripping experiment<br />

at 1:10 (volume/volume) using 2% HNO3 immediately prior to analysis.<br />

Standard solutions were prepared by diluting a 1000 g/mL Cu stock solution<br />

with 2% HNO3. A six-point external calibration curve (r 2 > 0.999) was used<br />

for quantitation for both Cu isotopes ( 63 Cu and 65 Cu), with 89 Y being<br />

used as an internal standard. The ICP-MS was an Agilent 7500c spectrometer<br />

operated under He collision gas mode. The instrumental detection<br />

limit was 1000 ng/mL), a Perkin-Elmer 4300 dual view ICP optical emission spectrometer<br />

was used for analysis.<br />

Statistical Analysis<br />

Analyses of the AUC were performed using two-sided, paired Student t-test.<br />

The probability value, P < 0.05, was considered significant.<br />

RESULTS<br />

General<br />

Due to the passive nature of the diffusion process, for all subjects a decreasing<br />

concentration gradient from the horny layer to the subcutaneous tissue<br />

became evident (Figs. 1 and 2). This concentration gradient was steep in<br />

the outermost layers of the SC due to its barrier function (Schaefer<br />

and Redelmeier, 1996) (39) and became less so towards the viable layers<br />

of the epidermis.


86 Hosty´nek et al.<br />

Figure 1 Plots of average values for copper removed by sequential tape stripping on<br />

the ventral forearm of three human volunteers after increasing periods of occlusion,<br />

as assessed by ICP-MS. Data points correspond to strip nos. 2, 3, 5, 10, 15, and 20.<br />

Figures 1–2 show copper content for strips nos. 2, 3, 5, 10, 15, and 20<br />

as a function of type and duration of occlusion, as resulting from ICP-MS<br />

analysis. Individual graphs are presented to illustrate the individually differing<br />

levels of copper naturally present in the skin of volunteers.


Diffusion of Copper Through Human Skin In Vivo 87<br />

Figure 2 Plots of average values for copper removed by sequential tape stripping<br />

on the ventral forearm of three human volunteers after increasing periods of<br />

semiocclusion, as assessed by ICP-MS. Data points correspond to strip nos. 2, 3,<br />

5, 10, 15, and 20.<br />

Copper Penetration Under Occlusion<br />

For purposes of range finding, occlusion was initially set at periods from<br />

minutes to 24 hours, as in earlier studies on nickel (23). However, over that


88 Hosty´nek et al.<br />

period copper values did not exceed background values, possibly due to<br />

greater corrosion resistance of copper on skin contact.<br />

Figures 1A–C show copper values in sequential SC strips in terms of<br />

amount per unit area of tissue, presented for individual volunteers and<br />

increasing periods of occlusion. Notable in individual tracings are the<br />

differing baseline levels of copper naturally present in the skin of volunteers.<br />

Only the initial three strips were analyzed for baseline.<br />

For all periods of occlusion, initially profiles were slightly elevated<br />

above baseline. By the 10th strip, however, copper levels had reverted to<br />

baseline values.<br />

Copper Penetration Under Semiocclusion<br />

Figure 2 shows copper values in sequential SC strips in terms of amount<br />

per unit area of tissue, recorded for individual volunteers and increasing periods<br />

of occlusion. Again, the individual tracings show the differing baseline<br />

levels of copper (strips 2–20) naturally present in the skin of volunteers.<br />

Copper values decreased from the superficial SC layers to the deeper layers,<br />

the gradients increasing commensurately with occlusion time (24, 48, and 72<br />

hours). After the 10th strip, the copper content in the SC approximated<br />

baseline levels, but at 72 hours occlusion continued above baseline values.<br />

The AUC values for the semioccluded experiments and their statistical<br />

significance are given in Table 1.<br />

DISCUSSION<br />

Results of this Study<br />

In our study of copper metal exposure, data obtained for the participating<br />

volunteers reveal a striking interindividual diversity in copper levels naturally<br />

occurring in the same anatomical area of the SC, determined before<br />

application of copper powder. Also the variable rate of SC penetration by<br />

Table 1 Copper Removed from Human Volar Lower Arm After<br />

Semiocclusion<br />

Occlusion time Mean AUC a (ng/cm 2 ) SD p-value<br />

0 38,033 18,086<br />

24 45,845 0.11<br />

48 55,960 0.03 b<br />

72 83,974 0.08<br />

a Average area under the curve (AUC) of triplicate stripping experiments on<br />

three volunteers removed by strips nos. 2–20.<br />

b Significant difference in copper removed after 48 hours versus background.


Diffusion of Copper Through Human Skin In Vivo 89<br />

the metal appears due to individual differences in metal oxidation in the<br />

microenvironment prevailing in the area of contact (chemical, i.e., exudates<br />

composition and sweating rate, and physical, i.e., ambient and skin<br />

temperature). A literature search for copper levels in the SC provided no<br />

information that could be used for direct comparison with our results. Only<br />

data on whole skin and hair were found.<br />

The normal copper concentration in healthy human skin tissue appears<br />

to vary according to anatomical site from 1 to 7 ppm dry weight, as determined<br />

by neutron activation analysis of biopsies from 15 individuals (24).<br />

The levels and distribution of copper in human skin (dermis and epidermis)<br />

was also investigated (25). The mean values determined by atomic absorption<br />

in dry tissue varied from 0.88 to 2.60 ppm, with a median value of 1.38 ppm.<br />

Using nuclear microscopy, hair from three normal subjects showed copper<br />

concentrations of 18 6 ppm in the cortex of hair, with marked increases<br />

in concentration at the periphery (approximately 100 ppm) (26). AA spectroscopy<br />

was used to analyze for copper content of hair in relation to age.<br />

Starting at 13 ppm at age 2, copper content increased to 60 ppm by the<br />

age of 12, and decreased again to levels of 10 ppm at age 80 (27).<br />

Discussion of natural levels of that essential trace element prevailing in<br />

the integument and, in particular, of those encountered in the volunteers<br />

participating in the present study lies outside the scope of this paper. Interindividual<br />

variability in the SC diffusion by copper, essentially going back to the<br />

rate of oxidation in the microenvironment of an individual’s skin, on the other<br />

hand, is readily explained, based on previous observations (28,29). Oxidation<br />

of a given metal in contact with the skin is a multifactorial process, as it will<br />

vary in function of prevailing environmental conditions, the skin’s pH, sweating<br />

rate, and the variability in the individual’s amount and composition of<br />

sebum, sweat, and salts, which is, in turn, a function of gender and age (30–36).<br />

The soluble metal complexes formed are capable of penetrating the<br />

corneocyte envelope, forming a reservoir of the metal in the outermost<br />

layers. The process of oxidation appears to be slower under occlusion,<br />

due to limited access of air (oxygen). As became evident from these in vivo<br />

experiments, availability of oxygen is one critical factor in the oxidation process<br />

of metals. In the presence of sweat as electrolyte, the electrochemical<br />

oxidation of copper leads to the formation of cupric ions (Cu 2þ ). This is<br />

coupled with the concurrent reduction of oxygen, which with water forms<br />

hydroxyl ions. In the absence of oxygen, the reaction cannot proceed and<br />

cupric ions are not liberated. One observation in this study points to the difference<br />

in ionization potential and thereby corrosivity of copper when compared<br />

to that of nickel described earlier (29).<br />

In general terms, oxidation of a metal to the ionic form and liberation<br />

of electrons proceeds according to Eq. (1):<br />

M ! M xþ þ x e<br />

ð1Þ


90 Hosty´nek et al.<br />

and the reduction of the (obligatory) oxygen present to form water is represented<br />

in Eq. (2):<br />

O2 þ 4H þ þ 4e ! 2H2O ð2Þ<br />

The difference in corrosion rates between the two metals is expressed<br />

as the standard oxidation potential in the equations below, characterizing<br />

the oxidation for elementary nickel [Eq. (3)] and elementary copper [Eq. (4)],<br />

respectively. While nickel, brought in intimate contact with the skin under<br />

occlusion, reacted to form skin-diffusible derivatives within minutes of<br />

exposure, copper seems not to significantly diffuse for up to 24 hours. This<br />

may be explained by the differences in oxidation potential between nickel<br />

(þ0.23 ev) and copper ( 0.34 ev), nickel thus being more easily oxidizable.<br />

This should not come as a surprise since copper belongs to the select group<br />

of the so-called noble, i.e., nonreactive metals.<br />

Nið0Þ !Ni þ2 þ 2e þ 0:23 ev ð3Þ<br />

Cuð0Þ !Cu þ2 þ 2e 0:34 ev ð4Þ<br />

Other metals besides copper, particularly when they are in intimate<br />

contact with other metals in alloys forming a galvanic element, are electrolytically<br />

oxidized by the skin’s sweat and sebum, forming skin-diffusible<br />

compounds: e.g., nickel and references therein; beryllium, cobalt, chromium,<br />

and mercury, and references therein (28,29). At the other end of the spectrum<br />

in the electromotive series, under specific circumstances skin reactions<br />

have also been attributed to gold in contact with skin, a metal even less reactive<br />

than copper (37). Positive patch test reactions were recorded in patients<br />

with facial and eyelid dermatitis that were ascribed to gold released from<br />

jewelry, apparently promoted by sweat and associated with the abrasive<br />

action of titanium dioxide in cosmetics and sunscreens. A contributing<br />

factor is presumed to be static absorption of loosened metal particles to titanium<br />

dioxide, which can act as adjuvant promoting penetration of skin<br />

strata (38). Nederost and Wagman concluded that a subset of gold-allergic<br />

patients would benefit from avoidance of gold jewelry coming in contact<br />

with skin to which cosmetics were applied containing titanium dioxide and<br />

becoming subject to substantial friction.<br />

The directionally increasing slope of copper concentration profiles with<br />

increasing time of exposure, traced up to 72 hours of occlusion, indicates<br />

a slow but gradual formation of a depot in the outer SC due to its barrier<br />

function (39). Such concentration gradients are typical for a passive diffusion<br />

process, as there are no active transport mechanisms involved through<br />

skin. Copper ions are highly electrophilic and readily complex with SC<br />

proteins, leading to such depot formation. Such depots in the SC merit<br />

consideration for purposes of risk assessment. In vitro diffusion experiments<br />

with several copper complexes showed that a substantial portion of the


Diffusion of Copper Through Human Skin In Vivo 91<br />

permeant is retained in the SC, epidermis, and dermatomed skin used for<br />

that purpose (Hostynek JJ, unpublished data). Such buildup in the SC is<br />

a fair indication that the exogenous agent (copper) eventually may become<br />

available systemically. For that reason also, measuring diffusion rates only<br />

presents part of the overall picture, as the chemical absorbed into the SC<br />

may continue to diffuse into the viable tissues, even after exposure has<br />

stopped. Further absorption of such material, appropriately referred to as<br />

the SC reservoir (40), is the result of a counter-current process that includes<br />

desquamation versus release.<br />

Results confirm that absorption of copper into the SC does occur.<br />

Because copper levels above baseline values, albeit approaching background<br />

and after prolonged semiocclusion, are still seen at the level of the 20th strip<br />

(0.4–1.4 mg/cm 2 ), the level of the stratum lucidum. This is taken as an indication<br />

that the metal has penetrated beyond the deepest SC layers, and thus<br />

is likely to have reached the viable epidermis.<br />

Although extremely fine particles may penetrate the loosely packed<br />

superficial stratum disjunctum, they would not be expected to migrate<br />

further into the deeper, tightly packed cells of the stratum compactum.<br />

Literature on human skin penetration by particulates indicates that follicles<br />

will be penetrated up to a size of 50 mm, and that the SC by an optimal diameter<br />

of 5 mm, migrating to a depth of ca. 10 tape strips, as observed by<br />

optical methods (41–44). In the studies cited above, the intent was to promote<br />

depth penetration of a dermatological preparation applied, achieved<br />

by massage of the treated skin area. Particles lodged in hair follicles would<br />

not appear as artifacts since tape stripping of skin only removes SC without<br />

associated hair follicles (22).<br />

Toxicological Considerations and Pharmacological Evidence<br />

for the Effects of Exogenous Copper<br />

Toxicity of the ionized form of this essential trace element is kept in check<br />

by an efficient homeostatic mechanism involving metallothionein and ceruloplasmin<br />

(45). Data available indicate that copper may have noxious effects<br />

only following chronic oral (or parenteral) exposure to high amounts of the<br />

metallo-element, particularly upon chronic oral ingestion with food (e.g.,<br />

water) that exposes the human organism to more than 5 mg/kg of copper<br />

per day (46).<br />

Sorenson (47) investigated the acute toxicity of AI copper complexes,<br />

partly listed in Table 2. Comparison with the respective chelating agent<br />

demonstrated that the copper complexes were less toxic and damaging to<br />

the organism than the parent drug.<br />

In a phase I trial to investigate the cutaneous absorption of copper in<br />

the skin, the metal was applied as copper (I) oxide and in its elemental form<br />

as an ointment (paraffin and Vaseline) under gauze. Based on daily application


92 Hosty´nek et al.<br />

Table 2 Acute Toxicity and Therapeutic Indices of Copper Complexes<br />

Copper complex LD50 (sc) (mg/kg)<br />

on the skin of volunteers over a four-week period, the authors concluded that<br />

dermal exposure to copper in concentrations of up to 20% does not present a<br />

toxic risk (48).<br />

The risk of copper toxicity may exist only by administering amounts of<br />

the metallo-element far greater than those that can be actually used for therapeutic<br />

purposes, by transdermal exposure in particular.<br />

CONCLUSIONS<br />

The present observations serve to confirm action of skin exudates (sweat<br />

and sebum) in reacting with copper metal on prolonged contact and in the<br />

presence of air, to form in situ diffusible salts, such as the chloride, pyruvate<br />

or lactate, and lipophilic derivatives (soaps) of likely diffusivity (8). The<br />

role of the skin as a toxicologically important route of exposure to environmental<br />

agents in general and metal compounds in particular is hereby<br />

underscored. Skin is an important port of entry not only to hazardous materials<br />

such as readily oxidized metals that harbor the potential for serious<br />

health effects, but also to essential elements such as copper, whose release<br />

and diffusivity through skin contact has the potential for beneficial and<br />

therapeutic action in the inflamed organism.<br />

These findings may contribute to the acceptance of the long-held<br />

belief in the AI effects of copper metal in direct contact with the skin in<br />

particular, but also may promote the concept of external AI therapy as<br />

alternative to systemic dosing, such as intravenous, intra-articular or intraperitoneal.<br />

Critical for AI activity may be the supplementation of endogenous<br />

copper with exogenous sources, irrespective of the agent’s nature: elemental,<br />

salt, complex, or as a covalent derivative, as endogenous formation of<br />

the chelate of maximum biological activity will occur by homeostasis. The<br />

mechanism of copper complexes acting as AI agents is not known in all its<br />

details, yet evidence so far points to the formation of a unique, as yet undefined<br />

metabolite that might be responsible for the suggested clinical AI effect.<br />

TI<br />

CFE PA<br />

Cupric acetate 350 70<br />

Cupric anthranilate 750 106 50 750<br />

Cupric aspirinate 760 100 150 760<br />

Abbreviations: TI, therapeutic index; CFE, carrageenan foot edema; PA, polyarthritis;<br />

sc, subcutaneous.<br />

Source: From Ref. 47.


Diffusion of Copper Through Human Skin In Vivo 93<br />

GLOSSARY<br />

Absorption Uptake into the organism.<br />

Coinage metals Copper, silver, and gold, with negative oxidation<br />

potential relative to the hydrogen (standard)<br />

oxidation potential defined as 0.00 V. Their<br />

characteristic electronic arrangements permit<br />

facile oxidation. Possible oxidation states are I, II,<br />

and III. They are often stabilized through<br />

formation of characteristically covalent<br />

complexes.<br />

Corrosion Electrochemical process involving redox reactions<br />

in the presence of electrolytes.<br />

Electromotive series Arrangement of metals by decreasing order in<br />

their ability to oxidize.<br />

Ionization potential Energy (electron volts, ev) required to remove an<br />

electron from its atomic orbit, with the value for<br />

the standard hydrogen electrode set at 0.00 ev as<br />

an arbitrarily selected standard reference.<br />

Metallic elements Characterized by the ability to form cations<br />

(positive ions), by luster, malleability,<br />

conductivity (thermal and electrical) and the<br />

formation of cations (positive ions).<br />

Noble metals Descriptive term used to characterize<br />

electrochemically inert metals, mostly copper,<br />

silver, gold, palladium, and platinum.<br />

Oxidation Process of electron removal from an atom or ion<br />

(e.g., the increase in the proportion of oxygen in a<br />

compound).<br />

Oxidation potential Electrical driving force toward electron loss,<br />

expressed as a potential value (in electron<br />

volts, ev).<br />

Penetration Passive diffusion process of a solute through skin.<br />

Permeation Diffusion through one or several layers.<br />

Reduction Process of electron gain by an atom or an ion<br />

(e.g., the increase in the proportion of hydrogen in<br />

a compound).<br />

ABBREVIATIONS<br />

AI anti-inflammatory<br />

AUC area under the curve<br />

ev electron volt<br />

SC stratum corneum


94 Hosty´nek et al.<br />

sc subcutaneous<br />

ICP-MS inductively coupled plasma–mass spectroscopy<br />

REFERENCES<br />

1. Lidén C, Carter S. Nickel release from coins. Contact Dermatitis 2001; 44:<br />

160–165.<br />

2. Wass U, Wahlberg JE. Chromated steel and contact allergy. Contact Dermatitis<br />

1991; 24:114–118.<br />

3. Black H. Dermatitis from nickel and copper in coins. Contact Dermatitis<br />

Newslett 1972; 12:326–327.<br />

4. Karlberg AT, Boman A, Wahlberg JE. Copper—a rare sensitizer. Contact<br />

Dermatitis 1983; 9:134–139.<br />

5. Saltzer EI, Wilson JW. Allergic contact dermatitis due to copper. Arch Dermatol<br />

1968; 98:375–376.<br />

6. Fat L, Gyorffy L. Occupational dermatitis due to copper exposure. Stockholm:<br />

OEESC, 2005:51–52.<br />

7. Hostynek JJ. Factors determining percutaneous metal absorption. Food Chem<br />

Toxicol 2003; 41:327–333.<br />

8. Hostynek JJ. Flux of nickel salts versus a nickel soap across human skin. Exog<br />

Dermatol 2003; 2:216–222.<br />

9. Walker WR, Griffin BJ. The solubility of copper in human sweat. Search 1976;<br />

7:100–101.<br />

10. Whitehouse MW, et al. Alternatives to aspirin, derived from biological sources.<br />

Agents Actions 1977; 7(Suppl 1):43–57.<br />

11. Odintsova NA. Permeability of human skin to potassium and copper ions and<br />

their ultrastructural localization. Chem Abstr 1978; 89:360.<br />

12. Bentur Y, et al. An unusual skin exposure to copper; clinical and pharmacokinetic<br />

evaluation. J Toxicol Clin Toxicol 1988; 26:371.<br />

13. Dollwet HH, Schmidt SP, Seeman RE. Anti-inflammatory properties of<br />

copper implants in the rat paw edema: a preliminary study. Agents Actions 1981;<br />

11:746–749.<br />

14. Bommannan D, Potts RO, Guy RH. Examination of stratum corneum<br />

barrier function in vivo by infrared spectroscopy. J Invest Dermatol 1990; 95:<br />

403–408.<br />

15. Cullander C, et al. A quantitative minimally invasive assay for the detection of<br />

metals in the stratum corneum. J Pharm Biomed Anal 2000; 22:265–279.<br />

16. Higo N, et al. Validation of reflectance infrared spectroscopy as a quantitative<br />

method to measure percutaneous absorption in vivo. Pharm Res 1993; 10:<br />

1500–1506.<br />

17. Loeffler H, Dreher F, Maibach HI. Stratum corneum adhesive tape stripping:<br />

influence of anatomical site, application pressure, duration and removal. Br J<br />

Dermatol 2004; 151:746–752.<br />

18. Rougier A, Lotte C, Maibach HI. In vivo percutaneous penetration of some<br />

organic compounds related to anatomic site in humans: predictive assessment<br />

by the stripping method. J Pharm Sci 1987; 76:451–454.


Diffusion of Copper Through Human Skin In Vivo 95<br />

19. van der Molen RG, et al. Tape stripping of human stratum corneum yields cell<br />

layers that originate from various depths because of furrows in the skin. Arch<br />

Dermatol Res 1997; 289:514–518.<br />

20. Parsons ML, Major S, Forster AR. Trace element determination by atomic spectroscopic<br />

methods-state of the art. Appl Spectrosc 1983; 37:411–418.<br />

21. Schwindt DA, Wilhelm KP, Maibach HI. Water diffusion characteristics of<br />

human stratum corneum at different anatomical sites in vivo. J Invest Dermatol<br />

1998; 111:385–389.<br />

22. Finlay A, Marks R. Determination of corticosteroid concentration profiles in the<br />

stratum corneum using skin surface biopsy technique. Br J Dermatol 1982;<br />

107:33–38.<br />

23. Hostynek JJ, et al. Human stratum corneum penetration by nickel: in vivo study<br />

of depth distribution after occlusive application of the metal as powder. Acta<br />

Dermato-Venereol (Suppl) 2001; 212:5–10.<br />

24. Molochia MM, Portnoy B. Neutron activation analysis of trace elements in skin<br />

IV. Regional variations in copper, manganese and zinc in normal skin. Br J Dermatol<br />

1970; 82:254–255.<br />

25. Meyer BJ, et al. Distribution of copper in the skin. South Afr Med J 1972;<br />

46:907–912.<br />

26. Watt F, et al. Analysis of copper and lead in hair using the nuclear microscope;<br />

results from normal subjects, and patients with Wilson’s disease and lead poisoning.<br />

Analyst 1995; 120:789–791.<br />

27. Petering HG, Yaeger DW, Witherup SO. Trace metal content of hair. Arch<br />

Environ Health 1971; 23:202–207.<br />

28. Flint GN. A metallurgical approach to metal contact dermatitis. Contact Dermatitis<br />

1998; 39:213–221.<br />

29. Hostynek JJ, Reagan KE, Maibach HI. Oxidative properties of skin exudates—<br />

a determinant for nickel diffusion: a review. Exogenous Dermatol 2002;<br />

1:7–17.<br />

30. Öhman H, Vahlquist A. The pH gradient over the stratum corneum differs in<br />

X-linked recessive and autosomal dominant ichthyosis: a clue to the molecular<br />

origin of the ‘‘acid skin mantle.’’ J Invest Dermatol 1998; 111:674–677.<br />

31. Guyton AC. In: Textbook of medical physiology. 8th ed. Philadelphia: W.B.<br />

Saunders Co., 1991:277; 801.<br />

32. Yousef MK, Dill DB. Sweat rate and concentration of chloride in hand and<br />

body sweat in desert walks: male and female. J Appl Physiol 1974; 36:82–85.<br />

33. Lampe MA, et al. Human stratum corneum lipids: characterization and regional<br />

variations. J Lipid Res 1983; 24:120–130.<br />

34. Schurer NY, Elias PM. The biochemistry and function of stratum corneum<br />

lipids. Adv Lipid Res 1991; 24:27–56.<br />

35. Wertz PW, et al. Composition and morphology of epidermal cyst lipids. J Invest<br />

Dermatol 1987; 89:419–425.<br />

36. Willing SK, Gamlen TR. Sweat osmolality values in normal adults. Clin Chem<br />

1987; 33:612–613.<br />

37. Nedorost S, Wagman A. Positive patch-test reactions to gold: patients’ perception<br />

of relevance and the role of titanium dioxide in cosmetics. Dermatitis 2005;<br />

16:67–70.


96 Hosty´nek et al.<br />

38. Yongchao Q, Yiping H, Wanlaui R. Adsorption behavior of noble metal ions<br />

(Au, Ag, Pd) on nanometer-size titanium dioxide with ICP-AES. Anal Sci 2003;<br />

19:1417–1420.<br />

39. Schaefer H, Redelmeier TE. Skin barrier—principles of percutaneous absorption.<br />

Basel: Karger, 1996:162–163.<br />

40. Schaefer H, Redelmeier TE. Skin barrier—principles of percutaneous absorption.<br />

Basel: Karger, 1996:129–152.<br />

41. Illel B. Formulation for transfollicular drug administration: some recent advances.<br />

Crit Rev Therap Drug Carrier Systems 1997; 14:207–219.<br />

42. Illel B, Schaefer H. Transfollicular percutaneous absorption. Acta Dermato-<br />

Venereol 1988; 68:427–430.<br />

43. Rolland A, et al. Site-specific drug delivery to pilosebaceous structures using<br />

polymeric microspheres. J Pharm Res 1993; 10:1738.<br />

44. Schaefer H, et al. Follicular penetration. In: Scott RC, Guy RH, Hadgraft J, eds.<br />

Prediction of percutaneous penetration. London: IBC, 1990:163–166.<br />

45. Hellman NE, Gitlin JD. Ceruloplasmin metabolism and function. Annu Rev<br />

Nutr 2002; 22:439–458.<br />

46. Aggett PJ, Fairweather-Tait S. Adaptation to high and low copper intakes: its<br />

relevance to estimate safe and adequate daily dietary intakes. Am J Clin Nutr<br />

1998; 67:1061S–1063S.<br />

47. Sorenson JRJ. Copper chelates as possible active forms of the antiarthritic<br />

agents. J Med Chem 1976; 19:135–148.<br />

48. Gorter RW, Butorac M, Cobian EP. Examination of the cutaneous absorption<br />

of copper after the use of copper-containing ointments. Am J Therap 2004;<br />

11:453–458.


6<br />

Irritation Potential of<br />

Copper Compounds<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

INTRODUCTION<br />

Following clarification of frequently encountered terms in dermatotoxicology<br />

we present a synopsis of irritant reactions of the skin to copper and its<br />

compounds: types of untoward skin reactions in general, aspects of human<br />

exposure to copper, and a description of predictive and diagnostic methods<br />

to assess irritancy through bioengineering methods, in vivo and in vitro, in<br />

humans and animals. The review discusses case studies, followed by critical<br />

examination of literature reports, with consideration given to a number of<br />

confounding factors in diagnosis. To a limited extent, the review also discusses<br />

immunotoxicity, copper pharmacology, and therapeutic benefits of exposure.<br />

EXPOSURE TO COPPER<br />

Natural sources of human copper exposure due to volcanic exhalations,<br />

weathering of mineral deposits, and runoff are a minor factor. The major<br />

release of copper stems from anthropogenic emissions, stemming from<br />

This chapter, in part, was reprinted from Hosty´nek JJ, Maibach HI. Review: skin irritation<br />

potential of copper compounds. Tox Mech Meth 2004; 14:205–213, with permission of Informa<br />

Healthcare.<br />

97


98 Hosty´nek and Maibach<br />

major industrial activities such as mining, smelting and refining, agricultural<br />

and industrial use of copper pesticides and preservatives, the burning of coal,<br />

waste incineration, and widespread consumer applications of copper (e.g.,<br />

brake-pad releases). Thus, occupational exposure is preponderant and<br />

mainly through inhalation. Concentrations of copper in the occupational<br />

setting are rarely reported, as the focus there lies mainly on other elements of<br />

greater toxicity. It is thus difficult to relate health effects from those environments<br />

specifically to copper. Most countries limit copper-containing dust to a<br />

range of 0.5–1.0 mg Cu/m 3 , and copper in fumes to 0.1 and 0.2 mg Cu/m 3 (1).<br />

For purposes of occupational hazards in the United States, a limited<br />

number of compounds are recognized as hazardous on cutaneous exposure,<br />

and are identified as such by a ‘‘skin’’ notation in the listing of hazardous chemicals<br />

by the American Conference of Governmental and Industrial Hygienists<br />

(ACGIH) in their listing of threshold limit values (TLVs). The purpose of such<br />

labeling is to raise attention to the fact that cutaneous absorption can present a<br />

significant risk of systemic toxicity. The criterion most frequently used for a<br />

‘‘skin’’ listing is acute animal toxicity from skin absorption, i.e., a dermal<br />

LD50 below 1000 mg/kg. This may be an indication of either rapid skin penetration<br />

or extreme toxicity, or both. The TLV values applicable to copper as<br />

fume are 0.2 and 1 mg/m 3 as respirable dust or mist, for purposes of irritation,<br />

gastrointestinal exposure, or metal fume fever (inhalation). In the 2001 edition<br />

of TLV guidelines, copper does not rate a skin notation (2).<br />

Exposure of the general population to this essential trace element is of<br />

minor importance, limited to normal dietary intake of copper naturally occurring<br />

in plants and meat, and the metal released into drinking water conveyed<br />

through copper tubing. Systemic exposure to copper occurs through its slow<br />

release from dental materials and intrauterine devices (IUDs). Topical<br />

exposure comes from the release of copper in alloys used in jewelry, as it is<br />

measurably released in contact with skin exudates.<br />

SOLUBILIZATION OF COPPER METAL<br />

Dermal<br />

Inflammatory skin reactions of different types are due mostly to exogenous<br />

factors, primarily chemical agents impacting the skin. To exert an irritant or<br />

inflammatory action they must penetrate the stratum corneum (SC), a layer<br />

of inert keratinized cells, before reaching the viable layers of the epidermis<br />

and dermis. Among the irritant chemicals are acids, bases, organic solvents,<br />

salts, soaps and detergents, and pharmacological agents.<br />

Copper and other elements in their metallic state have no effect on the<br />

skin. They become potential irritants or allergens only when they are corroded<br />

(oxidized) and thus become soluble through the action of exudates<br />

encountered on the skin surface, or in a relatively corrosive physiological<br />

environment such as the oral cavity or the uterus.


Irritation Potential of Copper Compounds 99<br />

By the action of salts and acids present in sweat and sebum on the skin,<br />

most base metals are converted to the hydrophilic (ionized salts) or lipophilic<br />

(soap) form, respectively. Sweat composition fluctuates considerably in<br />

function of the rate of sweat secretion (3). Besides sodium and chloride, other<br />

significant corrosive components of sweat are potassium, urea, lactate and<br />

pyruvate, amino acids, proteins, and acidic lipids. The formation of free acids<br />

in the SC and on the skin surface is the result of hydrolysis of those acidic lipids<br />

by lipolytic enzymes occurring in the sebaceous ducts and on the skin surface,<br />

and of bacterial decomposition (4,5). It is the oxidizing (corroding) action of<br />

such acids that results in the formation of soaps with copper (and metals in<br />

general) upon intimate and prolonged contact with articles of daily use, which<br />

potentially result in skin irritation or allergic reactions once they reach the<br />

viable structures of the skin, since these relatively lipophilic compounds penetrate<br />

the SC with relative ease as compared to ionized salts (electrolytes) (6).<br />

Metallic objects used in jewelry or drug-like devices (dental materials,<br />

orthopedic implants) as a rule are not made of copper alone, but the metal<br />

is incorporated in alloys that have corrosion (oxidation) characteristics quite<br />

different from those of the constituent metals. An exception is the wire used in<br />

IUDs, presumably made of high-grade copper only. The characteristics of<br />

alloys are determined by electrochemical characteristics of the elements in<br />

contact with each other; oxidation and formation of potentially allergenic ions<br />

will vary in function of alloy composition. The electrochemical potential (galvanic<br />

effect) between diverse elements in close proximity provide the driving<br />

force for such reactions resulting in enhanced corrosion (7). The more electropositive<br />

(baser) the element (e.g., nickel), the more stable it is in the ionized<br />

state, and will transfer electrons to the more electronegative, nobler metal<br />

(e.g., copper). The actual concentration of a metal in the alloy is thereby only<br />

of secondary importance. Ultimate biological activity of the alloy is determined<br />

by the rate at which metal ions are released, i.e., whether they reach<br />

a concentration sufficient to provoke a reaction in the adjacent tissues.<br />

Release of copper in synthetic sweat related to chloride ion concentration<br />

was determined by Boman et al. After 24 hours, copper dissolved from<br />

coins and copper thread in the range of 80–100 mg/mL sweat, with an inverse<br />

relationship between the concentration of copper and chloride ion (8).<br />

Lidén et al. determined the release of copper from gold-containing<br />

jewelry in artificial sweat. Amounts released over one week ranged between<br />

0.11 and 0.66 mg/cm 2 , depending on alloy composition (9).<br />

Systemic<br />

Corrosion and solution of copper in the physiological environment may be<br />

considered as equivalent to systemic dosing. Human plasma is an aggressive<br />

physiological medium for dissolving metals. Corrosion of the foreign object<br />

in this microenvironment releases components into the organism, some of<br />

which can then act as irritants or allergens. Levels of free ionic copper,


100 Hosty´nek and Maibach<br />

however, a relatively toxic metal, are moderated to the minimum levels<br />

required for physiological needs, 10 19 mol/L estimated in blood plasma,<br />

through binding to ceruloplasmin and metallothionein. The dynamic<br />

equilibrium between ceruloplasmin and metallothionein prevents toxic<br />

accumulation or deficiency of copper in mammals.<br />

Dental Alloys<br />

Amounts of copper released from commonly used dental casting alloys,<br />

measured in cell culture over 10 months, was 0.15 mg/cm 2 /day (10). Cytotoxicity<br />

of metals thus released from dental alloys could be considered a correlate<br />

of irritancy. Accordingly, Wataha et al. (11–14) investigated in vitro corrosion<br />

rates of dental casting alloys in various culture media to obtain a measure<br />

of biological risk to oral tissues in a number of investigations. Grimsdottir<br />

et al. (15) also studied the cytotoxic effect of orthodontic appliances in an<br />

attempt to obtain a measure of tissue irritation caused by corrosion. Such data<br />

do not allow derivation of an objective measure of copper irritancy; however,<br />

release of metal ion in a simulated environment is highly dependent on presence<br />

and concentration, and thus the (galvanic) interaction with other metals,<br />

resulting in variable and unpredictable concentrations/cytotoxicity of individual<br />

metal ions, e.g., copper, as most of the metal is protein bound.<br />

Intrauterine Devices<br />

Increases in systemic copper via parenteral entry from a contraceptive IUD can<br />

lead to adverse effects reported: systemic nonspecific contact dermatitis and<br />

immediate immunologic contact urticaria, even though the amounts liberated<br />

from such a device are relatively low. Copper levels determined in intrauterine<br />

fluids from women who had used the T-380 A device were 11.4 4.7 mg/mL<br />

after six months; 11.54 7.0 mg/mL after one year and 6.24 1.5 mg/mL after<br />

three years. Overall, concentrations over the entire period surveyed ranged from<br />

3.9 to 19.1 mg/mL (16). It is inferred that the toxic effect of copper ions thus<br />

released in the uterus (present in the form of complexes with proteins) are<br />

responsible for cutaneous eruptions, although most of the reported cases appear<br />

to belong to the category of nonimmunologic systemic contact dermatitis (16).<br />

These investigations on the release of copper ion from alloys in the<br />

physiological environment in vivo and in vitro and the potential biological<br />

effects from exposure help to explain the cases of systemic irritative response<br />

to IUDs and dental materials (17,18). On close examination, the immunologic<br />

relevance of many of those reports is unclear.<br />

INCIDENCE AND EPIDEMIOLOGY OF IRRITATION<br />

DUE TO COPPER<br />

Incidence of irritant contact dermatitis (ICD) is difficult to establish, as often<br />

patients do not consult a doctor. ICD, especially of the hands, is reported to


Irritation Potential of Copper Compounds 101<br />

be more common in women than men, possibly due to greater exposure<br />

to irritants in ‘‘wet work’’ in the household; also, women are more likely<br />

to consult with a doctor than men are. Studies in twins indicate that heredity<br />

is a factor in susceptibility to irritants, but variability is too great for generalizations.<br />

Atopic dermatitis seems to bring greater risk for ICD (18,19).<br />

Based on 5839 <strong>dermatology</strong> patients patch tested by the North American<br />

Contact Dermatitis Group, in which the role of occupational exposure to allergens<br />

and irritants was evaluated, 19% were found to be occupationally related.<br />

Of those, 60% were of allergic and 32% of irritant origin. The hands were the predominant<br />

part of the body affected, 80% of thosedue to exposure to irritants (20).<br />

For copper specifically, the aspects of epidemiology, prevalence, or<br />

population studies cannot be addressed since, in contrast to other metals such<br />

as nickel or chromium, reports of untoward reactions, systemic as well as<br />

cutaneous, are extremely rare. The two geographical areas with the most complete<br />

databases are the National Office for Occupational Health (Helsinki,<br />

Finland) and the State of California. The figures emanating from these<br />

sources are skewed in that they probably represent a small portion of the<br />

actual frequency of disease due to inherent weaknesses in reporting systems.<br />

PHARMACOLOGY OF COPPER<br />

Beneficial as well as adverse health effects due to copper, an essential trace<br />

element, are well characterized. Two pathological conditions stand out due<br />

to their chronicity. Menkes’ syndrome is remarkable in that there is no<br />

known cure and homocygots usually die early in life.<br />

Wilson’s disease (WD) is an inherited copper metabolism disorder,<br />

impairing biliary tract copper excretion that leads to excessive levels of the<br />

element in tissue, particularly in the liver if left untreated. Left untreated,<br />

such copper accumulation leads to hemolytic anemia, which over the years<br />

can result in progressive hepatic failure and ultimately death (21). The characteristic,<br />

brown ‘‘Fleisher rings’’ that develop in the eyes of Wilson’s disease<br />

patients are caused by the deposition of metallic copper. However, WD<br />

is very much treatable, if not cured, by penicillamine therapy and dietary<br />

control. WD patients lead seemingly normal lives as long as they are on<br />

medication and restrict copper intake.<br />

Deficiency of copper is associated with characteristic integumentary<br />

and skeletal abnormalities, defects in growth and development, and<br />

abnormalities in sensory perception (22). Copper status of the organism is<br />

reflected in ceruloplasmin levels. Plasma levels below 125 mg/dL are generally<br />

considered as indicative of copper deficiency (23).<br />

Menkes’ Syndrome<br />

Albinism, the striking absence of pigmentation in the skin, hair, and eyes, is<br />

characterized by the absence of the copper enzyme tyrosinase, which


102 Hosty´nek and Maibach<br />

converts tyrosine to melanin in the melanocyte (24). Menkes’ kinky hair<br />

syndrome, a hereditary defect in intestinal copper absorption that causes<br />

retardation in growth, focal cerebral and cerebellar degeneration, and hair<br />

to be abnormally sparse and brittle, becomes manifest in early infancy (25).<br />

Afflicted infants have low levels of copper and ceruloplasmin, dying usually<br />

within the first year of life. Although copper absorption and reabsorption<br />

are impaired, tissue copper levels of many epithelial tissues, including the<br />

skin fibroblasts, are elevated, and an increased production of metallothionein,<br />

the cysteine-rich protein that binds copper in cells, appears to be the<br />

cause of such accumulation. The biochemical defect underlying Menkes’<br />

syndrome however is largely unknown.<br />

Copper as Antimicrobial<br />

Copper itself proved highly antimicrobial in plumbing and in lab tests with<br />

several bacterial strains and some viruses.<br />

A chlorophyllin copper complex, derived from chlorophyll by replacing<br />

the chelated magnesium with copper, has anti-inflammatory and antimicrobial<br />

properties, as well as a marked stimulating effect on epithelial cell<br />

growth rates and cell regeneration. First established in tissue culture studies,<br />

these findings were confirmed clinically through wound healing and deodorizing<br />

characteristics observed in animal and man (26). Administered orally,<br />

chlorophyllin copper complex is classified as a safe and effective internal<br />

deodorant by the U.S. Food and Drug Administration (27).<br />

Transdermal Anti-inflammatory Action of Copper<br />

Exogenous copper has demonstrable anti-inflammatory effect, as several<br />

copper complexes like Cupralene, Dicuprene, Alcuprin, or Permalon are<br />

successfully employed in treating human arthritis (28–31). The potential<br />

for copper’s activity as an anti-inflammatory agent by transdermal delivery<br />

is subject to controversy, however. This is because scientific studies designed<br />

to demonstrate therapeutic benefits for arthritic conditions through dermal<br />

contact with metallic copper so far have been inadequate. Quantitative data<br />

for percutaneous penetration of copper’s putative oxidation products, which<br />

may be generated in contact of the metal with skin in humans, are still<br />

outstanding. One missing, important factor for a convincing case of such<br />

potential benefits is the deficiency in systematic and adequate scientific<br />

research into the penetration of copper through human skin in any of its<br />

forms, as polar mineral salts or as the more lipophilic complexes, and thus<br />

a lack of solid scientific data documenting the therapeutic value of transdermal<br />

copper delivery. It can be safely assumed that endogenous copper has<br />

natural anti-inflammatory activity, and that such activity may also be reinforced<br />

by exogenous copper. In a review of anti-inflammatory activity of<br />

exogenous copper, Milanino et al. (32) concluded that copper, indeed, is active


Irritation Potential of Copper Compounds 103<br />

as an acute anti-inflammatory agent irrespective of chemical form, including<br />

inorganic copper salts. That there is a direct connection between copper and<br />

rheumatoid arthritis is supported by the fact that low molecular weight<br />

copper concentrations in plasma and synovial fluids increase in response to<br />

the disease, and when such increases are induced further by administration<br />

of exogenous copper, they are observed to have a definite anti-inflammatory<br />

effect in both laboratory animals and humans.<br />

COPPER IRRITANCY IN SKIN AND MUCOSA<br />

In Vivo Assays<br />

Kinetics and specificity of nickel hypersensitivity were assessed by Siller and<br />

Seymour in mice presensitized with nickel sulfate and challenged with Cu (II)<br />

sulfate, chromic chloride, cobaltous chloride, nickel chloride, and nickel sulfate.<br />

The challenge concentration for the metal salts was 0.0152 M, and for<br />

Cu (II) sulfate, 0.003 M. A reaction occurred at 24 hours, resolving at<br />

48 hours, consistent with an irritant reaction. Cu (II) sulfate was found to<br />

be ‘‘profoundly more irritant than the other metals’’ (specific numbers not<br />

given) (33).<br />

The biocompatibility and metal release were investigated in vivo<br />

through implantation of representative specimen alloys in rats, and in vitro<br />

in a battery of cell culture tests (7). In addition, combinations of dissimilar<br />

alloys were investigated in relation to possible enhanced corrosion by galvanic<br />

effects. Implantation and cytotoxicity tests on epithelial cells, macrophages,<br />

and erythrocytes were performed, and the results compared. The severity of<br />

tissue response in implantation tests corresponded to the nobleness of the casting<br />

alloys joined to amalgam. The most severe reaction occurred in the tissue<br />

in proximity of the LG-1 alloy, probably due to its high copper content. Similar<br />

results were obtained in the in vitro macrophage test. All of the alloys<br />

except the high-gold alloy (LM-Hard) had a toxic effect on epithelial cells.<br />

The combination of the casting alloys with amalgam diminished such toxicity.<br />

For the study, limit ratios of the metals used in the alloy were evaluated<br />

in order to test the biological significance of the galvanic currents with<br />

respect to these materials.<br />

Implantation Test<br />

Twenty-eight Wistar rats, weighing 250–300 g, were implanted subcutaneously<br />

with the various alloy combinations for time periods of 7, 30, and<br />

60 days (Table 1). Two rats in each group were allocated for each alloy combination.<br />

Polystyrene implants serving as controls were left in 10 rats for the<br />

same observation times. After the allotted time, the animals were sacrificed<br />

with ether. The specimens including the surrounding tissues, submandibular<br />

glands, liver, kidney, spleen, and part of the spinal cord were examined.


104 Hosty´nek and Maibach<br />

Table 1 The Compositions of the Examined Alloys (wt%)<br />

Alloy Au Pt Pd Ag Cu Sn Zn<br />

LM-hard 76.9 1 1.4 9.6 11.1<br />

LG-1 a<br />

50.0 9.0 4.0 34.0 2.9<br />

Micro 5.7 1.0 25.0 67.2 1.0<br />

Midi 44.9 0.1 3.0 39.0 12 1.0<br />

ANA 68 67.6 5 26.2 0.26<br />

Revalloy 69.6 2.8 26.9 0.96<br />

a Experimental alloy.<br />

The connective tissue reactions to the implants were diagnosed as mild,<br />

moderate, or severe on the basis of the degree of infiltrate, vascularity,<br />

and fibrosis. In addition, special attention was paid to estimation of the<br />

giant cells and foreign bodies in the tissues removed. Whenever foreign<br />

bodies were histologically recognized, Energy dispersion X-ray (EDAX)<br />

analysis was performed to reveal their constituents.<br />

Polystyrene control: The response to polystyrene was an uncomplicated<br />

repair of the surgical wound. Collagen fibers were present at day 7,<br />

and a thin compact collagenous capsule enclosed the implant by day 30, followed<br />

by an acellular capsule detectable by day 60.<br />

LM-Hard gold alloy: At day 7, the tissue surrounding the implant<br />

showed a moderate inflammatory cell infiltration and proliferating fibroblasts.<br />

Giant cells and a well-defined capsule were detectable at day 30.<br />

The capsule matured to a dense connective tissue membrane by day 60. Foreign<br />

bodies were still detectable around the implants after 30 and 60 days.<br />

EDAX showed the presence of Au, Cu, and Fe in these bodies.<br />

LM-Hard/ANA 68 combination: The inflammatory response around<br />

the implant was extensive by day 7. Granulation tissue formation was<br />

delayed, and characterized by numerous macrophages and extensive capillary<br />

proliferation. Foreign body aggregates found around the implant were<br />

verified by EDAX to consist of Au, Ag, Hg, Cu, Sn, and Zn. A subacute<br />

inflammation with prominent vascularity still persisted at day 30. Foreign<br />

bodies were shown to contain Cu, Hg, Fe, Sn, and Zn. At day 60, the inflammation<br />

had resolved into the mild stage, and a collagenous capsule<br />

surrounded the implant.<br />

Micro/ANA 68 combination: The initial reaction of the tissue against<br />

the Micro/ANA 68 combination presented heavy inflammation due to granulocytes,<br />

plasma cells, and macrophages. The reaction subsided by day 30,<br />

but the vascularity still remained prominent. By day 60, the tissue outside<br />

the fibrous capsule contained a few accumulations of lymphocytes. EDAX<br />

analysis disclosed the presence of Au, Ag, Cu, Fe, Hg, Sn, and Zn in the<br />

foreign bodies adjacent to the implants.


Irritation Potential of Copper Compounds 105<br />

Midi/ANA 689 combination: After the seven-day observation period,<br />

the Midi/ANA 68 implant had induced a strong cellular reaction with pronounced<br />

vascularity. Macrophages were abundant. After 30 days, there was<br />

a fibrous inflammatory region adjacent to the implant, contiguous with a<br />

zone of granulation tissue composed mainly of fibroblasts, mononuclear<br />

cells, and small blood vessels. Foreign bodies around the implant contained<br />

Ag, Au, Cu, Hg, Pd, and Sn. On day 60, a capsule with well-oriented collagen<br />

fibers existed, with only a few inflammatory cells present.<br />

LG-1/ANA 68 combination: By day 60 there still was a subacute inflammatory<br />

infiltration with high cellularity, plasma cells, and lymphocytes in<br />

predominance. Giant cells, macrophages, and occasional granulocytes were<br />

also detected. Some collagen was apparent, but it was poorly orientated.<br />

At this stage, foreign bodies were seen containing Ag, Au, Cu, Hg, and Sn<br />

in abundance.<br />

Histopathology: None of the biopsies from different parenchymal<br />

organs showed any morphological changes due to implants. Occasionally,<br />

foreign bodies or blackish precipitates were present in liver, kidney, and<br />

spleen. EDAX analysis showed them to contain calcium, chloride, sulfur,<br />

silicon, potassium, and iron in varying proportions. In addition, occasional<br />

copper particles were found in the kidney following the implantation of the<br />

LG-1/ANA 68 combination. Also, a few particles containing Ag were<br />

detected in the spleen of the same animals.<br />

The results obtained in the investigations of alloy combinations<br />

showed that when implanted in living tissue they caused reactions different<br />

in character and intensity, which finally led to the formation of fibrous capsules.<br />

The authors conclude that the differences in the severity of the<br />

responses observed can, in most instances, be explained on the basis of electrochemical<br />

reactions due to the different electrical potentials responsible<br />

for the release of metal ions. In the present study, EDAX analysis showed<br />

the presence of alloy elements in the surrounding tissue in every case. The<br />

composition of the elements was not identical to that of the original alloys,<br />

which indicates in situ corrosion, rather than particles dislodged from the<br />

test alloy during the implantation procedure. The severity and duration of<br />

the inflammatory reaction around the implants fully corresponded to the<br />

nobleness of the alloys, and, surprisingly, not to the suggested electric potential<br />

difference generated between the combined alloys.<br />

Using EDAX analyses, the foreign bodies adjacent to the gold (LM-<br />

Hard) implant were always shown to contain both gold and copper, thus<br />

suggesting that gold may be complexed to copper within cells, or evoking<br />

a copper-like biological response that is also causing localized accumulation<br />

of copper. Whether such a possible gold–copper complex is related to the<br />

adverse effects of gold or is a normal pathway in gold metabolism is not<br />

known. The finding that capsules around the amalgam implants contained<br />

mercury and tin particles are in agreement with previous observations (34).


106 Hosty´nek and Maibach<br />

The extensive reaction to the LG-1/ANA 68 combination is apparently<br />

related to the release of copper from the LG-1 alloy. In vitro studies<br />

on binary Cu–Pd alloys have shown that a preferential dissolution of Cu<br />

is followed by an enrichment of Pd on the alloy surface (35). Amalgam, on<br />

the other hand, corrodes continuously. The high copper content of LG-1<br />

(34%) accounts for a continuous copper release with a slow rate of Pd<br />

enrichment, thus maintaining a persistent inflammation with high cellularity<br />

adjacent to the implant. These findings are consistent with recent reports<br />

dealing with tissue response to Ag–Pd–Cu–Au I alloys and pure copper<br />

implants (36,37).<br />

The abundance of macrophages and copper around the LG-1/ANA 68<br />

implants supports the results of McNamara and Williams (37) who showed<br />

that the pigmented material found in connection with Cu implants was composed<br />

of Cu-containing macrophages. The cells had absorbed large amounts<br />

of copper and remained damaged in the area, attracting more macrophages<br />

to these sites. Cu particles could seldom be found in the liver after implantation<br />

of the LG-1/ANA combination. This is contradictory to the findings of<br />

Yli-Urpo and Parvinen (38), who always found elevated levels of Cu and Hg<br />

in the liver and kidney after implantation of different alloy combinations.<br />

This discrepancy can be explained by the different methods used. The disadvantage<br />

of EDAX analysis is that only the surface of the specimen to a depth<br />

of 2–3 mm can be analyzed.<br />

Agarose Overlay Test<br />

The effects of alloys and their combinations on cultured human epithelial<br />

cells were examined. The cytotoxic effect of the test alloy was evaluated<br />

by measuring the zone of cell lysis around the alloy.<br />

Midi produced the most prominent cytotoxicity, whereas LM-Hard<br />

had no effect. All of the alloy combinations were less cytotoxic than the constituent<br />

alloys when tested separately. The diminishing cytotoxicity was<br />

most prominent with the combination of Midi/ANA 68.<br />

The reaction between the alloy and the culture medium can result in the<br />

leakage of metal ions from the alloy into the culture medium, while the cells<br />

themselves have no detectable effect on the corrosion process. LG-l, Micro,<br />

Midi, and ANA 68 alloys showed a marked cytotoxic activity in the agarose<br />

overlay test. The release of copper could be the major factor responsible for<br />

the observed rapid cytotoxic effect of Midi. The minor degree of cytolysis<br />

caused by LG-1, despite its high copper content, might be due to the preferential<br />

release of the least noble metal, zinc, thus retarding the release of the<br />

more noble constituents, copper included. The equal degree of cytolysis<br />

caused by Micro and ANA 68 (both containing equal amounts of silver) substantiates<br />

the concept of the role of silver as a cytotoxic agent. Surprisingly,<br />

the degree of cytolysis diminished when the casting alloys were combined<br />

with ANA 68. This is probably due to the electrochemical passivation, which


Irritation Potential of Copper Compounds 107<br />

was most pronounced in the Midi/ANA 68 combination, and least in the<br />

LG-l/ANA 68 combination.<br />

Erythrocyte Lysis Assay<br />

When the hemolytic activities of the alloys were tested, Midi, Revalloy, and<br />

LM-Hard were shown to possess a slight hemolytic activity. Microscopy of<br />

the cell pellet did not show any hemagglutination, which otherwise might<br />

cause low hemolysis values.<br />

Incubation of Midi, Revalloy, and LM-Hard with erythrocytes resulted<br />

in a slight degree of hemolysis. The mechanisms by which the metal particles<br />

produce their biological effects are not known in detail. It has been proposed<br />

that interaction between the erythrocyte membrane and the particles would<br />

be the most important factor in hemolysis (29). Additional mechanisms conferring<br />

the hemolytic activity are the chemical nature at the metal surfaces,<br />

particle size, and their surface charge. Since LG-1 did not show any hemolytic<br />

activity, cytotoxicity cannot be attributed to copper release. Further studies<br />

are needed, however, in order to elaborate on the elements and membrane<br />

components involved in the hemolytic mechanisms of dental alloys.<br />

Toxicity Test Using Murine Macrophages<br />

Latex and Revalloy particles are phagocytized faster than the other alloy particles.<br />

In the cultures of macrophages, which had been in contact with LG-1, a<br />

phagocytosis rate of only 25% was detectable, as compared to 80% due to<br />

Revalloy. The number of alloy particles phagocytized per macrophage was<br />

significantly lower than that of the Latex particles. The proportion of nonviable<br />

macrophages after exposure to the alloys, except Microalloy, was slight.<br />

More pronounced cellular damage with pyknosis and vacuolization appeared<br />

after exposure to Microalloy. No difference in toxicity was observed after one<br />

day compared to one-hour exposure to the alloys except for LG-1, which<br />

showed cell damage comparable to that due to Microalloy. A considerable<br />

amount of lactic dehydrogenase (LDH) was released by Microalloy. In contrast,<br />

little LDH release occurred when the cultures were exposed to Revalloy<br />

or latex particles.<br />

Solubility of particulate alloys into the macrophage culture medium: The<br />

concentration of zinc in medium from alloy cultures was higher than in<br />

the controls. The solubility of Zn was most prominent from Midi alloy.<br />

In addition, copper release from LG-1 and Midi alloys was found. No<br />

release of Au, Ag, or Sn was detected in any of the culture media (Table 2).<br />

The corrosion of metal implants in human and mammalian organisms<br />

(due to body fluids) may lead to local reactions in the surrounding tissues.<br />

The tissue response will depend on the corrosive behavior of the metal, the<br />

rate of release of metal ions, and their physiological activity. Since each<br />

element will be released at a different rate from a complex alloy and many


108 Hosty´nek and Maibach<br />

Table 2 Soluble Metals Concentration in Macrophage Culture After One Hour<br />

Particulate alloy Zn SD (mg/mL) Cu SD (mg/mL)<br />

LM-hard 0.62 0.15 0.21 0.10<br />

LG-1 0.71 0.09 0.71 0.30<br />

Micro 0.70 0.14 0.14 0.04<br />

Midi 1.45 0.61 0.52 0.44<br />

Revalloy 0.83 0.21 0.13 0.17<br />

Control 0.56 0.13 0.17 0.10<br />

have a different mechanism of toxicity, it is difficult to establish the biocompatibility<br />

of the alloy using a single test method. A combination of test<br />

methods was used in an attempt to assess the behavior of a variety of complex<br />

dental alloys in different bioenvironments.<br />

Amalgam particles were phagocytized faster than the other alloy particles.<br />

This might be due to the differences in particle size. The present results<br />

indicated that particulate Microalloy was the most toxic of the alloys tested,<br />

whereas particulate Revalloy was well tolerated by the cells. Analyses of the<br />

soluble elements in alloys revealed only relative low concentrations of copper<br />

and zinc. Gold, silver, and tin were not detectable in any of the supernatant<br />

determined from the experiments described. It seems that the alloys are not<br />

sufficiently soluble in tissue culture medium for their effects to be exerted<br />

with extracellular toxic levels. These findings are in agreement with previous<br />

reports where no definite correlations could be found between the solubility<br />

of the particles and toxicity. Thus, it seems more probable that the alloys<br />

exert their toxic effects directly intracellularly after being phagocytized.<br />

Copper has been shown to cause degenerative changes in macrophage<br />

morphology, which could explain the increased LDH values due to LG-1<br />

and Micro, despite only mild and moderate changes in cell morphology.<br />

Comparison of the different tests: Some correlation was seen between<br />

the in vivo implantation test and the in vitro macrophage test. This can<br />

be explained by the central role of macrophages in the manifestation of<br />

inflammation. Furthermore, macrophages are the first cells with which foreign<br />

bodies come into contact in living tissue. The in vitro results obtained<br />

from the agarose overlay test and the erythrocyte lysis test did not correlate<br />

well with the in vivo results. The toxicity established by the agarose overlay<br />

test would indicate the toxicity of soluble silver and copper rather than that<br />

of the alloy in itself. In hemolysis, on the other hand, interaction of the alloy<br />

constituents with biomembranes is one of the likely mechanisms involved in<br />

the toxicity of particulate alloys. Some evidence exists that materials that<br />

have not been phagocytized but that come into contact with the cell surface<br />

can cause macrophage destruction comparable to hemolytic activity. The<br />

interpretation of the results obtained by the different test methods is difficult,<br />

and the dynamic state of cells and their possible metabolic alterations due to


Irritation Potential of Copper Compounds 109<br />

the implants are not fully understood. The behavior of alloys in a biological<br />

environment and the precise effect of each constituent element in different<br />

tests need to be studied more extensively.<br />

The severity of tissue response in the implantation test corresponded<br />

with the nobleness of the alloys combined with ANA 68. The most severe<br />

reaction was seen in the area surrounding LG-l, probably due to its high<br />

copper content. Similar results were obtained in the in vitro macrophage<br />

test. The agarose overlay test showed a somewhat similar zone of lysis for<br />

all the alloys except for LM-Hard. The combination of the alloys with<br />

ANA 68 reduced the lytic zone, which could be accounted for by a surface<br />

passivation of the alloy. LM-Hard, Midi, and Revalloy showed a slight<br />

hemolytic activity. A poor correlation was established between the agarose<br />

overlay, the erythrocyte hemolysis, and implantation tests.<br />

In Vitro Assays<br />

Schmalz et al. evaluated the suitability of a commercially available model<br />

system based on a recombined coculture of human fibroblasts and human<br />

epithelial cells for assessing mucosal irritancy of metals used in dentistry, as<br />

no valid animal or in vitro model exists for this purpose (39). That model<br />

had been introduced for evaluating the time-dependent irritancy of cosmetic<br />

products, where cell viability and prostaglandin E2 (PGE2) release from<br />

the cells were used as markers for the irritative potential of test materials. The<br />

human fibroblast–keratinocyte cocultures were exposed to test specimens<br />

fabricated from copper, zinc, palladium, nickel, tin, cobalt, indium of high<br />

purity (99.98–99.99%), and from a dental ceramic. Cell survival rates decreased<br />

after exposure to copper (14–25%), cobalt (60%), zinc (63%), indium<br />

(85%), nickel (87%), and the nonoxidized/oxidized high noble cast alloy<br />

(87%/90%) compared to untreated control cultures. Dental ceramic, palladium,<br />

and tin did not influence cell viability. In parallel, the PGE2 release<br />

was continuously monitored up to 24 hours using a competitive displacement<br />

enzyme immunoassay. PGE2 release increased most highly in the cultures<br />

exposed to copper (6–25-fold), cobalt (seven-fold), indium (four-fold), and<br />

zinc (two-fold) compared to untreated control cultures. The PGE2 determination<br />

proved to be a nondestructive method for continuous monitoring of cell<br />

reactions in the same culture. The model used seems promising for evaluating<br />

the time-dependent mucosal irritancy of dental cast alloys.<br />

Cell viability of exposed cell cultures was determined by the MTT test<br />

after 24 hours. Survival rates were calculated relative to values obtained in<br />

untreated cultures. For PGE2 release, assay aliquots (100/mL) were taken<br />

from exposed media and the amount of PGE2 released from treated and<br />

untreated cell cultures was quantified against a standard curve of purified<br />

PGE2, using a competitive displacement enzyme immunoassay. Threedimensional<br />

fibroblast–keratinocyte cocultures were exposed to one high<br />

noble dental cast alloy and various metals frequently found in cast alloys.


110 Hosty´nek and Maibach<br />

Identical levels of cell viability were found in untreated control cultures and<br />

in cultures exposed to a dental ceramic, which was used as a negative control<br />

material. Pure copper was the most toxic metal tested. In copper-exposed cultures,<br />

a time-dependent decrease of cell viability at a level of 14% to 25% of<br />

untreated cell cultures was observed. Because of the demonstrated high<br />

toxicity, copper was routinely included as a positive reference material in<br />

all subsequent experiments evaluating the effects of other test materials.<br />

Cobalt and zinc induced a moderate decrease of cell viability to a level of<br />

about 60% of untreated cell cultures. Pure nickel, indium, and oxidized and<br />

nonoxidized specimens of the high noble cast alloy, were weakly toxic. Similar<br />

to the dental ceramic, no cytotoxicity was observed after exposure of the<br />

cocultures to palladium and tin specimens. Survival rates after exposure to<br />

copper, zinc, indium, cobalt, nickel, and the high noble alloy (oxidized and<br />

nonoxidized) were significantly different from those of untreated control<br />

cultures. Total amounts of PGE2 released from cell cultures exposed to test<br />

materials and from untreated control cultures steadily increased during the<br />

exposure period. The spontaneous PGE2 release from untreated tissues was<br />

identical with values obtained from cultures exposed to specimens of the<br />

dental ceramic and nontoxic metals. The amounts of PGE2 released after<br />

exposure to copper were about 10-fold higher than those released from<br />

untreated cultures after a 24-hour exposure. After 30-minute exposure to copper<br />

specimens, significantly higher PGE2 levels were already found compared<br />

to untreated controls. In contrast, no differences were found between the<br />

PGE2 levels measured in media of untreated tissues and tissues treated with<br />

all other test materials. In repeated experiments, the amounts of PGE2<br />

released from cultures exposed to copper varied, being 6–25-fold higher than<br />

those released spontaneously. Indium and cobalt in contrast produced<br />

increases that were considerably lower than those elicited by copper in the<br />

same experiments (4–7-fold). The induction of an increased PGE2 release<br />

from human fibroblast–keratinocyte cocultures was inversely related to cell<br />

viability measurements after exposure to copper. The dramatic effect of<br />

copper on cell viability is in accordance with data from other in vitro and in<br />

vivo studies (40,41). This is due to the oxidative potential of pure copper<br />

and the toxicity of copper ions in vitro (42,43). As a consequence of copper<br />

toxicity, the cell viability was reduced to about 15% to 25% of untreated control<br />

cultures. The increases of PGE1 levels by factors of 2 (zinc) to 25 (copper)<br />

are among the highest observed in vitro so far (44,45). The model system based<br />

on a recombined coculture of human fibroblasts and human epithelial cells<br />

seems promising for evaluation of the mucosal irritative potential of dental<br />

materials; however, further studies, particularly on interexperimental variations,<br />

are needed before it can be established as a routine test model candidate.<br />

Cell viability as measure of cytotoxic potential in HaCaT cells<br />

(a spontaneously immortalized human kertinocyte line) and, indirectly, of<br />

irritancy in vivo, was determined on human keratinocytes in vitro by Brosin


Irritation Potential of Copper Compounds 111<br />

et al. (46) for five metal salts. The endpoint used to assess cellular viability<br />

was metabolism of the tetrazolium salt XTT [2,3-bis (2-methoxy-4-nitro-<br />

5-sulfophenyl)-5-(phenylamino carbonyl)-2H-tetrazolium hydroxide]. The<br />

metal salts showed the following rank order in cytotoxicity at an exposure<br />

time of 24 hours: potassium bichromate > Cu (II) sulfate > cobalt chloride<br />

and palladium chloride > nickel sulfate. The authors found an excellent correlation<br />

to the rank order of the metals’ known irritative potency, as it was<br />

determined in vivo for purposes of contact allergy screening by the ICDRG,<br />

but recognized that such a test hardly applies to the complex pathomechanism<br />

of skin irritation. As such, the presented XTT assay on HaCaT cells would be<br />

well-suited for an initial screening of substances to establish a relative order of<br />

irritancy as part of a battery of tests targeting different aspects of skin irritation.<br />

This could be subsequently followed by irritation tests in humans.<br />

CONCLUSIONS<br />

Data on the dermal irritation by copper and its compounds is scant, and<br />

its irritancy has not determined, e.g., in terms of an irritant dose ID50.<br />

Irritancy of copper can only be comparatively characterized in relation<br />

to other metal salts. A rank order for the irritancy of metal compounds<br />

can be inferred from the patch test concentrations recommended as nonirritating<br />

for the purpose of cutaneous allergy testing: potassium dichromate<br />

0.5% in petrolatum; copper sulfate, cobalt chloride, and palladium chloride<br />

ex equo: 1% in aqueous solution; and nickel sulfate: 5% in petrolatum.<br />

With the exception of its mineral salts, copper (II) compounds (complexes,<br />

soaps) exhibit low irritancy and several have been adapted as therapeutics<br />

for epicutaneous applications as antiseptics or deodorants (e.g., the<br />

chlorophyllin copper complex, gluconate, oleate, or citrate) or in<br />

transdermal drugs (copper salicylate, copper phenylbutazone).<br />

Because of the increasing need for reliable skin irritation tests and in<br />

order to reduce the number of animal experiments, in vitro alternatives<br />

have been developed. So far, in vitro studies show that different chemicals<br />

induce irritant inflammatory responses, which vary considerably in the time<br />

course of the response, and that there are differences in the components of<br />

the inflammatory response to different irritants. Although no single test can<br />

be considered as an indirect, though reliable, measure of skin irritation in<br />

vivo, a battery of tests, each addressing a different aspect of such multifactorial<br />

phenomena leading to skin irritation, may well be a critical step<br />

preparatory to in vivo testing in humans.<br />

Distinguishing between irritant and allergic contact dermatitis can be<br />

challenging; thus, copper cross-reactivity/concomitant sensitization with<br />

other transition metals and failure by practitioners to resort to patch testing<br />

for resolution of questionable skin reactions, in many cases, leads to questionable<br />

diagnosis of irritation.


112 Hosty´nek and Maibach<br />

Cu (II) sulfate is clearly an irritant when applied in pet under occlusion<br />

for 48 hours. However, there are no currently available data that allow us to<br />

determine the threshold for induction of acute or cumulative irritancy<br />

dermatitis for copper or any of its salts. Fortunately, the technology to<br />

define this is readily available (cumulative irritancy testing). These are now<br />

being generated in this laboratory.<br />

ABBREVIATIONS<br />

ACD allergic contact dermatitis<br />

ARL adult rat lung<br />

ELISA enzyme-linked immunoassay<br />

ICD irritant contact dermatitis<br />

ICU immunologic contact urticaria<br />

IUD intrauterine device<br />

LDH lactate dihydrogenase<br />

MM mitochondrial membrane<br />

MTT mitochondrial toxicity test<br />

NICU nonimmunologic contact urticaria<br />

SC stratum corneum<br />

TEWL transepidermal water loss<br />

TLV threshold limit value<br />

REFERENCES<br />

1. ILO. Occupational exposure limits for airborne toxic substances. Occupational<br />

safety and health series. 3rd ed. International Labor Organisation, Geneva,1991.<br />

2. ACGIH. Threshold limit values for chemical substances and physical agents and<br />

biological exposure indices. Cincinnati, Oh: American Conference of<br />

Governmental Industrial Hygienists, 2001.<br />

3. Pilardeau PA, Lavie F, Vayasse J, et al. Effect of different work-loads on sweat<br />

production and composition in man. J Sports Med Phys Fitness 1988; 28:247–252.<br />

4. Lampe MA, Burlingame AL, Whitney J, et al. Human stratum corneum lipids:<br />

characterization and regional variations. J Lipid Res 1983; 24:120–130.<br />

5. Wertz PW, Swartzendruber DC, Kathi C, Downing DT. Composition and<br />

morphology of epidermal cyst lipids. J Invest Dermatol 1987; 89:419–425.<br />

6. Collins KJ. The corrosion of metal by palmar sweat. Br J Ind Med 1957; 14:<br />

191–194.<br />

7. Syrjänen S, Hensten-Pettersen A, Kangasniemi K, Yli-Urpo A. In vitro and<br />

in vivo biological responses to some dental alloys tested separately and in combinations.<br />

Biomaterials 1985; 6:169–176.<br />

8. Boman A, Karlberg AT, Einarsson O, Wahlberg JE. Dissolving of copper by<br />

synthetic sweat. Contact Dermatitis 1983; 9:159–160.


Irritation Potential of Copper Compounds 113<br />

9. Lidén C, Nordenadler M, Skare L. Metal release from gold-containing jewellery<br />

materials: no gold release detected. Contact Dermatitis 1998; 39:281–285.<br />

10. Wataha JC, Lockwood PE. Release of elements from dental casting alloys into<br />

cell-culture medium over 10 months. Dent Mat 1998; 14:158–163.<br />

11. Wataha JC, Craig RG, Hanks CT. The release of dental casting alloys into<br />

cell-culture medium. J Dent Res 1991; 70:1014–1018.<br />

12. Wataha JC, Hanks CT, Craig RG. In vitro synergistic, antagonistic and duration<br />

of exposure effects of metal cations on eukaryotic cells. J Biomed Mat Res 1992;<br />

26:1297–1309.<br />

13. Wataha JC, Hanks CT. Correlation between cytotoxicity and the elements<br />

released by dental casting alloys. Int J Prosthodont 1995; 8:9–14.<br />

14. Wataha JC, Hanks CT, Sun Z. In vitro reaction of macrophages to metal ions<br />

from dental biomaterials. Dent Mat 1995; 11:239–245.<br />

15. Grimsdottir MR, Hensten-Pettersen A, Kullmann A. Cytotoxic effect of orthodontic<br />

appliances. Europ J Orthodont 1992b; 14:47–53.<br />

16. Frentz G, Teilum D. Cutaneous eruptions and intrauterine contraceptive copper<br />

device. Acta Derm Venereol (Stockh) 1980; 60:69–71.<br />

17. Vilaplana J, Romaguera C. Contact dermatitis and adverse oral mucous<br />

membrane reactions related to the use of dental prostheses. Contact Dermatitis<br />

2000; 43:183–185.<br />

18. Holst R, Moller H. One hundred twin pair patch tested with primary irritants. Br<br />

J Dermatol 1975; 93:145–149.<br />

19. Rystedt I. Factors influencing the occurrence of hand eczema in adults with<br />

a history of atopic dermatitis in childhood. Contact Dermatitis 1985; 12:185–191.<br />

20. Rietschel RL, Mathias CGT, Fowler JF Jr., et al. Relationship of occupation to<br />

contact dermatitis: evaluation in patients tested from 1989 to 2000. Am J<br />

Contact Dermatol 2002; 13:170–176.<br />

21. Cartwright GE, Markowitz H, Shields GS, Wintrobe MM. Studies on copper<br />

metabolism 29. A critical analysis of serum copper and ceruloplasmin concentrations<br />

in normal subjects, patients with Wilson’s disease and relatives of patients<br />

with Wilson’s disease. Am J Med 1960; 28:555–563.<br />

22. Burch RE, Hahn HKJ, Sullivan JF. Newer aspects of the roles of zinc, manganese<br />

and copper in human nutrition. Clin Chem 1975; 21:501–520.<br />

23. Miller SJ. Nutritional deficiency and the skin. J Am Acad Dermatol 1989; 21:<br />

1–30.<br />

24. Lerner AB, Fitzpatrick TB, Calkins E, Summerson WH. Mammalian tyrosinase:<br />

the relationship of copper to enzymatic activity. J Biol Chem 1950; 187:793–802.<br />

25. Menkes JH, Alter M, Steigleder GK, Weakley DR, Sung JH. A sex-linked recessive<br />

disorder with retardation of growth, peculiar hair and focal cerebral and cerebellar<br />

degeneration. Pediatrics 1962; 29:764–779.<br />

26. Chernomorsky SA, Segelman AB. Biological activities of chlorophyll derivatives.<br />

New Jersey Med 1988; 85:669–673.<br />

27. DHHS. Deodorant drug products for internal use for over-the-counter human<br />

use CFR 21. Part 357. Fed Reg 1985; 50:25,162–167.<br />

28. Sorenson JRJ. Progress in medicinal chemistry. Amsterdam: Elsevier, 1978.<br />

29. Sorenson JRJ. The anti-inflammatory activities of copper complexes. In: Siegel<br />

A, ed. Metal ions in biological systems. New York: Marcel Dekker, 1982:77–123.


114 Hosty´nek and Maibach<br />

30. Hangarter W. Inflammatory diseases and copper. Clifton, NJ: Humana Press,<br />

1982.<br />

31. Sorenson JRJ, Berthon G. Copper potentiation of non-steroidal antiinflammatory<br />

drugs. New York: Marcel Dekker, 1995.<br />

32. Milanino R, Marrella M, Gasperini R, Pasqualicchio M, Vela G. Copper and<br />

zinc body levels in inflammation: an overview of the data obtained from animal<br />

and human studies. Agents Actions 1993; 39:195–209.<br />

33. Siller GM, Seymour GJ. Kinetics and specificity of nickel hypersensitivity in the<br />

murine model. Australasian J Dermatol 1994; 35:77–81.<br />

34. Eley BM. Tissue reaction to implanted dental amalgam, including assessment by<br />

energy dispersive X-ray micro-analysis. J Pathol 1982; 138:251–272.<br />

35. Gniewek J, Pezy J, Baker BG, Bokris JOM. The effect of noble metal addition<br />

upon the corrosion of copper. J Electrochem Soc 1978; 125:17–23.<br />

36. McNamara A, Williams DF. The response to intramuscular implantation of<br />

pure metals. Biomaterials 1981; 2:33–40.<br />

37. McNamara A, Williams DF. Enzyme histochemistry of the tissue response to<br />

pure metals implants. J Biomed Mater Res 1984; 18:184–206.<br />

38. Yli-Urpo A, Parvinen T. Metal degradation and tissue accumulation following<br />

subcutaneous implantation of combinations of materials. Proc Finn Dent Soc<br />

1980; 76:124–128.<br />

39. Schmalz G, Arnholt-Bindsley D, Hiller K-A, Schweikl H. Epithelium-fibroblast<br />

co-culture for assessing mucosal irritancy of metals used in dentistry. Eur J Oral<br />

Sci 1997; 105:86–91.<br />

40. Borenfreund E, Puerner JA. Cytotoxicity of metals, non-metal and metalchelator<br />

combinations assayed in vitro. Toxicology 1986; 39:121–134.<br />

41. Romeu-Moreno A, Aguilar C, Arola L, Mas A. Respiratory toxicity of copper.<br />

Environ Health Perspect 1994; 102(suppl 3):339–340.<br />

42. Schedle A, Samarapoompichit P, Rausch-Fan XH, et al. Response of L-929<br />

fibriblasts, human gingival fibroblasts, and human tissue mast cells to various<br />

metal cations. J Dent Res 1995; 74:1513–1520.<br />

43. Reuling N, Pohl-Reuling B, Keil M. Histomorphometrische Untersuchungen der<br />

Gewebevertraeglichkeit dentaler Legierungen. Deutsche Zahnaerztliche Zeitschrift<br />

1991; 46:215–219.<br />

44. Gate Y, Niisat N, Sakurai T, Furuyama S, Sugiya H. Comparison of the<br />

characteristics of human gingival fibriblasts and periodontal ligament cells.<br />

J Periodontol 1995; 66:1025–1031.<br />

45. Ratkay LG, Waterfield JD, Tonzetich J. Stimulation of enzyme and cytokine<br />

production by methyl mercaptan in human gingival fibroblasts and monocyte<br />

cell cultures. Arch Oral Biol 1995; 40:337–344.<br />

46. Brosin A, Wolf V, Mattheus A, Heise H. Use of XTT-assay to assess the cytotoxicity<br />

of different surfactants and metal salts in human keratinocytes (HaCaT).<br />

Acta Derm-Venereol (Stockh) 1997; 77:26–28.


7<br />

Copper Hypersensitivity: Dermatologic<br />

Aspects—Overview<br />

Jurij J. Hosty´nek and Howard I. Maibach<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

INTRODUCTION<br />

Reports of immune reactions of both the immediate and delayed types due to<br />

cutaneous or systemic exposure to copper have been reviewed, in the endeavor<br />

to draw a comprehensive profile of the immunogenic potential of that metal<br />

and its compounds. Also the metal’s immunotoxic potential is briefly reviewed.<br />

In principle, as noted for other transition metals, the electropositive<br />

copper ion is potentially immunogenic due to its ability to diffuse through<br />

biological membranes to form complexes in contact with tissue protein.<br />

Based on predictive guinea pig test and the local lymph node assay,<br />

copper has a low sensitization potential. Reports of immune reactions<br />

to copper include immunologic contact urticaria (ICU), allergic contact dermatitis,<br />

systemic allergic reactions, and contact stomatitis, but considering<br />

the widespread use of copper intrauterine devices (IUDs) and the importance<br />

of copper in coinage, items of personal adornment, and industry, unambiguous<br />

reports of sensitization to the metal are extremely rare, and even fewer<br />

are the cases that appear clinically relevant.<br />

This chapter, in part, was reprinted from Hosty´nek JJ, Maibach HI. Copper hypersensitivity:<br />

dermatological aspects—an overview. Rev Environ Health 2003: 18:153–185, with permission.<br />

115


116 Hosty´nek and Maibach<br />

Reports of immune reactions to copper mainly describe systemic exposure<br />

as cause to IUD, and to prosthetic materials in dentistry, implicitly<br />

excluding induction of hypersensitivity from contact with the skin as a risk<br />

factor. We provide a diagnostic algorithm that might clarify the frequency<br />

of copper hypersensitivity.<br />

Objective<br />

Intent of the overview is to establish a synopsis of dermatologic immune<br />

reactions ascribed to copper exposure, and to examine the criteria applied<br />

in such diagnosis, since not always has such causation been demonstrated<br />

unequivocally. The review discusses metallurgy of copper, predictive and<br />

diagnostic tests and describes types of immune reactions and the potential<br />

for the copper ion to act as sensitizer, followed by critical examination of<br />

literature reports applying strict diagnostic criteria, with consideration given<br />

to a number of confounding factors that may have led earlier investigators to<br />

erroneous interpretation of signs, symptoms, or test results.<br />

The last decade has seen a marked expansion in interest in metalallergic<br />

contact dermatitis—from a focus mainly on nickel and chromate<br />

to currently gold, cobalt, palladium, and others. Case report methodology<br />

now is much of the literature citations in this area. Here, we critically review<br />

the citations and suggest diagnostic criteria that might clarify how often<br />

copper hypersensitivity occurs in man.<br />

The skin is a target organ and indicator for allergy. While the stratum<br />

corneum (SC) is a partial barrier to the passive penetration of allergens, to<br />

electrophilic, protein-reactive metals in particular, live tissue of the epidermis<br />

and dermis actively process penetrants or systemically absorbed<br />

allergens that reach it. Such immune reactions to chemicals in the skin are<br />

broadly categorized into two distinct classes:<br />

1. Allergic contact dermatitis (ACD) or delayed-type reactions<br />

mediated by allergen-specific T lymphocytes. It expresses as a wide<br />

range of cutaneous eruptions upon (a second) dermal contact or<br />

systemic exposure to haptens in individuals with preformed cellular<br />

immunity (type IV allergic reactions).<br />

2. ICU or immediate-type hypersensitivity, which involves IgE antibody.<br />

The latter most notably results in respiratory allergy, but<br />

can also manifest in separate stages collectively described as ‘‘contact<br />

urticaria syndrome’’ (1), local or generalized urticaria, urticaria with<br />

extracutaneous reactions such as asthma, rhinoconjunctivitis, and<br />

gastrointestinal involvement, and ultimately anaphylaxis (type I<br />

reactions).<br />

Copper has been alleged to sensitize de novo on systemic exposure following<br />

inhalation or implantation. The resulting dermatosis thus induced is


Copper Hypersensitivity 117<br />

described as systemic contact dermatitis or urticaria (2–5). Copper complexes<br />

are also known to elicit skin reactions upon systemic challenge in<br />

the previously sensitized organism (6).<br />

METALLURGY OF COPPER AND ITS ALLOYS,<br />

AND ITS ROLE AS SENSITIZER<br />

Dissimilar metals, combined in alloys for the fabrication of medical devices<br />

such as dental materials, evoke currents in electrolytic media such as saliva<br />

and degrade, resulting in a steady release of metal ions. In immediate proximity<br />

of dental restorations or IUDs, this can lead to adverse (intraoral or<br />

intrauterine) reactions such as lichenoid lesions of the oral or genital mucosa.<br />

Beyond local effects at the implant site, ions can be transported into distal<br />

tissues such as the skin, giving rise to pathological processes such as manifest<br />

allergic reactions. Among the metals that commonly form allergenic<br />

ions are nickel, cobalt, chromium, and mercury. Exposure type, duration,<br />

and environmental conditions (sweat, oxygen supply) in proximity of the<br />

metal are critical for mobilization of ions leading to induction or elicitation<br />

of immune reactions. As most articles of common human contact are alloys<br />

and not made of the pure metal itself, electrochemical interaction between<br />

components are significant for the release of allergenic ions potentially leading<br />

to immune reactions (7).<br />

Reports of copper as immunogen are few, and rarely could the clinical<br />

relevance of copper sensitivity be demonstrated with certainty. Consequently,<br />

the question as to incidence or prevalence of copper sensitivity<br />

among the general population is moot, the number of cases too low to<br />

express as percentages. Nevertheless, two characteristics of copper in contact<br />

with tissues put the metal into a category that renders appropriate a<br />

discussion of its role in inducing reactions in the immune system.<br />

Copper belongs to the family of electrophilic transition metals, which<br />

makes the copper ion highly protein reactive, i.e., likely to be haptenized,<br />

thus recognizable by the immune system as non-self or foreign.<br />

Although belonging to the nobler metals highly resistant to corrosion<br />

(oxidation, dissolution), in the physiological environment (IUD, dental<br />

materials, implants) or in contact with skin exudates, elemental copper is<br />

converted to diffusible forms that can penetrate biological membranes.<br />

This latter factor merits detailed discussion, also to lay the groundwork<br />

for demonstrating how copper and other metals eventually become<br />

biologically available from contact with endothelial and epithelial barriers.<br />

The oxidation of copper (0) in body fluids has been investigated as a<br />

factor that may determine induction or elicitation of immune reactions.<br />

Release of metal ions experimentally determined in synthetic body fluids<br />

may not adequately mimic the degree of corrosion (oxidation, release) as it<br />

occurs in contact with live skin or in the physiological environment,


118 Hosty´nek and Maibach<br />

however. This is due to the fact that composition of such media used for<br />

routine experimentation lacks important components that, in contact of<br />

foreign materials with a living organism, determine the nature of reaction<br />

product, the rate of reaction, and thus the path of diffusion of the end products<br />

through biological barriers.<br />

These formulas appear to omit important factors, for instance those<br />

present in skin exudates, which can play a determining role in metal oxidation:<br />

proteins, and, most importantly, free fatty acids in the sebum (8–13).<br />

Together with metal ions the latter are likely to form lipophilic soaps,<br />

presumably diffusible via the intercellular lipid matrix of the SC. Evidence<br />

for skin diffusivity in a model experiment was obtained by in vivo application<br />

of copper oleate over 24 hours on human back skin. Urinary copper levels<br />

were subsequently seen to increase significantly over several days (14). While<br />

it is a good indication of facilitated permeation, that result in itself does not<br />

indicate the actual path followed by the permeant, however. Evidence for an<br />

actual path of diffusion was obtained in a different experiment; localization of<br />

copper in the intercellular spaces was made visible through electron microscopy<br />

following application of copper acetate on human skin (15).<br />

Human plasma or serum are the most corrosive physiological media<br />

and can play a decisive role on the path towards systemic immunization.<br />

Comparative tests simulating corrosion of implant metals in vitro demonstrated<br />

that the electrochemical process of oxidation in the presence of<br />

enzymes, proteins, and other components of actual serum is accelerated in<br />

comparison to standard simulating media (16). Corrosion testing of<br />

implants thus becomes more relevant for in vivo conditions when it is conducted<br />

in a proteinaceous medium (whole blood, serum, saliva) (16,17).<br />

The present synopsis of hypersensitivity cases arising from contact<br />

with copper amply, albeit indirectly, confirms the diffusivity of copper derivatives<br />

through biological barriers. In addition, copper derivatives (abietate,<br />

naphthenate, oleate, sulfate, 8-quinolinolate) used as pesticides are reported<br />

to act as irritants when coming in contact with the skin—evidence for their<br />

diffusion beyond the SC, reaching the live strata of the skin (18–20).<br />

The practice of using copper compounds, including metallic copper, as<br />

patch test materials for diagnostic purposes in <strong>dermatology</strong> also is based on<br />

empirical evidence gathered for their diffusion to reach the live strata of the<br />

epidermis when applied under occlusion.<br />

Finally, conversion of copper metal to diffusible compounds has been<br />

demonstrated in our laboratory in a semiquantitative manner (unpublished<br />

data). The SC of human volunteers was analyzed in depth for copper<br />

content following application of finely distributed metal on the skin under<br />

semiocclusive conditions. After application of the metal as micronized powder<br />

on the volar forearm for periods up to 72 hours, inductively coupled<br />

plasma mass spectroscopy analysis of sequential tape strips showed that<br />

the gradients of copper distribution profiles increased proportionally with


Copper Hypersensitivity 119<br />

occlusion time, from 24 to 72 hours, rising to 10 ppm after the longest period,<br />

significantly above the initial background level of 2 ppm.<br />

PREDICTIVE IMMUNOLOGY TEST RESULTS FOR COPPER<br />

Thus far, copper has been tested for sensitization potential in two predictive<br />

tests: the guinea pig maximization test (GPMT), a standard method used as<br />

predictor of skin sensitization potential, and in the local lymph node assay<br />

(LLNA) (21,22).<br />

In the GPMT, on 20 guinea pigs Boman et al. (23) noted two positive<br />

reactions at 24 hours and seven at 48 hours after using 1% copper sulfate<br />

pentahydrate in pet. Karlberg et al. (24) later found no difference between<br />

copper-exposed and control animals at 1% to 0.1% CuSO4 in pet. Basketter<br />

et al. (25,26) obtained a 0% response in the same test, but later in the LLNA<br />

the result was positive.<br />

In the LLNA adapted to test for allergenicity of metal salts also, under<br />

modified conditions, cupric ion significantly increased lymph node cell proliferation.<br />

Testing of cupric ion as the chloride in dimethyl sulfoxide at 1%,<br />

2.5%, and 5% concentrations showed significant increases in lymph node<br />

cells proliferation, with ratios of test to control lymphocyte proliferation<br />

of 8.1, 13.8, and 13.6, respectively. Also, mice could be sensitized in the<br />

LLNA by application of copper (II) sulfate (27,28). When the National Toxicology<br />

Program Interagency Center for the Evaluation of Alternative<br />

Toxicological Methods tested cuprous chloride in the LLNA, that copper<br />

salt was also found to increase lymph node cells proliferation, resulting in<br />

a positive test reading (29).<br />

DIAGNOSTIC TESTS FOR HYPERSENSITIVITY<br />

A differential diagnosis of chemically induced urticaria (ICU), immediatetype<br />

irritant (non-immunologic contact urticaria) and ACD is sometimes<br />

difficult, particularly when dealing with strong irritants. In simplest terms,<br />

it is mainly based on concentration of the xenobiotic (agent) necessary to<br />

induce a skin reaction, and on the time course of reaction.<br />

The Open Test<br />

The material is applied to intact skin or slightly dermatotic skin, with wheal and<br />

flare developing in minutes, a positive indication of or ICU (see above).<br />

The SPT for Immediate-Type Allergy (Contact Urticaria)<br />

Sensitization is defined as a positive skin prick test (SPT) response with or<br />

without clinical symptomatology. One drop (20 mL) of putative allergen in<br />

an appropriate solvent (e.g., propylene glycol), vehicle (negative control),


120 Hosty´nek and Maibach<br />

and histamine in physiological saline (positive control) is placed on three<br />

separate sites on the volar aspect of the forearm. Using a sterile prick device<br />

inserted through the drop, the underlying superficial epidermis is gently<br />

pricked. One needle is used per skin site and discarded. Immediately after<br />

pricking, each skin site is blotted dry. After 15 to 30 minutes the skin sites<br />

are evaluated for wheal and flare response. An edematous reaction (wheal)<br />

of at least 3 mm in diameter, surrounded by a flare, and at least half the<br />

size of the histamine control is considered positive in the absence of such<br />

a reaction in the vehicle control. SPT positives are retested to confirm the<br />

response. Ultimately, diagnosis should be based on clinical history and<br />

negative controls (30). Unlike with the open test, controls are mandatory.<br />

Radioallergosorbent Test<br />

Immediate allergic hypersensitivity can be diagnosed by radioallergosorbent<br />

test (RAST), an in vitro immunologic procedure designed to detect specific<br />

IgE antibodies in serum (31). Initially, a hapten–protein conjugate between<br />

a reactive compound and human serum albumin (HAS) has to be synthesized<br />

for the radioimmunoassay. The allergen (hapten–protein conjugate)<br />

is coupled to a paper disc. IgE antibodies in a serum sample, which are specific<br />

for the conjugate, bind to the conjugate epitopes on the disc, and the<br />

portion of bound IgE is detected by 125 I-labeled anti-human IgE. Usually,<br />

the results are expressed as a percentage of the total activity, the ratio<br />

between the binding to the hapten-HAS disc and a disc onto which HAS<br />

had been coupled and run in the same experiment.<br />

The RAST Inhibition Test<br />

Also cross-reactivity of various haptens can be determined by the RAST<br />

method. Serial dilutions of conjugate are allowed to react with an individual’s<br />

serum. The mixture is then used for RAST determination. Degree<br />

of reduction of the serum RAST values after absorption are expressed as<br />

percent inhibition (32).<br />

The Patch Test for Delayed-Type Allergy (ACD)<br />

The key diagnostic tool for ACD is patch testing. Objective is to reproduce<br />

the skin reaction to a suspected allergen under controlled conditions,<br />

by dosing the substance (or a standard series of allergens) in a suitable<br />

vehicle at a nonirritant concentration on adhesive tape and placing it on<br />

the skin. Penetration through the SC is promoted by airtight occlusion.<br />

The test is left on for at least 48 hours. Some agents elicit reactions only after<br />

a substantial delay (late phase reactions), such as copper or gold, possibly<br />

due to their known anti-inflammatory activity (33–39). Compounds of poor<br />

skin diffusivity such as transition metal salts may produce reactions with<br />

considerable delays also (36,37).


Copper Hypersensitivity 121<br />

It is suggested that in order to resolve doubtful cases a more differentiated<br />

approach using dilution series is advisable for diagnostic purposes.<br />

This would include using a dilution series rather than the standard practice<br />

of applying the single 1% or 2% copper sulfate solution for patch test assessment<br />

of allergic response to the metal salt.<br />

Detection of allergens by patch testing with salts dissolved in water, dispersed<br />

in petrolatum, or in their elemental form, and subsequent removal of<br />

the allergen resulting in clinical improvement are the simplest and most direct<br />

connections between cause and effect. As described in greater detail below, a<br />

confounding factor in etiology and diagnosis for a number of transition<br />

elements, and particularly in the case of copper, is the well-documented<br />

cross-reactivity with other metal ions, primarily nickel, but also palladium.<br />

These are reactions occurring when haptens of similar size and electron shell<br />

configuration are transferred to the same carrier protein. In fact, in the<br />

majority of copper sensitivity cases reported, the patients, when tested for<br />

multiple metal allergies, were positive to two or more metals, nickel being<br />

the most frequent one (40). That is an allergen that can induce clinical<br />

manifestations in even minute amounts, and is ubiquitous in the normal<br />

environment (41). Thus, false-positive reactions to copper may be due to presence<br />

of trace contaminants in the putative cause for allergy, e.g., the IUD, or<br />

even in the diagnostic test material, e.g., the metal disc (also see later).<br />

Because of the inherent irritancy of copper sulfate under patch test<br />

occlusion, and the relatively small number of normal volunteer controls used,<br />

we estimate that the nonirritating dose for diagnostic testing approximates<br />

1% to 2% in petrolatum. For verification of copper allergic hypersensitivity,<br />

application of 1% copper sulfate in water or petrolatum is recommended by<br />

the International Contact Dermatitis Research Group, or of an occluded<br />

copper (metal) disc over 2 to 4 days. Petrolatum is the recommended vehicle<br />

for copper sulfate, although uniform distribution of the crystalline salt is<br />

problematic and poor penetration from petrolatum makes that choice less<br />

than ideal. Any positive reactions warrant further evaluation to ascertain<br />

clinical relevance (42).<br />

One alternative method advocates the use of metals in the elemental state<br />

for diagnostic skin tests, and several authors have used copper discs or currency<br />

for patch testing (33,43–53). The diagnostic value of this approach is<br />

put in question by one investigation, however, where metallic copper was<br />

immersed in synthetic sweat to analyze for metal release. Over 24 hours, the<br />

final copper concentration was 0.01%, considered by the authors to be too<br />

low to elicit a reaction except in highly sensitized individuals (54).<br />

Role of Vehicle in Patch Testing<br />

In choosing a vehicle for percutaneous penetration, a factor for consideration<br />

is the effect it will have on the skin membrane and thus its barrier


122 Hosty´nek and Maibach<br />

properties, since the solvent of a xenobiotic (metal) can significantly influence<br />

its diffusivity and thus bioavailability. Petrolatum, for instance, is a<br />

poor solvent for metal salts because the permeant remains suspended as fine<br />

particles affording less than ideal uniformity in skin contact, but on the<br />

other hand it has an occlusive effect that would increase skin hydration and<br />

thus promote diffusion of a hydrophilic compound. Another solvent that<br />

enhances penetration is dimethylsulfoxide (DMSO) (see later for mercury<br />

chloride). As an instance, Sharata and Burnette point to dimethyl formamide<br />

and dimethyl acetamide associated with DMSO, which cause swelling<br />

of basal SC cells and disrupt the normal keratin pattern. They located the<br />

electron-dense metal ions mercury and nickel in the intercellular spaces<br />

and corneocytes, whereas in control membranes those metals were seen<br />

almost exclusively in the intercellular space. Thus, certain solvents may<br />

modify intercellular solute diffusion to include the transcellular path (55).<br />

How the nature of the vehicle can either influence the rate of release of a<br />

compound or modify the barrier properties, thus determining the level of<br />

percutaneous absorption of xenobiotics, is illustrated by further examples<br />

from the literature. An instance of practical importance is the choice of<br />

vehicle in standard diagnostic skin patch testing for sensitization, with the<br />

aim of optimum release of allergen into the viable epidermis while avoiding<br />

allergic or irritant contact dermatitis, leading to false-positive reactions<br />

caused by the vehicle itself.<br />

The enhancing effect on skin penetration by DMSO was demonstrated<br />

by experimental results on guinea pigs in vivo. The use of neat DMSO as<br />

vehicle for 0.239 M HgCl2 significantly increased the skin penetration of<br />

the compound relative to water as vehicle, and thereby the percutaneous<br />

toxicity (56). Mortality for the test animals at three weeks post-treatment<br />

increased from 20% with water to 80% with DMSO. Permeability coefficients<br />

for mercury in DMSO averaged 2.7 10 3 –4 10 3 cm/hr for the first<br />

five hours, as compared to 0.44 10 3 –1.47 10 3 cm/hr when the carrier<br />

was water.<br />

Poorer penetration of salts formulated in petrolatum was demonstrated<br />

repeatedly. Fullerton et al. (57) explored the effect of water as vehicle<br />

for NiCl2 and of petrolatum for both NiCl2 and NiSO4 at l.32 mg Ni/mL<br />

through in vitro experiments with full-thickness human skin.<br />

Petrolatum was the poorest vehicle for NiC12 as less than 1 mg Ni/cm 2<br />

reached the receptor in 70 to 98 hours compared to 12.6 mg Ni/cm 2 in water.<br />

For NiSO4 in petrolatum, a poorer penetrant than NiCl2, no detectable<br />

nickel reached the receptor phase over 163 hours.<br />

The permeability coefficients for 2.4% zinc chloride through human<br />

skin in vitro were compared when applied from petrolatum and from a<br />

hydrogel over 72-hour periods. The permeability coefficient from petrolatum<br />

was 0.082 10 4 cm/hr. Applied from the hydrogel base, the permeability<br />

coefficient was 0.29 10 4 cm/hr, more than three times higher (58,59).


Copper Hypersensitivity 123<br />

One method for determining the optimal solvent in diagnostic skin<br />

patch testing of allergens is its application on hypersensitive patients and<br />

recording the ratio of positive elicitation reactions. Thereby the solvent is<br />

identified, which better promotes diffusion of the xenobiotic. The reaction<br />

threshold to nickel sulfate was tested in 53 sensitized patients in both water<br />

and petrolatum at equal concentrations (390 ppm) (60): in petrolatum three<br />

patients were positive, and five to the aqueous solution, leading to the conclusion<br />

that the mean reaction threshold for nickel sulfate in water is lower<br />

(0.43%) than in petrolatum (51%) due to better diffusivity in the former. Also<br />

the irritation potential of a chemical can be assessed by application in different<br />

solvents in dermatological diagnostics aiming to minimize the chemical’s<br />

potential for irritation and thus false-positive reactions. The irritant reactivity<br />

of nickel salts in petrolatum was found to be greater than in that of<br />

water (61). These two concordant examples from dermatological practice serve<br />

to illustrate the differentiated promotion of diffusivity by different vehicles.<br />

From experience in dermatological practice, particularly in consideration<br />

of the clinical picture emerging from the few cases that document<br />

copper allergy, and because of the complexity of the irritant dermatitis<br />

syndrome, adhering to the criteria set out in the Operational Definition of<br />

ACD is recommended when a definition of clinical relevance is sought (42,62).<br />

TEST CONCENTRATIONS FOR COPPER ACD<br />

Since relatively few dermatotoxicological investigations have researched<br />

copper’s characteristics as allergen, no definite value has been assigned as<br />

to copper sulfate’s threshold-inducing sensitization, nor is an optimal<br />

concentration defined that would reliably elicit reactions in the sensitized<br />

organism. Thus, the patch test doses vary: 1% aq. (Gruppo Italiano<br />

di Ricerca Dermatiti da Contatto); 1% to 2% pet.; in a dental screening tray<br />

concentrations include 1% aq. and 2% aq. (63,64).<br />

IMMUNOGENIC POTENTIAL OF COPPER<br />

Systemic ACD<br />

Systemically induced allergic disease that can be caused by T-cell-mediated<br />

reactions to metals (copper) (65), potentially occur when copper or coppercontaining<br />

alloy materials used in IUDs, implants in replacement surgery or<br />

orthodontic appliances, are oxidized with release of free copper ions. These<br />

are absorbed through the epithelia and carried to the skin and the mucosa<br />

via blood and the lymphatic circulation. There the allergen is intercepted<br />

by antigen-presenting cells and recognized by T cells that migrate to the<br />

lymph nodes with blastic transformation, proliferation of cytotoxic lymphocytes,<br />

and production of cytokines. These in turn recall neutrophils and<br />

eosinophils to the reaction site, cause capillary dilation and increased


124 Hosty´nek and Maibach<br />

permeability, resulting in cutaneous inflammation appearing as wheal and<br />

flare (Menne, 1996 membrane); lichen planus and asymptomatic contact<br />

hypersensitivity (dental alloy contact dermatitis) are increasingly being linked<br />

with oral exposure to materials used in dental fillings, orthodontic prostheses,<br />

cements and components of dentures, bridges, bands, and wires. Such reactions<br />

can be either immunologic contact stomatitis or systemic anaphylactic<br />

stomatitis (type I reactions), or delayed contact stomatitis (type II). In a few<br />

instances, copper was implicated as possible cause for the latter, as copper is<br />

commonly a part of alloys used in dental materials (Table 1) (33,66,67). In<br />

a study investigating the release of copper from a selection of orthodontic<br />

appliances in organic and inorganic solutions made up to different pH values<br />

to imitate the oral environment, Stoffolani et al. found that the levels of metal<br />

mobilized were well below those ingested with a normal daily diet. From that<br />

of result they concluded that the quantities released should be of no concern.<br />

The relevance of that conclusion, particularly for purposes of immunology,<br />

invite further discussion, however, to be pursued elsewhere (68).<br />

A study of professionals (dental technicians, orthodontists and their<br />

assistants) involved in making and handling such materials reveals that in<br />

Table 1 Copper (0) and Nickel (0) Content (wt%) of Dental Materials<br />

Alloy Cu, wt%<br />

Ni, wt%<br />

(other metals) Manufacturer<br />

Copper (0) 100 N/R AB JS Sjöding, Kista, Sweden<br />

JSC—gold alloy 11.5 N/R (Au, Ag,<br />

Pt, Zn, Ir)<br />

AB JS Sjöding, Kista, Sweden<br />

Degussa Training 87.5 N/R<br />

Hereaus Kultzer GmbH, Hanau,<br />

Metal<br />

(Zn, Sn, Co) Germany<br />

Trindium 87.0 1.0<br />

Trindium Corp. of America,<br />

(Al, Mn) Los Angeles, California, U.S.A.<br />

Duracast MS 81.6 4.1<br />

Duracast Inc., Brasilia,<br />

(Al, Fe, Mn) Sao Paulo, Brazil<br />

Goldent 76.0 0.5<br />

Goldent Inc., Brasilia,<br />

(Al, Zn, Mn) Sao Paulo, Brazil<br />

Modulay 77.0 N/R<br />

J. F. Jelenko, Armenac, NewYork,<br />

(Au, Ag, Pd) U.S.A.<br />

ANA 68 5.0 N/R<br />

AB Nordiska Affineriet ANA,<br />

(Ag, Sn, Hg) Helsingborg, Sweden<br />

Dispersalloy TM<br />

12.4 N/R<br />

J & J Dental Products Co.,<br />

(Ag, Sn, Zn) Tallaght, Dublin, Ireland<br />

Valiant TM<br />

20.0 N/R<br />

L.D. Caulk Co., Dentsply Intl,<br />

(Ag, Sn, Pd) Inc., Milford, Delaware (U.S.A.)<br />

Gallium alloy GF 15.0 N/R<br />

Tokuriki Honten Co. Ltd., Tokyo,<br />

(Ag, Sn, Pd) Japan<br />

Abbreviation: N/R, none reported.


Copper Hypersensitivity 125<br />

handling dental devices they also run the risk of developing hypersensitivity<br />

to allergenic materials, metals among them, as in one study 40% of orthodontists<br />

and 43% of dental assistants reported work-related skin problems (69).<br />

Copper Intrauterine Devices<br />

Copper metal in contact with biological substrates (as in IUDs) is highly<br />

reactive and releases free copper ions. Release in vivo was determined<br />

at 0.71 I-1 micro mol/day (45 I-1 micro g) from a surface area of 200 mm 2 and<br />

1.29 I-1 micro mol/day (82 mg) in a culture medium in vitro (70).<br />

After it was discovered that copper metal placed in the uterus of animals<br />

had a contraceptive effect (71), the principle was applied to humans: a<br />

plastic T-shaped device with copper wire or a copper sleeve was introduced<br />

as a pharmacologic agent and became widely used as an IUD to regulate<br />

fertility. Research suggests that copper prevents fertilization rather than<br />

implantation (72).<br />

Reports of untoward reactions by women using the IUD (generalized<br />

eczema, edema) sometimes mention that the condition worsens during the<br />

perimenstrual period of the cycle, and patients often test positive to patches<br />

of copper as metal or as the sulfate (48). Upon removal of the device<br />

patients usually experienced complete remission.<br />

Dual Immune Response to Copper<br />

While organic compounds infrequently cause both types of reactions, dual<br />

immune response appears more common for metals and metallic compounds.<br />

Their reactivity towards protein results in a complete antigen that triggers<br />

both IgE antibody production (type I) and cellular (T cell, type IV) immune<br />

reactions. Immunogenic effects that result from exposure to metals can be<br />

attributed to the same factors that determine their toxicological and biological<br />

effects. Metal ions in general, and certainly those belonging to the transition<br />

group of elements, such as copper, have an ionic radius too small to be<br />

antigenic. Containing a partially filled d shell, these metals oxidize to highly<br />

electropositive cations that can act as haptens interacting with tissue protein.<br />

They form bonds that range from fully ionized to fully chelated complexes,<br />

and have the ability to modify the native protein configuration. These are<br />

recognized as non-self by hapten-specific T cells in the host immune<br />

system (73), leading to allergic reactions of the two different types.<br />

Copper is one of several metals causing more than one type hypersensitivity<br />

presenting with multiple symptoms in allergic responses, in part<br />

depending on type of exposure: immediate type, immunologic contact urticaria<br />

sometimes associated with respiratory hypersensitivity, delayed-type<br />

cutaneous hypersensitivity, systemic allergic reactions, and contact stomatitis<br />

(2–4,33,45,47,51,66,74,75).<br />

Concurrent occurrence of immediate- and delayed-type sensitivity has<br />

also been observed in the same individual (50,76).


126 Hosty´nek and Maibach<br />

It should be noted, however, that, overall, human case reports of<br />

copper-induced immunologic reactions are rare.<br />

The Major Factors with the Potential to Induce Copper Sensitivity<br />

Dental Materials<br />

Over the past 15 years, there has been considerable effort to replace the use<br />

of traditional materials such as mercury in dental restorative work, and<br />

dental casting alloys contain increasing amounts of copper. This use of various<br />

substitute, metal-based materials has proceeded without the necessary<br />

corollary knowledge of their irritant and allergenic potential, however.<br />

In reports of contact stomatitis (contact allergy of the oral mucosa), no<br />

reports of rechallenge have been published which would strengthen the<br />

causal association with copper.<br />

Tables 2–4 list the different types of allergic reactions associated with<br />

exposure to copper.<br />

Recommended Patch Test Procedure in Suspected Copper Allergy<br />

of the Delayed Type<br />

Establish clinical history (anamnesis) determining nature of contact<br />

and physical form of putative allergen.<br />

Physical examination of the patient.<br />

Patch testing with 2% CuSO4 in petrolatum. In case of positive<br />

outcome follow up with serial dilution patch testing (1%, 0.5%,<br />

and 0.1%).<br />

Repeat open application test (ROAT) or provocative use test<br />

(PUT) with a dilution series of CuSO4: 2%, with at least 10 na€ve<br />

control subjects to demonstrate that positive reaction is not irritant<br />

in nature, then further at 1% and 0.5%. The substance is applied<br />

once or twice daily for 14 to 28 days (107,108). A positive reaction<br />

usually appears within four days, less frequently between five and<br />

seven days. Delayed reactions have been noted in patch testing<br />

with copper.<br />

To confirm positive patch test reactions and identify false-negative<br />

reactions on patch testing, intradermal tests may be considered<br />

(109). Herbst et al. (110) provide the scientific background of intradermal<br />

testing for ACD.<br />

As an ‘‘alternative’’ predictive test for ACD, the local lymph node<br />

assay was developed on mice for the detection of contact allergens (22). It<br />

has been adapted to test for allergenicity of metal salts also. Under modified<br />

conditions, cupric ion was seen to significantly increase lymph node cell<br />

proliferation, as mice could be sensitized by application of copper (II)<br />

sulfate (27,28).


Copper Hypersensitivity 127<br />

Table 2 Immunologic Contact Urticaria Due to Copper<br />

Visible signs and<br />

symptoms Diagnostic test Challenge reaction CR References<br />

Cases Cohort/etiology<br />

1 Dental Generalized rash None None 1? 77<br />

1 Occup., Cu–ammonia Bronchial asthma Cu/NH3 inhal.n Dyspnea 1 78<br />

1 Dermatology patient Generalized uricaria Scratch, 1% aq. CuSO4 Erythema 2 2<br />

(IUD)<br />

1 Dermatology patient Pruritus and Scratch, CuSO4 Positive 2 79<br />

(IUD)<br />

urticaria<br />

2 Patients (IUD) Rhinitis Cu prick test Positive 1–2 80<br />

1 Patient (IUD; dental) Urticaria, flushing, Cu metal, 5% aq. CuSO4, Punctate wheals 1? 47<br />

pruritus<br />

actyl choline patch<br />

1 Adhesive pads Urticaria, eczema Open/closed patch 5% Pos. to copper acetyl 3 76<br />

aq. CuSO4<br />

acetonate/urticaria; not to<br />

Cu metal<br />

1 Occup. exposure to Urticaria Copper metal 2% aq. CuSO4 Positive at 20 min. Positive 3 50<br />

copper<br />

at 24 hr<br />

Abbreviations: CR, clinical relevance; IUD, intrauterine device.


128 Hosty´nek and Maibach<br />

Table 3 Allergic Contact Dermatitis Due to Copper<br />

Visible signs and<br />

symptoms Patch test Challenge reaction CR References<br />

Cases Cohort/etiology<br />

3 Dermatol. patients Hand eczema Copper metal<br />

Positive<br />

0–1 43<br />

Brass metal<br />

Positive<br />

1 Metal contact Dermatitis 1% aq. CuSO4 Positive patch 2–3 81<br />

2 Dermatol. patients Dermatitis 10% aq. CuSO4 Positive patch 0–1 82<br />

1 Teleph. lineman Dermatitis Copper metal Positive 1 44<br />

1 Jewelry Eczema Copper coins<br />

Positive at 72 hr<br />

1–2 45<br />

1.25–5% aq. CuSO4 Positive at 24 hr<br />

1 Metalworker Eczema 2% aq. CuSO4, multiple Multiple positives, including 0–1 83<br />

salts<br />

Cu(II)<br />

10 Furniture polishers Dermatitis 5% aq. CuSO4 Positive–irritation? 1? 84<br />

1 Welder Eczema 0.1–2% CuSO4 pet. Positive at 72–96 hr 2 74<br />

6 Dermatol. patients Eczema 0.25–5% CuSO4 pet. Positive to Cu and Ni 0 85<br />

4 Agricultural Dermatitis 1% CuSO4, pet. Positive at 24–72 hr 0 19<br />

2 Jewelry Dermatitis 5% CuSO4 aq. Positive to Cu and Ni; irr? 1? 86<br />

140 Dermatol.<br />

Dermatitis Copper metal Positive 0–1 49<br />

patients/jewelry<br />

1 Occupational Itching, eczema 2% CuSO4 aq. Necrosis at 24 hr 1 50<br />

1 Dental patient Oral symptoms 1% CuSO4 pet. Positive patch 0–1 87<br />

2 Dentistry students None 1% CuSO4 aq. Positive patch 1 88<br />

1 Enameller Dermatitis 5% CuSO4, pet. Positive 0–1 89<br />

3 Jewelry Dermatitis 2% CuSO4 pet. Positive to Cu and Ni 0–1 90<br />

1 Painter Itching, edema 10% Cu pigment, in pet. Positive patch 1–2 91<br />

5 Agricultural Dermatitis 2% CuSO4, pet. Positive at 48–96 hr 0–1 92<br />

3 Dermatol. patients Oral symptoms 1% CuSO4 Positive patch 0–1 93<br />

1 Dermatol. patients Dermatitis 2% CuSO4, pet.<br />

Positive at 72 hr<br />

2–3 53<br />

Cu metal<br />

Positive at 72 hr<br />

Abbreviation: CR, clinical relevance.


Copper Hypersensitivity 129<br />

Table 4 Systemic Allergic Contact Dermatitis Due to Copper<br />

Visible signs and<br />

symptoms Patch test Challenge reaction CR References<br />

Cases Cohort/etiology<br />

1 Dermatol. patient Lychen planus Cu metal, CuO 1% Positive at 96 hr, negative 2–3 33<br />

(dental)<br />

aq. CuSO4<br />

1 Dermatol. patient Rash Cu metal Aerythema, vesicles, 0–1 46<br />

(dental)<br />

excoriation<br />

1 Dermatol. patient (IUD) Generalized eczema 5% aq. CuSO4 Positive, negative to other 2 3<br />

metals<br />

1 Dermatol. patient (IUD) Generalized dermatitis 5% CuSO4 10% NiSO4 Positive 1 94<br />

4 Dermatol. patients (IUD) Generalized dermatitis 2% aq. CuSO4 Positive patch 1 pos. to 2 4<br />

NiSO4<br />

3 Dermatol. patients (IUD) Generalized dermatitis, 0.01% CuSO4 Cu metal Pos. patch at 24/48 h. Pos. 1 48<br />

itching<br />

to Cu metal at 24 hr<br />

1 Dermatol. patient (IUD) Generalized dermatitis 1% aq. CuSO4 Positive patch 2 95<br />

1 Dermatol. patient (IUD) Pruritus and dermatitis 0.1%–1% CuSO4 Positive patch at 24 and 0–1 96<br />

72 hr<br />

3 Dermatol. patients (IUD) Eczema, edema CuSO4 None 1? 97<br />

1 Dermatol. patient (IUD) Edema, erythema 1% aq. CuSO4 Vesicles and spongiosis 1 98<br />

2 Dentistry students, staff, None 5% aq. CuSO4 Positive at 24 and 72 hr 0–1 99<br />

patients<br />

1 Dermatol. patient (IUD) Nephritis CuSO4 Pos. to CuSO4 and NiSO4 1 100<br />

2 Dermatol. patients Oral lesions 5% aq. CuSO4 Positive patch at 72 hr 1 67<br />

(dental)<br />

1 Dermatol. patient Multiple metal<br />

Cu and other metal Eczema 0–1 51<br />

(dental)<br />

sensitivities<br />

patches<br />

(Continued)


130 Hosty´nek and Maibach<br />

Table 4 Systemic Allergic Contact Dermatitis Due to Copper (Continued )<br />

Visible signs and<br />

symptoms Patch test Challenge reaction CR References<br />

Cases Cohort/etiology<br />

1 Dermatol. patient Gen. oral eruptions 1% CuSO4 pet. Positive patch 0–1 101<br />

(dental)<br />

40 Dentistry students Metal allergies 1% aq. CuSO4 Positive patch 0 102,103<br />

1 Dermatol. patient (IUD) Perimenstrual<br />

0.01–1% aq. CuSO4 Positive patch, positive 1 52<br />

angioedema<br />

Cu metal<br />

1 Dermatol. patient (IUD) Generalized skin Copper salt (?) Positive to copper and 0–1 104<br />

reactions<br />

silver salt<br />

1 Dermatol. patient (IUD) Perimenstrual dermatitis 2% aq. CuSO4, other Positive 1? 105<br />

metal salts<br />

1 Dermatol. patient Lychen planus 2% aq. CuSO4, other Positive to Cu and other 0–1 68<br />

(dental)<br />

metal salts<br />

salts at 4 days<br />

3 Dermatol. patient Orodynia, lychen planus 2% aq. CuSO4 Positive at 48 hr 0–1 106<br />

(dental)<br />

Abbreviations: CR, clinical relevance; IUD, intrauterine device.


Copper Hypersensitivity 131<br />

Recommended Screening Procedure in Suspected<br />

Copper Urticaria<br />

Open test: Application on healthy skin first and observation of the<br />

test area for 60 minutes. If reaction is negative, on previously<br />

affected skin (as suggested by patient’s anamnesis) spreading of<br />

2% aq. CuSO4 on a 3 3 cm area. Immunologically mediated<br />

reactions usually appear within 15–20 minutes, nonimmunologic<br />

ones within 45–60 minutes after application (111). This difference<br />

in delay is a major distinction between specific and nonspecific contact<br />

urticaria. A positive reaction is seen as edema or erythema<br />

(wheal and flare). A minimum of 10 na€ve background controls<br />

with the test solution is suggested. A nonimmunologic reaction will<br />

appear in the controls due to release of inflammatory mediators<br />

from the cells without participation of specific IgE antibody (112).<br />

A use test is suggested, handling the suspected agent and recreating<br />

the original scenario inducing the reaction (108).<br />

When open application is negative, a prick test with 2% aq. CuSO4<br />

is suggested. A group of more than 10 background controls is<br />

required in prick testing using physiologic saline solution to ascertain<br />

that copper does not produce such lesions in normal controls.<br />

The occluded application of a copper disk over 48 hours can also<br />

confirm suspected sensitization.<br />

In case of a positive test the open application may be repeated for<br />

verification.<br />

Confounding Factors in Copper Allergy Test Results: Cross-Reactivity,<br />

Contaminants, Irritation, and Angry Back Syndrome<br />

In many cases where copper allergy is suspected, positive patch tests to<br />

copper (as metal or the sulfate) are equivocal, and assignment of clinical<br />

relevance can be difficult or impossible because case reports in the literature<br />

most often lack relevant details. One element of uncertainty in the diagnosis<br />

of copper allergy is its cross-reactivity with other (adjacent) transition metals<br />

in the periodic system of elements. Observations of multiple sensitivity to<br />

metals have been made frequently, attributed to cutaneous or systemic contact<br />

with alloys, and it is challenging for the investigator to ascribe the clinical<br />

observation either to concomitant sensitization or to cross-reactivity. Often<br />

patients react to compounds that are not the primary sensitizer. Originally,<br />

Epstein (82) had raised the question of nickel and copper cross-sensitization<br />

in 1955, and since then many cases of simultaneous sensitivity to nickel and<br />

copper in the same organism have been reported (Tables 2–5). The immunologic<br />

mechanism involved in hypersensitivity to multiple metals and crossreactivity<br />

between copper and other transition elements has been investigated<br />

in two independent in vitro studies and the event is well characterized now,


132 Hosty´nek and Maibach<br />

Table 5 Population Studies of Copper Hypersensitivity<br />

Visible signs and<br />

symptoms Patch test Challenge reaction CR References<br />

Cases Cohort/etiology<br />

10 10 Furniture polishers Dermatitis 5% aq. CuSO4 Positive; irritation? 1? 84<br />

0 37 Patients Dermatitis 5% pet. CuSO4 Negative – 113<br />

0 1190 Dermatol. patients Eczema 2%–0.125% pet. CuSO4 Negative – 24<br />

4 652 Agricultural workers Dermatitis 1% CuSO4, pet. Positive at 24–72 hr 0 19<br />

140 964 Dermatol.<br />

Dermatitis Copper metal Positive 0–1 49<br />

patients/jewelry<br />

1 10,936 Dermatol. patients Allergy 2%–0.01% aq./5% pet. Positive 1–2 114<br />

CuSO4<br />

1 190 Enamellers and decorators Dermatitis 5% CuSO4, pet. Positive 0–1 89<br />

2 12 Dentistry students None 1% CuSO4 aq. Positive patch 1 88<br />

32 60 Dentistry students Metal allergies 5% aq. CuSO4 Positive patch 0 102,103<br />

5 46 Agricultural workers Dermatitis 2% CuSO4, pet. Positive at 48–96 hr 0–1 92<br />

5 311 Dental patients Allergies Metals (?) Metals (?) 0 115<br />

7 233 Metal workers Metal allergies 1% aq. CuSO4 ? 0 116<br />

3 520 Dermatol. patients Oral symptoms 1% CuSO4 Positive patch 0–1 93<br />

1 2660 Dermatol. patients Dermatitis 2% CuSO4, pet. Positive at 72 hr 2–3 52<br />

Cu metal<br />

Positive at 72 hr<br />

1 60 Dermatol. patients Dermatitis 1% aq. CuSO4<br />

Positive patch 2 40<br />

Copper alloy


Copper Hypersensitivity 133<br />

making it possible to put the numerous case reports on copper-induced allergy<br />

in better perspective. Specifically, nickel ion-specific T-cell clones appear to be<br />

recognized both by copper and palladium ions, but not by others such as<br />

cobalt. This reactivity is likely to be favored by their bivalency and proximity<br />

to nickel in the periodic table of elements. Investigations showed that among a<br />

large panel of nickel-specific T-cell clones four different types of reactivity can<br />

occur: reactivity to nickel only, cross-reactivity between nickel and palladium,<br />

cross-reactivity of nickel to copper, or to both palladium and copper ion,<br />

which both neighbor nickel in the periodic table of elements (117,118). In light<br />

of these results, copper-positive patients are now more often screened for<br />

allergy to other metals also, but only few among them are found to be truly<br />

copper-sensitive. To illustrate the possibility of cross-reactions due to dental<br />

materials in particular, a number of commercial alloys are tabulated, with<br />

special attention given to the listed presence of copper and nickel (Table 1).<br />

Purity of test materials can be a source of diagnostic equivocation with<br />

the potential for false-positive results. Copper patch test material may contain<br />

nickel as an impurity, as analytical grade copper sulfate was shown to<br />

contain up to 0.002% nickel; high-purity copper wire in IUDs, which is also<br />

used for skin testing, contain 0.0003% (3 ppm) nickel (24). Note that with<br />

metal ACD in humans, highly sensitized subjects can react down to a few<br />

parts per million of the hapten (119).<br />

A potential cause of false-positive, clinically nonrelevant reactions that<br />

can result in patch testing is hyper-reactive skin, also known as the excited skin<br />

syndrome or ‘‘angry back’’ (120–122). This condition can result from multiple<br />

inflammatory skin conditions or from strong positive patch-test reactions,<br />

magnifying adjacent patch test responses or inducing nonspecific reactions. This<br />

is a potential occurrence in testing for copper when several different metal<br />

patches are simultaneously applied on the patient. Multiple positive reactions<br />

may require separate, sequential tests with the involved substances.<br />

Finally, several studies, especially those involving retrospective reviews<br />

or large population groups, routinely examine skin reactions at 48 hours,<br />

missing potential late-phase (72 hr) reactions after patch application (Table 5)<br />

(33–39). They may result in false-negative diagnoses and under-reporting of<br />

hypersensitivity to copper.<br />

Determining Clinical Relevance<br />

The open literature has been critically reviewed for clinical relevance of the<br />

cases reported. A problem encountered often in the evaluation of diagnostic<br />

tests from patients reacting to chemical substances is understanding the clinical<br />

relevance of test results, because little or no data are reported to qualify<br />

positive results. This becomes particularly difficult in the interpretation of<br />

tests, which appear to indicate a compound as primary sensitizer that<br />

is known to have no or little sensitization potential, such as copper.


134 Hosty´nek and Maibach<br />

Benezra et al. have addressed the problem of classification by suggesting a<br />

systematic analysis of available data to arrive at an expression of degree<br />

of confidence in the results reported by investigators, thus to better define<br />

morbidity of a putative allergen. A degree of confidence has been assigned<br />

to all cases listed in Tables 2–5, listing the literature reports. Although<br />

Benezra et al. (123) designed the system with skin contact sensitizers in<br />

mind, which lead to delayed-type reactions, the approach appears more<br />

generally valid and is applied to all cases reviewed here.<br />

Criteria for Assignment of Degree of Confidence<br />

Presence of vehicle-treated or untreated controls<br />

Concentration of test substance judged sufficient to elicit a<br />

response<br />

Use of an appropriate vehicle<br />

Purity of test reagent to exclude possible reaction to contaminants<br />

Sufficient number of cases for meaningful response<br />

The evidence provided in the reports is evaluated towards classification<br />

of the agent (copper) as allergen and a degree of confidence on a scale<br />

from 0 to 5 is assigned to indicate how well the test results demonstrate that<br />

the chemical does or does not induce the immune reaction:<br />

5 ¼ results meet all of the criteria<br />

4 ¼ all criteria met, but number of cases is marginal<br />

3 ¼ parameters such as controls are missing but reports point to<br />

substance as sensitizer<br />

2 ¼ controls are absent and there are no other details indicating<br />

substance as sensitizer<br />

1 ¼ results not considered to be reliable<br />

0 ¼ test fails all of the criteria<br />

Since evaluation of criteria is subjective, degree of confidence should be<br />

viewed within a range of 1 of the number assigned. Listed in the following<br />

are two categories of reports relative to copper hypersensitivity: populationbased<br />

studies (also listed in Table 5), selected from published reports of immune<br />

reactions to copper, which surveyed larger samples—random cross-sections of<br />

the population, cohorts of specific occupational exposure, wearers of IUDs, dermatological<br />

clinic data bases, or groups exposed to copper in dental materials.<br />

That section is followed by selected case reports of more anecdotal value.<br />

SUMMARIES OF POPULATION-BASED STUDIES<br />

Barranco, 1972<br />

Upon review of the literature the author noted six cases of ACD to copper:<br />

three cases attributed to contact with brass, and one each to exposure to


Copper Hypersensitivity 135<br />

copper sulfate, copper metal, and jewelry. The author also reported on a case<br />

of dermatitis attributed to the use of a copper IUD. Although of questionable<br />

clinical relevance due to patch testing with 5% CuSO4, it holds a<br />

somewhat historical interest as it is the first report of eczematous dermatitis<br />

to copper due to systemic exposure. Tested for the other frequent metal<br />

allergens: Ni, Cr, Co, and Hg besides Cu, all patch tests were negative except<br />

for a strong reaction to 5% CuSO 4. Remission was noted after removal of<br />

the IUD (3).<br />

Dhir, 1977<br />

A cohort of 10 furniture polishers who had developed skin reactions on<br />

handling ethyl alcohol tinted with 5% copper sulfate were tested with that<br />

solution and aq. 5% CuSO4. All 10 patients reacted to both materials; the<br />

test with the same materials were negative on 15 control subjects (84).<br />

Jouppila, 1979<br />

Assessed were 37 patients wearing copper IUD and presenting with skin<br />

rashes. Epicutaneous tests for copper, nickel, and cobalt allergy showed<br />

reactions to nickel (four) and cobalt (one), but none to copper. The authors<br />

concluded that allergy to copper was not likely to be the cause of the side<br />

effects (124).<br />

Karlberg, 1983<br />

Of 1190 eczema patients tested with serial dilutions (2–0.125%) CuSO4 in<br />

pet. over a three-year period, none had a reaction to copper only, 13 reacted<br />

to copper and other metals. Thus, no sensitization to copper specifically<br />

became evident, leading to the assumption that the (multiple) reactions<br />

noted were due to metals contaminating the test allergen. According to<br />

Karlberg, highest-grade copper metal contains 0.0003% nickel, analytical<br />

grade copper sulfate up to 0.002%. In the GPMT using dilution series of<br />

0.1–0.01% CuSO4 for induction and 1–0.05% in pet. for elicitation, Karlberg<br />

determined that copper sulfate was a grade I allergen. In her review of the<br />

literature prior to 1982, Karlberg noted four relevant and 20 probably<br />

relevant cases of copper hypersensitivity. Over 90 cases were classified as<br />

uncertain or not relevant (24).<br />

Lisi, 1987<br />

The authors studied the prevalence of irritant or ACD from pesticides by<br />

patch tests on 652 outpatients with skin disorders. Of 564 subjects tested<br />

with 1% CuSO4, four cases showed positive reactions, none of which were


136 Hosty´nek and Maibach<br />

irritant morphology. They presumed that allergic reactions could not be<br />

considered of definite relevance due to the scarcity of clinical details. In particular,<br />

data are missing on confirmatory retesting of positive tests conducted<br />

2–3 months later (19).<br />

Romaguera, 1988<br />

Nine hundred and sixty-four <strong>dermatology</strong> patients complaining of metal<br />

intolerance, experienced mostly in contact with jewelry, were patch tested<br />

with standard allergens and metal washers. Of 52% of patients giving positive<br />

reactions to nickel, 14% were also positive to copper (among other<br />

metals), none to CuSO4 (concentration not given). The relevance of copper<br />

sensitivity is uncertain due to the minimal experimental details given, and<br />

the probability of contamination of the metal washers used (48).<br />

Zabel, 1990<br />

Records on 10,936 patch test reactions collected in a <strong>dermatology</strong> clinic over<br />

the period 1975–1985 were reviewed, in addition to patch tests conducted on<br />

118 patients wearing IUDs. Besides the record of patients with positive reactions<br />

to multiple metals (mostly nickel), one eczematous IUD-wearing<br />

patient reacted to CuSO4 at 5% in pet. only. After removal of the IUD<br />

the eczema resolved. The causative role of copper is uncertain due to lack<br />

in supporting evidence in that case (116).<br />

Motolese, 1993<br />

The authors reported on skin sensitization to metals encountered in a cohort<br />

of enamellers and decorators. Relevance of the only positive reaction to copper<br />

was uncertain due to the high concentration of 5% CuSO4 used in the<br />

test. Also, too few clinical details were given to establish a firm causeand-effect<br />

relationship in that case (89).<br />

Kawahara, 1993<br />

The cause of occupational allergies was investigated in a dental technology<br />

school by testing a cohort of 12 students with 40 potential contact allergens<br />

occurring in the manufacture of prostheses and was determined to be dust,<br />

mist, and fumes in their environment. Two reacted to 1% aq. CuSO4; the<br />

reactions could not be assessed as to their clinical relevance due to lack of<br />

any further details (88).<br />

Tschernitschek, 1998<br />

Over the period 1982–1997 in a dental clinic, of 311 patients who were patch<br />

tested for dental materials-induced hypersensitivity, 13% showed positive


Copper Hypersensitivity 137<br />

reactions. Most frequent among the sensitizing materials were metals (77 of<br />

107). Three among those reacted to copper and cadmium, two to copper<br />

only. Significance cannot be assigned due to total lack of experimental<br />

details (115).<br />

Candura, 1999<br />

Of 233 ACD outpatients patch tested with the standard GIRDCA test series<br />

in a <strong>dermatology</strong> clinic, three had positive reactions to copper along with<br />

other metals, four to copper only. The importance of the causative role of<br />

copper cannot be assessed due to a total lack of experimental details (116).<br />

Vilaplana, 2000<br />

A testing program including 520 patients with dental prostheses who presented<br />

with adverse oral mucous membrane reactions was conducted using<br />

a special metal test series that included 1% CuSO4 in pet. Of 289 patients<br />

with one or more positive reactions, one patient only reacted to copper, classified<br />

as a reaction of past relevance (sic) by the authors; two patients had<br />

reactions to copper with unknown relevance (93).<br />

Wöhrl, 2001<br />

In the endeavor to assess the relevance and diagnostic value of positive reactions<br />

to copper, 2660 routine patch tests recorded in an allergy clinic over<br />

2.5 years were screened for positive reactions to copper (2% CuSO4 in pet.)<br />

and the other metals in the immediate vicinity in the periodic system of<br />

elements: nickel, palladium, cobalt, and mercury. Of 94 cases that were<br />

copper-positive, 26 were enrolled in a retest program involving CuSO4 at<br />

5%, 2%, 1%, 0.6%, 0.2%, and 0.05% aq. Testing with copper foil was also<br />

included. Of the original 26 inductees, 10 were positive to copper on retesting<br />

with 5% CuSO4 in pet., but eight of those also reacted to a nickel patch.<br />

Two of 10 showed unequivocally positive reactions to 2% CuSO4 in pet. Two<br />

were positive to copper foil. Only one case showed an isolated sensitivity to<br />

copper and not to any of the other test allergens, presenting with chronic<br />

eczema of the fingertips. That patient’s occupation as electrician would<br />

characterize the case as ACD to copper induced through cutaneous contact.<br />

One other patient with multiple metal sensitivities appeared to have clinically<br />

relevant sensitivity to copper. Presenting with eczema to a golden ring<br />

(test to gold negative), the condition resolved when the patient exchanged<br />

the gold ring with one made of silver. Although authors concluded on<br />

copper–nickel cross-reactivity on the T-cell level in 9 of the 10 cases, with<br />

a high statistical association, and copper sensitivity being of low clinical<br />

relevance, all reactions cleared at 96 hours, a delay that is typical for irritant<br />

reactions rather than ACD (52).


138 Hosty´nek and Maibach<br />

SUMMARY OF SELECTED CASE REPORTS OF IMMUNE<br />

REACTIONS TO COPPER<br />

The process of diagnosis and remediation of copper allergy recorded in the<br />

literature so far follows an overly direct and simple path, with little or no<br />

clinical data. Solidly worked-up cases are rare, which makes assignment<br />

of relevance difficult and is the reason for the low scores in clinical relevance<br />

assigned to the reported cases in Tables 2–5. In most cases the examining<br />

physician (dermatologist), having confirmed ACD, often by CuSO4 patch<br />

test only, without controls or tests with other metals, advises the patient<br />

to remove amalgam or antifertility device. As a rule, then, improvement<br />

of the condition or complete remission follows promptly. As the signs of<br />

allergy are no longer present, no follow-up testing is done. Cases where<br />

no improvement was noted have not been published. With few exceptions,<br />

sensitization to copper results mostly from two types of exposure: leaching<br />

of the metal from dental amalgams, and from copper containing IUDs, due<br />

to corrosion in the physiological environment. The majority of signs and<br />

symptoms of allergy reported point to the delayed type, ACD, or systemic<br />

allergic contact dermatitis (SACD).<br />

At first glance, the literature search produced eight records with nine<br />

cases of ICU (Table 2), 21 records with 160 cases of ACD (Table 3), and<br />

21 records with 71 cases of SACD (Table 4). Many of the cases, diagnosed<br />

by using a 5% CuSO4 patch in pet., are classified as having borderline relevance,<br />

because patch tests at that strength are now considered to cause<br />

probable irritation—rather than ACD (3,89).<br />

A few of the cases listed in the tables merit detailed discussion due<br />

to their extraordinary characteristics. A number of them also show that<br />

sensitization originally thought to be due to copper actually resulted from<br />

exposure to other metal(s) or chemical agents.<br />

SELECTION OF INDIVIDUAL REPORTS OF IMMUNE<br />

REACTIONS TO COPPER<br />

Frykholm, 1969<br />

An example of SACD due to dental materials was reported by Frykholm et<br />

al. A patient with oral lichen planus-1ike reactions to dental restorative<br />

materials showed positive skin reactions to epicutaneous tests with copper<br />

metal, Cu (II) hydroxide, Cu (II) oxide, and Cu (I) oxide at 72 hours, while<br />

the test sites were unreactive at 48 hours. Tests carried out on 20 controls<br />

were negative. The patient’s oral lesions flared up in conjunction with the<br />

skin tests. On repeated retesting at month-long intervals with metallic copper<br />

and Cu (II) oxide, positive reactions to all test materials were until the<br />

fourth day (33).


Copper Hypersensitivity 139<br />

The late appearance of the skin reactions (24–96 hr) is characteristic of a<br />

‘‘delayed allergic reaction,’’ to gold in particular due to its anti-inflammatory<br />

activity (125,126). The delays in tissue involvement seen with copper may also<br />

be ascribed to that characteristic, which makes the metal an effective antirheumatic<br />

agent. An alternative explanation for the delayed reaction may<br />

involve the protein reactivity and depot formation by copper, resulting in<br />

delays in SC penetration as seen in other electrophilic metals, e.g., aluminum,<br />

silver, mercury, or chromium known for their retention in the SC (127–131).<br />

Shelley, 1983<br />

A case of potential contact urticaria, which defies interpretation, was<br />

reported for a woman with occult sensitivity to copper. Wearing an IUD<br />

and dental fillings with copper amalgam, she developed urticaria and flushing<br />

with pruritus upon physical exercise, emotional stress, or overheating,<br />

more severe during the perimenstrual period. On testing, metallic copper<br />

and copper-containing coins elicited no response, but on the test sites she<br />

developed punctate wheals when challenged with exercise or on injection<br />

with acetylcholine in subthreshold concentration. Also challenge with 5% aq.<br />

CuSO4 induced the same response as the copper metal. Tests with nickel<br />

metal and the sulfate were negative. That cholinergic urticaria reaction<br />

observed is attributed to a collaborative effect of acetylcholine and copper<br />

on mast cell membranes inducing degranulation, a clinical response that an<br />

antibody–antigen reaction on the mast cell surface alone could not produce.<br />

No subsequent such patients have been reported (46).<br />

Hocher, 1992<br />

A woman with IUD presented with interstitial nephritis and renal failure.<br />

Patch tests were positive for copper, nickel, and cobalt. An in vitro<br />

lymphocyte-stimulating test with copper was also positive. On removal of<br />

the contraceptive device renal function returned to normal. No similar cases<br />

have been published (100).<br />

Laubstein, 1990<br />

A case of occupational exposure to copper metal presented with an itching<br />

eczematous condition, which healed on absence and returned on resumption<br />

of work. That highly sensitized individual reacted with necrosis on 24-hour<br />

testing with 2% aq. CuSO4, and also on contact with copper metal. Followup<br />

brought an urticarial reaction after 20-minute exposure to copper metal,<br />

and to serial dilution of aq. CuSO4 after 24 hours, reactions that extended<br />

to necrotic ulceration. Upon changing occupation the patient became free<br />

of symptoms. This description suggests two mechanisms—ACD documented<br />

with a positive response to a copper disk, and immunologic contact<br />

urticaria (49).


140 Hosty´nek and Maibach<br />

Sterry, 1985<br />

A female patient reported with eczema and pruritus while wearing selfadhesive<br />

pads treated with the disinfecting agent copper-acetyl acetonate.<br />

Reactions to patches with copper-acetyl acetonate on 12 control subjects were<br />

weakly positive at 24 hours, disappearing after 48 hours. The test with metallic<br />

copper and CuSO4 on the patient was negative, but positive to the disinfectant<br />

at 50% pet. and 50% aq. On open patch testing, the patient was positive to<br />

both copper-acetyl acetonate 50% aq. and acetyl acetone (100%) alone. This<br />

suggests that allergy was probably due to acetyl acetonate and not copper (76).<br />

COMMENTS<br />

Many case reports of sensitization attributed to copper may be difficult to<br />

classify as such with certainty. Copper sensitivity may overlap with nickel<br />

hypersensitivity, or nickel alone may even be the only causative agent, as<br />

in dermatological or dental practice they can only be distinguished with<br />

difficulty when assessing exposure in the individual patient. As results from<br />

several in-depth investigations, patients with a positive test to copper also<br />

appear sensitized to nickel, and vice versa. This can be attributed to cellbiological<br />

and metallurgical factors:<br />

Investigations at the cellular level have established cross-reactivity<br />

between the two metals, which may account for the frequency of<br />

copper hypersensitivity reported (117,118).<br />

At the exposure level, often copper and nickel are associated in IUDs<br />

or orthodontic materials. Copper of highest purity still contains traces of<br />

nickel; thus, sensitization observed may be concomitant (24,124,132).<br />

In dermatological practice, diagnostic test materials copper sulfate<br />

or copper metal discs also contain low levels of nickel sufficient to<br />

elicit a reaction in an organism highly sensitive to nickel, leading<br />

to a false-positive diagnosis.<br />

There may be true allergic reactions to copper exposure, topical or systemic:<br />

to copper salts, to the metal, or to its alloys. Judging from the cases<br />

reviewed so far, such responses are rare.<br />

CONCLUSIONS<br />

Systemic as well as topical exposure to copper can cause both immediate<br />

and delayed-type sensitization. Contact dermatitis and urticaria attributed<br />

to copper metal or its compounds has been suggested, with effects from<br />

dental materials and IUDs as the main etiological factors. Immune reactions<br />

occurring in industry are few, considering the number of copper smelters


Copper Hypersensitivity 141<br />

and refinery workers in daily contact with the metal. The majority of sensitization<br />

reports may be due to copper cross-reactivity with nickel and<br />

palladium. Thus, true allergic reactions to copper appear rare, particularly<br />

those induced by skin contact, which is consistent with copper’s rating as<br />

a grade I allergen in the guinea pig maximization test. Most cases of confirmed<br />

copper allergy result from its presence in orthodontic materials,<br />

and those reactions are mostly of the delayed type.<br />

Firmer chemical and epidemiologic judgments will be possible when:<br />

1. Additional experimental data become available on the nonirritating<br />

dose(s) suitable for diagnostic patch testing (in petrolatum and<br />

water), and in water for prick testing. On the basis of Wöhrl’s<br />

data, 2% in petrolatum may be appropriate (52).<br />

2. Authors describe their clinical experimental data with details of<br />

the several steps as documented in the Operational Definition<br />

of ACD (56), specifically re-patch testing upon indication, serial<br />

dilution patch testing, and use testing (PUT/ROAT). Those steps<br />

will help clarify clinical relevance.<br />

ABBREVIATIONS<br />

ACD allergic contact dermatitis<br />

aq aqua<br />

ICDRG International Contact Dermatitis Research Group<br />

ICU immunologic contact urticaria<br />

IUD intrauterine device<br />

LLNA local lymph node assay<br />

pet. petrolatum<br />

PUT provocative use test<br />

ROAT repeat open application test<br />

GPMT guinea pig maximization test<br />

REFERENCES<br />

1. Lahti A, Maibach HI. Immediate contact reactions. In: Middleton E, Reed CE,<br />

Ellis EF, Adkinson NF, Yunginger JW, Busse WW, eds. Allergy: Principles<br />

and Practice. St. Louis: Mosby, 1993:1641–1647.<br />

2. Barkoff JR. Urticaria secondary to a copper intrauterine device. Int J Derm<br />

1976; 15:594–595.<br />

3. Barranco VP. Eczematous dermatitis caused by internal exposure to copper.<br />

Arch Dermatol 1972; 106:386–387.<br />

4. Romaguera C, Grimalt F. Contact dermatitis from a copper-containing intrauterine<br />

contraceptive device. Contact Dermatitis 1981; 7:163–164.


142 Hosty´nek and Maibach<br />

5. Menné T, Veien N, Sjolin K-E, Maibach HI. Systemic contact dermatitis. Am J<br />

Contact Dermatitis 1994; 5:1–12.<br />

6. Veien NK, Menné T, Maibach HI. Systemically induced allergic contact<br />

dermatitis. In: Menné T, Maibach HI, et al., eds. Exogenous Dermatoses:<br />

Environmental Dermatitis. Boca Raton: CRC Press, 1991:267–282.<br />

7. Flint GN. A metallurgical approach to metal contact dermatitis. Contact<br />

Dermatitis 1998; 39:213–221.<br />

8. Rubin RW, Penneys NS. Subpicogram analysis of sweat proteins using twodimensional<br />

polyacrylamide gel electrophoresis. Anal Biochem 1983; 131:520–524.<br />

9. Sens DA, Simmons MA, Spicer SS. The analysis of human sweat proteins by<br />

isoelectric focusing. I. Sweat collection utilizing the Macroduct system demonstrates<br />

the presence of previously unrecognized sex-related proteins. Pediatr<br />

Res 1985; 19:873–878.<br />

10. Marshall T. Analysis of human sweat proteins by two-dimensional electrophoresis<br />

and ultrasensitive silver staining. Anal Biochem 1984; 139:506–509.<br />

11. Elias P. Epidermal lipids, barrier function, and desquamation. J Invest Dermatol<br />

1983; 80:44s–49s.<br />

12. Lampe MA, Burlingame AL, Whitney J, et al. Human stratum corneum lipids:<br />

characterization and regional variations. J Lipid Res 1983; 24:120–130.<br />

13. Schurer NY, Elias PM. The biochemistry and function of stratum corneum<br />

lipids. Adv Lipid Res 1991; 24:27–56.<br />

14. Schmid R, Winkler J. Über die Kutane Kupferresorption aus einer Kupfer<br />

Enthaltenden Salbe. Klin Wochenschr 1938; 17:559–561.<br />

15. Odintsova NA. Permeability of human skin to potassium and copper ions and<br />

their ultrastructural localization. Chem Abs 1978; 89:360.<br />

16. Brown SA, Merritt K. Electrochemical corrosion in saline and serum. J Biomed<br />

Mater Res 1980; 14:173–179.<br />

17. Samitz MH, Katz SA. Nickel dermatitis hazards from prostheses. Br J Dermatol<br />

1975; 92:287–290.<br />

18. Cronin E. Metals. Contact dermatitis. Edinburgh: Churchill Livingstone,<br />

1980:328–335.<br />

19. Lisi P, Caraffini S, Assalve D. Irritation and sensitization potential of pesticides.<br />

Contact Dermatitis 1987; 17:212–218.<br />

20. Adams RM. Metals. Chromium. In: Adams RM, et al., eds. Occupational Skin<br />

Diseases. Philadelphia: Saunders, 1990:208–217.<br />

21. Magnusson B, Kligman AM. The identification of contact allergens by animal<br />

assay. The guinea pig maximization test. J Invest Dermatol 1969; 52:568–576.<br />

22. Kimber I. The murine local lymph node assay: principles and practice. Am J<br />

Contact Dermatitis 1993; 4:42–44.<br />

23. Boman A, Wahlberg JE, Hagelthorn G. Sensitizing potential of beryllium,<br />

copper and molybdenum compounds studied by the guinea pig maximization<br />

method. Contact Dermatitis 1979; 5:332–333.<br />

24. Karlberg AT, Boman A, Wahlberg JE. Copper—a rare sensitizer. Contact<br />

Dermatitis 1983; 9:134–139.<br />

25. Basketter DA, Roberts DW, Cronin M, Scholes EW. The value of the local<br />

lymph node assay in quantitative structure-activity investigations. Contact<br />

Dermatitis 1992; 27:137–142.


Copper Hypersensitivity 143<br />

26. Basketter DA, Gerberick GF, Kimber I, Loveless SE. The local lymph node<br />

assay: a viable alternative to currently accepted skin sensitization tests. Food<br />

Chem Toxicol 1996; 34:985–997.<br />

27. Ikarashi Y, Ohno K, Tsuchiya T, Nakamura A. Differences of draining lymph<br />

node cell proliferation among mice, rats and guinea pigs following exposure to<br />

metal allergens. Toxicology 1992; 76:283–292.<br />

28. Ikarashi Y, Tsuchiya T, Nakamura A. Detection of contact sensitivity of<br />

metal salts using the murine local lymph node assay. Toxicol Lett 1992;<br />

62:53–61.<br />

29. Haneke KE, Tice RR, Carson BL, Margolin BH, Stokes WS. ICCVAM evaluation<br />

of the murine local lymph node assay. Regul Toxicol Pharm 2001; 34:274–86.<br />

30. Amin S, Engasser P, Maibach HI. Sensitive skin: what is it? In: Baran R,<br />

Maibach HI, eds. Textbook of Cosmetic Dermatology. London: Martin Dunitz,<br />

Ltd., 1998:343–349.<br />

31. Wide L, Bennich H, Johansson SGO. Diagnosis of allergy by an in vitro test for<br />

allergen antibodies. Lancet 1967; 2:1105–1107.<br />

32. Halmepuro L, Lovenstein H. Immunological investigation of possible structural<br />

similarities between pollen antigens and antigens in apple, carrot and<br />

celery tuber. Allergy 1985; 40:264–272.<br />

33. Frykholm KO, Frithiof L, Fernström A ˚ IB, et al. Allergy to copper derived<br />

from dental alloys as a possible cause of oral lesions of lichen planus. Acta<br />

Derm Venereol (Stockh) 1969; 49:268–281.<br />

34. Cousins RJ. Absorption, transport, and hepatic metabolism of copper and zinc:<br />

special reference to metallothionein and ceruloplasmin. Physiol Rev 1985;<br />

65:238–309.<br />

35. Denko CW, Petricevic M, Whitehouse MW. Inflammation in relation to dietary<br />

intake of zinc and copper. Int J Tissue React 1981; 3:73–76.<br />

36. Hosty´nek JJ. Gold: an allergen of growing significance. Food Chem Toxicol<br />

1997; 35:839–844.<br />

37. Hosty´nek JJ. Metals. In: Amin S, Lahti A, Maibach HI, eds. Contact Urticaria<br />

Syndrome. Boca Raton, Florida: CRC Press LLC, 1997:189–211.<br />

38. Milanino R, Conforti A, Franco L, Marrella M, Velo G. Copper and<br />

inflammation—a possible rationale for the pharmacological manipulation of<br />

inflammatory disorders. Agents Actions 1985; 16:504–513.<br />

39. Powanda MC. Systemic alterations in metal metabolism during inflammation<br />

as part of an integrated response to inflammation. Agents Actions 1981(suppl 8):<br />

121–136.<br />

40. Lisi P, Brunelli L, Stingeni L. Co-sensitivity between cobalt and other transition<br />

metals. Contact Dermatitis 2003; 48:172–173.<br />

41. Uter W, Fuchs T, Häusser M, Ippen H. Patch test results with serial dilution of<br />

nickel sulfate (with and without detergent), palladium chloride, and nickel and<br />

palladium metal plates. Contact Dermatitis 1995; 32:135–142.<br />

42. Ale SI, Maibach HI. Operational definition of allergic contact dermatitis. In:<br />

Maibach HI, ed. Toxicology of Skin. Philadelphia: Taylor & Francis, 2001:<br />

345–356.<br />

43. Gaul LE. Metal sensitivity in eczema of the hands. Ann Allergy 1953; 11:<br />

758–762.


144 Hosty´nek and Maibach<br />

44. van der Meer DBJ, te Heerenveen H. Een geval van contactallergie voor koper<br />

en zink. Ned T Geneesk 1957; 101:2166–2167.<br />

45. Saltzer EI, Wilson JW. Allergic contact dermatitis due to copper. Arch Dermatol<br />

1968; 98:375–376.<br />

46. Trachtenberg DI. Allergic response to copper. J Periodontol 1972; 705–707.<br />

47. Shelley WB, Shelley ED, Ho AKS. Cholinergic urticaria: acetylcholinereceptor-dependent<br />

immediate-type hypersensitivity reaction to copper. Lancet<br />

1983; 1:843–846.<br />

48. Forck G. Allergie gegen Kupfer-T-Spirale. Allergologie 1984; 7:343.<br />

49. Romaguera C, Grimalt F, Vilaplana J. Contact dermatitis from nickel: an<br />

investigation of its sources. Contact Dermatitis 1988; 19:52–57.<br />

50. Laubstein B. Kupferallergie—eine seltene Sensibilisierung. Dermatol Monatsschr<br />

1990; 176:421–425.<br />

51. Bezzon OL. Allergic sensitivity to several base metals: a clinical report. J Prosthet<br />

Dent 1993; 69:243–244.<br />

52. Purello d’Ambrosio F, Ricciardi L, Isola S, et al. Systemic contact dermatitis to<br />

copper-containing IUD. Allergy 1996; 51:658–659.<br />

53. Wöhrl S, Hemmer W, Focke M, Götz M, Jarisch R. Copper allergy revisited.<br />

J Am Acad Dermatol 2001; 45:863–870.<br />

54. Boman A, Karlberg AT, Einarsson O, Wahlberg JE. Dissolving of copper by<br />

synthetic sweat. Contact Dermatitis 1983; 9:159–160.<br />

55. Sharata HH, Burnette RR. Effect of dipolar aprotic permeability enhancers on<br />

the basal stratum corneum. J Pharm Sci 1988; 77:27–32.<br />

56. Wahlberg JE, Skog E. The effect of dimethyl sulfoxide on the percutaneous<br />

absorption of mercuric chloride in the guinea pig. Acta Derm Venereol (Stockh)<br />

1967; 47:209–215.<br />

57. Fullerton A, Andersen JR, Hoelgaard A. Permeation of nickel through human<br />

skin in vitro—effect of vehicles. Br J Dermatol 1988; 118:509–516.<br />

58. Pirot F, Panisset F, Agache P, Humbert P. Simultaneous absorption of Cu and<br />

Zn through human skin in vitro. Skin Pharmacol 1996; 9:43–52.<br />

59. Pirot F, Millet J, Kalia YN, Humbert P. In vitro study of percutaneous absorption,<br />

cutaneous bioavailability and bioequivalence of zinc and copper from<br />

five topical formulations. Skin Pharmacol 1996; 9:259–269.<br />

60. Wahlberg JE. Vehicle role of petrolatum. Absorption studies with metallic test<br />

compounds in guinea pigs. Acta Derm Venereol (Stockh) 1971; 51:129–134.<br />

61. Wahlberg JE. Nickel: the search for alternative, optimal and non-irritant patch<br />

test preparations. Assessment based on laser Doppler flowmetry. Skin Res<br />

Technol 1996; 2:136–141.<br />

62. Ale SI, Maibach HI. Clinical relevance in allergic contact dermatitis. Derm<br />

Beruf Umwelt 1995; 43:119–121.<br />

63. de Groot AC. Patch testing concentrations and vehicles for testing contact<br />

allergens. In: Kanerva L, Elsner P, Wahlberg JE, Maibach HI, eds. Handbook<br />

of Occupational Dermatology. New York: Springer, 2000:1257–1276.<br />

64. Kanerva L, Estlander T, Jolanki R. Occupational skin allergy in the dental<br />

profession. Dermatol Clin 1994; 12:517–532.<br />

65. Veien NK. Systemically induced eczema in adults. Acta Dermato-Venereol<br />

(Stockh) 1989; 147(suppl):12–55.


Copper Hypersensitivity 145<br />

66. Nordlind K, Lidén S. Patch test reactions to metal salts in patients with oral<br />

mucosal lesions associated with amalgam restorations. Contact Dermatitis<br />

1992; 27:157–160.<br />

67. Hay IC, Ormerod AD. Severe oral and facial reaction to 6 metals in restorative<br />

dentistry. Contact Dermatitis 1998; 38:216.<br />

68. Stoffolani N, Damiani F, Lilli C, et al. Ion release from orthodontic appliances.<br />

J Dent 1999; 27:449–454.<br />

69. Jacobsen N, Hensten-Pettersen A. Occupational health problems and adverse<br />

patient reactions in orthodontics. Eur J Orthodont 1989; 11:254–260.<br />

70. Chantler E, Critoph F, Elstein M. Release of copper from copper-bearing<br />

intrauterine contraceptive devices. BMJ 1977; 6062:288–291.<br />

71. Zipper J, Medel M, Prager R. Suppression of fertility by intrauterine copper<br />

and zinc in rabbits: a new approach to intrauterine concepts. Am J Obstet<br />

Gynecol 1969; 105:529–534.<br />

72. Alvarez F, Brache E, Fernandez B, Guerrero B, Guiloff R, Hess R. New<br />

insights on the mode of action of intrauterine contraceptive devices in women.<br />

Fertil Steril 1988; 49:768–773.<br />

73. Sinigaglia F. The molecular basis of metal recognition by T cells. J Invest<br />

Dermatol 1994; 102:398–401.<br />

74. Förström L, Kiistala R, Tarvainen K. Hypersensitivity to copper verified by<br />

test with 0.1% CuSO4. Contact Dermatitis 1977; 3:280–281.<br />

75. van Joost TH, van Ulsen J, van Loon LA. Contact allergy to denture materials<br />

in the burning mouth syndrome. Contact Dermatitis 1988; 18:97–99.<br />

76. Sterry W, Schmoll M. Contact urticaria and dermatitis from self-adhesive pads.<br />

Contact Dermatitis 1985; 13:284–285.<br />

77. Reid DJ. Allergic reaction to copper cement. Br Dent J 1968; 124:92.<br />

78. Popa V, Teculescu D, Stanescu D, Gavrilescu N. Bronchial asthma and asthmatic<br />

bronchitis determined by simple chemicals. Dis Chest 1969; 56:395–404.<br />

79. Beckmann M, Wagner H. Allergische Reaktionen bei kupferhaltigen Intrauterinpessaren.<br />

Medizinische Welt 1979; 30:1855–1856.<br />

80. Sabbah A, Drouet M, Hergon E. Rhinite par sensibilite au cuivre. Allergie et<br />

Immunologie 1983; 15:209–210.<br />

81. Gaul LE. Incidence of sensitivity to chromium, nickel, gold, silver, and copper<br />

compared to reactions to their aqueous salts including cobalt sulfate. Ann<br />

Allergy 1954; 12:429–444.<br />

82. Epstein S. Cross-sensitivity between nickel and copper. J Invest Dermatol 1955;<br />

55:269–274.<br />

83. Bockendahl H, Remy W, Masuch E. Untersuchungen zum Mechanismus des<br />

Kontaktekzems gegen Kupfer. Archiv fuer Dermatologische Forschung 1974;<br />

250:167–171.<br />

84. Dhir GG, Rao DS, Mehrotra MP. Contact dermatitis caused by copper<br />

sulfate used as coloring material in commercial alcohol. Ann Allergy 1977;<br />

39:204.<br />

85. Walton S. Investigation into patch testing with copper sulfate. Contact Dermatitis<br />

1983; 9:89–90.<br />

86. van Joost T, Habets JMW, Stolz E, Naafs B. The meaning of positive patch tests<br />

to copper sulphate in nickel allergy. Contact Dermatitis 1988; 18:101–102.


146 Hosty´nek and Maibach<br />

87. Hackel H, Miller K, Elsner P, Burg G. Unusual combined sensitization to<br />

palladium and other metals. Contact Dermatitis 1991; 24:131–132.<br />

88. Kawahara D, Oshima H, Kosugi H, Nakamura M, Sugai T, Tamaki T. Further<br />

epidemiologic study of occupational contact dermatitis in the dental clinic.<br />

Contact Dermatitis 1993; 28:114–115.<br />

89. Motolese A, Truzzi M, Giannini A, Seidenari S. Contact dermatitis and contact<br />

sensitization among enamellers and decorators in the ceramics industry. Contact<br />

Dermatitis 1993; 28:59–62.<br />

90. Giorgini S, Brusi C, Francalanci S, Acciai MC, Sertoli A. Prodotti alternativi e<br />

prevenzione della dermatite allergica da contatto. II. Prevenzione della dermatite<br />

allergica da contatto da orecchini di bigiotteria. Annali Italiani di<br />

Dermatologia Clinica e Sperimentale 1994; 46:151–158.<br />

91. Raccagni AA, Baldari U, Righini MG. Airborne dermatitis in a painter. Contact<br />

Dermatitis 1996; 35:119–120.<br />

92. Rademaker M. Occupational contact dermatitis among New Zealand farmers.<br />

Australasian J Dermatol 1998; 39:164–167.<br />

93. Vilaplana J, Romaguera C. Contact dermatitis and adverse oral mucous membrane<br />

reactions related to the use of dental prostheses. Contact Dermatitis<br />

2000; 43:183–185.<br />

94. Dry J, Leynadier F, Bennani A, Piquet P, Salat J. Intrauterine copper contraceptive<br />

devices and allergy to a copper and nickel. Ann Allergy 1978; 41:1978.<br />

95. Rongioletti F, Rivara G, Rebora A. Contact dermatitis to a copper-containing<br />

intra-uterine device. Contact Dermatitis 1985; 13:343.<br />

96. Hausen BM, Hohlbaum W. Verursachen Kupfer-Intrauterinpessare eine<br />

Kontaktallergie? Dtsch Med Wochenschr 1986; 111:1016–1021.<br />

97. Lauter H. Messingallergie? Allergologie 1987; 10:156–157.<br />

98. Siliotti F, Caroti S, Caroti A, Alborino F. Considerazioni su di un caso di allergia<br />

a IUD medicato al rame. Ginecological Clinics 1987; 8:197–200.<br />

99. Namikoshi T, Yoshimatsu T, Suga K, Fujii H, Yasuda K. The prevalence of<br />

sensitivity to constituents of dental alloys. J Oral Rehabil 1990; 17:377–381.<br />

100. Hocher B, Keller F, Krause PH, Gollnick H, Oelkers W. Interstitial nephritis<br />

with reversible renal failure due to a copper-containing intrauterine contraceptive<br />

device. Nephron 1992; 61:111–113.<br />

101. Vilaplana J, Romaguera C, Cornellana F. Contact dermatitis and adverse oral<br />

mucous membrane reactions related to the use of dental prostheses. Contact<br />

Dermatitis 1994; 30:80–84.<br />

102. Kansu G, Aydin AK. Evaluation of the biocompatibility of various dental alloys:<br />

part 1—toxic potentials. Eur J Prosthodont Restorative Dent 1996; 4:129–136.<br />

103. Kansu G, Aydin AK. Evaluation of the biocompatibility of various dental<br />

alloys: part 2—allergenical potentials. Eur J Prosthodont Restorative Dent<br />

1996; 4:155–161.<br />

104. Fedorov SM, Ado VA, Mokronosova MA, Seliskii GD, Perlamutrov YN,<br />

Samuilova TL. Allergic dermatitis due to metal sensitizers contained in jewellery<br />

and intrauterine devices. Vestnik Dermatologie i Venereologie 1997; 1:49–50.<br />

105. Pujol RM, Randazzo L, Miralles J, Lomar A. Perimenstrual dermatitis secondary<br />

to a copper-containing intrauterine contraceptive device. Contact<br />

Dermatitis 1998; 38:288.


Copper Hypersensitivity 147<br />

106. Santosh V, Ranjith K, Shrutakirthi DS, Sachin V, Balachandran C. Results of<br />

patch testing with dental materials. Contact Dermatitis 1999; 40:50–51.<br />

107. Hannuksela M, Salo H. The repeated open application test (ROAT). Contact<br />

Dermatitis 1986; 14:221–227.<br />

108. Nakada T, Hosty´nek JJ, Maibach HI. Use tests: ROAT (repeated open application<br />

test)/PUT (provocative use test): an overview. Contact Dermatitis 2000;<br />

43:1–3.<br />

109. Wilkinson SM, Heagerty AHM, English JSC. A prospective study into the<br />

value of patch and intradermal tests in identifying topical corticosteroid allergy.<br />

Br J Dermatol 1992; 127:22–25.<br />

110. Herbst R, Lauerma A, Maibach HI. Intradermal testing in the diagnosis of<br />

allergic contact dermatitis—a reappraisal. Contact Dermatitis 1993; 29:1–5.<br />

111. Lahti A, Maibach HI. Contact urticaria syndrome. In: Moschella SL, Hurley HJ,<br />

eds. Dermatology. 3rd eds. Philadelphia: WB Saunders Co., Harcourt Brace<br />

Jovanovich, Inc., 1992:433–440.<br />

112. Hannuksela M, Lahti A. Contact urticaria from foods. In: Roe D, ed. Nutrition<br />

and the Skin. New York: Alan R.: Riss, 1986.<br />

113. Joules H. Asthma from sensitisation to chromium. Lancet 1932; 2:182–183.<br />

114. Zabel M, Lindscheid KR, Mark H. Kupfersulfatallergie unter besonderer<br />

Berucksichtigung der internen Exposition. Z Hautkr 1990; 65:481–486.<br />

115. Tschernitschek H, Wolter S, Korner M. Allergien auf Zahnersatzmaterialien.<br />

Dermatosen 1998; 46:244–248.<br />

116. Candura SM, Verni P, Dellabianca A, et al. Sensibilizzazione epicutanea a<br />

metalli e dermatite allergica da contatto: analisi di una casistica ambulatoriale.<br />

Giornale Italiano di Medicina del Lavoro ed Ergonometria 1999; 21:40–45.<br />

117. Moulon C, Vollmer J, Weltzien HU. Characterization of processing requirements<br />

and metal cross-reactivities in T cell clones from patients with allergic<br />

contact dermatitis to nickel. Eur J Immunol 1995; 25:3308–3315.<br />

118. Pistoor FHM, Kapsenberg ML, Bos JD, Meinardi MMHM, von Blomberg BME,<br />

Scheper RJ. Cross-reactivity of human nickel-reactive T-lymphocyte clones with<br />

copper and palladium. J Invest Dermatol 1995; 105:92–95.<br />

119. Jerschow E, Hosty´nek JJ, Maibach HI. Allergic contact dermatitis elicitation<br />

thresholds of potent allergens in humans. Food Chem Toxicol 2001; 39:1095–1108.<br />

120. Mitchell JC. Egregious blunder of maximization by the angry back and a note<br />

on unconfirmed ergodata. Contact Dermatitis 1996; 35:131–132.<br />

121. Mitchell J, Maibach HI. Managing the excited skin syndrome: patch testing<br />

hyperirritable skin. Contact Dermatitis 1997; 37:193–199.<br />

122. van der Burg CKH, Bruynzeel DP, Vreeburg KJJ, von Blomberg BME,<br />

Scheper RJ. Hand eczema in hairdressers and nurses: a prospective study.<br />

Contact Dermatitis 1986; 14:275–279.<br />

123. Benezra C, Sigman CC, Perry LR, Helmes CT, Maibach HI. A systematic<br />

search for structure–activity relationships of skin contact sensitizers. I.<br />

Methodology. J Invest Dermatol 1985; 85:351–356.<br />

124. Jouppila P, Niinimäki A, Mikkonen M. Copper allergy and copper IUD.<br />

Contraception 1979; 19:631–637.<br />

125. Aro T, Kanerva L, Hayrinenimmonen R, Silvennoinenkassinen S. Long-lasting<br />

allergic patch test reaction caused by gold. Contact Dermatitis 1993; 28:276–281.


148 Hosty´nek and Maibach<br />

126. Möller H, Larsson A ˚ , Björkner B, Bruze M. The histological and immunohistochemical<br />

pattern of positive patch test reactions to gold sodium thiosulfate.<br />

Acta Derm Venereol (Stockh) 1994; 74:417–423.<br />

127. Samitz MH, Katz SA. Nickel-epidermal interactions: diffusion and binding.<br />

Environ Res 1976; 11:34–39.<br />

128. Dupuis D, Rougier A, Roguet R, Lotte C, Kalopissis G. In vivo relationship<br />

between hotny layer reservoir effect and percutaneous absorption in human<br />

and rat. J Invest Dermatol 1984; 82:353–356.<br />

129. Alder JF, Batoreu MCC, Pearse AD, Marks R. Depth concentration profiles<br />

obtained by carbon furnace atomic absorption spectrometry for nickel and<br />

aluminium in human skin. J Anal Atom Spectrom 1986; 1:365–367.<br />

130. Fullerton A, Hoelgaard A. Binding of nickel to human epidermis in vitro. Br J<br />

Dermatol 1988; 119:675–682.<br />

131. Santucci B, Cannistraci C, Cristaudo A, Camera E, Picardo M. The influence<br />

exerted by cutaneous ligands in subjects reacting to nickel sulfate alone and<br />

in those reacting to more transition metals. Exp Dermatol 1998; 7:162–167.<br />

132. Frentz G, Teilum D. Cutaneous eruptions and intrauterine contraceptive copper<br />

device. Acta Derm Venereol (Stockh) 1980; 60:69–71.


8<br />

Copper in Medicine and Personal<br />

Care: A Historical Overview<br />

Roberto Milanino<br />

Facoltà di Medicina e Chirurgia, Sezione di Farmacologia,<br />

Dipartimento di Medicina e Salute Pubblica, Università di Verona,<br />

Verona, Italy<br />

Remember always that some ideas that seem dead and buried may at<br />

one time or another rise up to life again, more vital than ever before.<br />

—Louis Pasteur<br />

INTRODUCTION<br />

In the pre- and protohistoric ages, medicine and therapy were both strongly<br />

linked with religion and thus were mainly presented to communities and<br />

patients in magic-religious clothing, although empiricism actually lay at<br />

the core of these arts.<br />

Medicinal plants were widely employed as drugs, and, in some cases,<br />

their active principles are really valuable and still used clinically today, e.g.,<br />

atropine (Atropa belladonna, Datura stramonium—L.), digitalin (Digitalis<br />

purpurea—L.), opium and derivatives (Papever somniferum—L.), quinine<br />

(Chincona offinicinalis—L.), reserpine (Rauwolfia serpentina—L.), salicin<br />

and derivatives (Salix alba—L.), tubocurarine (Chondrodendon tomentosum—<br />

R. & P.), etc. Drugs from the animal kingdom were numerous, such as the<br />

donkey fat, rhinoceros horns, and pig liver; the latter, for instance, was<br />

149


150 Milanino<br />

considered a good remedy for anemia (an empiric indication consistent with<br />

our modern medical knowledge). Minerals, for example those containing<br />

antimony, arsenic, copper, gold, iron, lead, mercury, sulfur, or zinc, were<br />

also used frequently. Many of the above ingredients were routinely mixed<br />

according to magic-empirical criteria in recipes, often kept secret, and usually<br />

administered accompanied by complex esoteric rituals.<br />

The aim of this review, however, is focused on the use of copper in therapy<br />

during the prescientific millennia preceding our modern medical culture.<br />

Humans have employed copper in medicine since before 3500 B.C., mostly<br />

referring to it as a ‘‘generic drug,’’ but also recognizing its antiseptic as well<br />

as anti-inflammatory potentials. In particular, attributing anti-inflammatory<br />

properties to copper appears to have been an extremely clever intuition. What<br />

is even more surprising is that this metallo-element was believed to be endowed<br />

with therapeutic properties by almost all major ancient cultures. In particular,<br />

while its diffusion among Mediterranean, near-, middle-, and far-Eastern civilizations<br />

may be explained on the basis of their frequent mutual contacts, the<br />

presence of copper in the ‘‘pharmacopoeia’’ of the pre-Columbian Meso- and<br />

South-American cultures seems to derive entirely from the autochthonous<br />

experience of these populations.<br />

THE SUMERIC CULTURE: CIRCA 4000–2300 B.C.<br />

Present day knowledge ascribes to this culture the basic merit of having invented<br />

the near- and middle-Eastern ideographic writing, which is conventionally considered<br />

as the beginning of the historical age. This society developed recurrent<br />

commercial and cultural relations with the neighboring areas and cultures (1).<br />

Malachite (basic cupric carbonate) was one of the products that<br />

Sumerians frequently exported to Egypt as well as closer regions, mainly<br />

for making jewels and cosmetic products, and also for pharmaceutical preparations.<br />

It is documented, through indirect evidence (mostly Egyptians),<br />

that the utilization of pulverized malachite for generic medical purposes<br />

was a practice commonly used by the Sumerians themselves (2).<br />

THE ANCIENT EGYPTIAN CULTURE<br />

The Predynastic Age to the II Intermediate Period (XVII Dynasty):<br />

Circa 3900–1550 B.C. (3,4)<br />

Probably the first significant medical document known today is the Kahun<br />

papyrus, which was most likely written during the kingdom of Seosostris II<br />

(about 1880 B.C., XII dynasty), and is essentially focused on gynecological<br />

problems (4,5). The Kahun papyrus appears to make no specific mention of<br />

the use of copper as a drug.<br />

However, we know from other minor written fragments of those times,<br />

as well as numerous and much more exhaustive subsequent medical


Copper in Medicine and Personal Care 151<br />

manuscripts, that the utilization of pulverized malachite was extremely common,<br />

since the beginning of the predynastic period, for preparing a typical<br />

blue-green eye makeup (5).<br />

In the middle and late period of this Egyptian age, this cosmetic<br />

became dark gray, as shown by several tomb pictures, probably because<br />

pulverized galena (lead sulfide) was already added to malachite (2,6). This<br />

eye makeup was used not only for decorative ritual purposes exclusively<br />

related to the Osiris cult but also for the prevention and cure of eye infections,<br />

widespread in the ancient as well as modern Egypt. These infections<br />

were mainly caused by the dryness of the climate and sandy winds characterizing<br />

the area (2,5–7). Actually, in this practice, it may be possible to<br />

recognize the first empiric attribution of antiseptic properties to copper<br />

preparations.<br />

The XVIII Dynasty to the Ptolemaic Dynasty: Circa<br />

1550 B.C. to 30 A.D.<br />

The most famous and comprehensive documentation of Egyptian medical<br />

science are the Ebers’ and Smith’s papyri. They summarize medical information<br />

of those times based on, if not mainly, previous orally transmitted and<br />

written traditions. Both these treatises belong to the XVIII dynasty, the first<br />

of the New Kingdom (1550–1075 B.C.) (5,7).<br />

The papyrus of Ebers (circa 1500 B.C.) is a true medical encyclopedia,<br />

dealing with both generic medical and therapeutic problems. The pharmacological<br />

sections of the Ebers’ papyrus emphasize the importance of<br />

personal cleanliness, and reports the use of many drugs mainly coming from<br />

medicinal plants (castor oil, senna, tamarisk, thyme, etc.), but minerals are<br />

also not neglected. For example, this text clearly codifies, for the first time,<br />

the preparation of kohl, i.e., the mixture of malachite and galena. This compound<br />

is certainly identical to that used in previous centuries, and is still<br />

routinely applied as an eye cosmetic, for the same decorative–ritual and<br />

practical medical purposes as described earlier (2,5,6).<br />

The papyrus of Smith (about 1450 B.C.) mainly addresses surgical problems<br />

as Egyptian society (like the later Roman one) was frequently involved<br />

in wars and, consequently, was forced to develop efficient emergency<br />

medical procedures (5). This papyrus also mentions the use of pulverized<br />

malachite thought to be endowed with astringent, healing, and antiseptic<br />

properties for the treatment of postoperative wounds (2,5). Interestingly,<br />

these antiseptic and healing potentials of copper are presently fully recognized<br />

(8), although no longer routinely employed in modern medical practice.<br />

However, copper preparations, in particular cupric sulfate and basic cupric<br />

sulfate (9), are still widely used in agriculture; copper is also a potent antimycotic<br />

agent, especially effective in the treatments of plants, such as<br />

tomatoes and grapes, against fungal infections.


152 Milanino<br />

THE BABYLONIAN–ASSYRIAN CULTURE: CIRCA 1750–539 B.C.<br />

The medical and pharmacological knowledge of these societies is reported<br />

on approximately 800 medical clay tablets recovered from circa 100,000<br />

found during the excavation of the Assyrian Library of Nineveh<br />

(about 650 B.C.) (2,10). Babylonian–Assyrian medicine particularly focused<br />

on studying the liver as a basic tool of its diagnostic. This organ, however,<br />

was not examined according to modern semeiotic procedures (directly on<br />

patient), but instead physicians examined the livers as well as the viscera of<br />

animals sacrificed during the propitiatory rituals performed in order to recognize<br />

the disease, then chose the appropriate therapy (2). This practice was<br />

clearly an esoteric superstition called Haruspecism. Most likely, Haruspecism<br />

reached its highest levels over 900 years later with the development of the<br />

Etrurian culture in Italy (11). In fact, the Etrurian haruspices not only ‘‘specialized’’<br />

in this kind of ‘‘medical approach’’ but also transformed it into a more<br />

general divinatory procedure, which included the magically inspired interpretation<br />

of many other natural phenomena, such as the flight of birds (11).<br />

About 250 drugs of plant origin, and also 120 compounds derived from<br />

minerals, are cited in the Babylonian–Assyrian pharmacopoeia. Among the<br />

minerals, those containing copper are mentioned specifically, being qualified<br />

as generic therapeutic remedies (2). However, most likely for the first time,<br />

the use of a copper bracelet is quoted in these reports as useful, albeit nonspecific,<br />

pharmacological tool (10).<br />

THE ANCIENT INDIAN CULTURE: CIRCA 2800–1000 B.C.<br />

The medical knowledge acquired since the remote origins of the ancient<br />

Indian culture and orally transmitted generation to generation was later<br />

summarized in two major medical books: the Samhita Charaka and the<br />

Susruta Samhita (written in Sanskrit during the Brahamanic age, approximately<br />

between 800 B.C. and 1000 A.D.) (1,2). Surgery was highly developed<br />

in the ancient Indian culture and, unsurprisingly, their pharmacopoeia made<br />

a wide use of medicinal plants, among which is mentioned Cannabis indica<br />

L. as an analgesic (cannabinoid derivatives). This drug was frequently<br />

employed, together with black henbane (Hyoscyamus niger—L.), which contains<br />

such analgesic–narcotic principles as hyoscyamine and scopolamine, as<br />

a general anesthetic in surgical operations (2).<br />

Although no other significant information is available, the use of<br />

copper (as sulfide or sulfate) for making preparations used for nonspecific<br />

medical purposes is reported in these texts (2).<br />

THE ANCIENT CHINESE CULTURE: CIRCA 3000 B.C. TO 1100 A.D.<br />

According to legend, the medical book Nei Ching (‘‘The Canon of Medicine’’)<br />

was written by the mythical king Huang Ti (the ‘‘Yellow Emperor’’) about<br />

2700 years B.C. (2). Actually, this treatise was most likely edited much later


Copper in Medicine and Personal Care 153<br />

and, moreover, appeared in two separate parts, about 10 centuries apart.<br />

In fact, the first part dates back to approximately the second century B.C., the<br />

second part to about the eighth century A.D. However, ‘‘The Canon of<br />

Medicine’’ is the first-known compendium in which millennia of Chinese<br />

medical and therapeutic traditions are summarized (2).<br />

The pharmacological use of copper sulfate (or sulfide) is undeniably<br />

documented in the above text, not only for the topical treatment of skin<br />

and eye diseases but also for ‘‘blood purification.’’ The latter is, probably,<br />

the first-known indication for oral administration of copper (2).<br />

THE PRE-COLUMBIAN MESO- AND SOUTH-AMERICAN<br />

CULTURES: CIRCA 600 B.C. TO 1500 A.D.<br />

The medical traditions of the better-known Meso- and South-American civilizations<br />

(i.e., Maya, Aztec, and Inca) are referred to in numerous documents,<br />

albeit those written exclusively by the Spanish invaders (2).<br />

In particular, the cranial trepanation procedure deserves an especial<br />

attention. Although known and occasionally practiced since at least the fifth<br />

to fourth millennium B.C. among most of the world’s prehistoric and historic<br />

cultures, including that of Egypt, this kind of surgery was chiefly practiced<br />

by Meso- and South-American societies between the fifth century B.C. and<br />

the fifth century A.D. (12,13). Craniotomy, however, reached an astonishing<br />

level of specialization among the Inca ‘‘surgeons,’’ who frequently made use<br />

of this peculiar technique and whose patients actually had an extraordinary<br />

survival rate, assessed far beyond 50%. In fact, archeologists discovered<br />

skeletons of a great number of Incas whose skulls underwent this procedure,<br />

and who survived the distressing experience (12). Interestingly, a human<br />

cranium found in the necropolis of Cuzco (Peru) clearly shows the holes<br />

resulting from two successive cranial trepanations; obvious signs of bone<br />

regeneration surrounding these holes testify to the individual’s long survival<br />

following the surgical procedures (12). The main purpose of this intervention<br />

was undoubtedly a ritual–magical one (2,12,13); nonetheless, the<br />

craniotomy also bears real clinical value in reducing intracranial pressure<br />

due to post-traumatic meningeal edemas or hemorrhages (10).<br />

Notably, ‘‘gauze’’ soaked in a copper sulfate solution was routinely used<br />

to ‘‘disinfect’’ the surgical wounds after the removal of the skull section (2,13).<br />

THE ANCIENT GREEK CULTURE<br />

It is relevant to note that the chronological classification of Ancient Greek<br />

society and culture is more conventional than factual. For instance, the<br />

dating of Archaic Greek, of the classic Greek, and, especially, of the official<br />

end of this unique civilization (which is made to coincide whit the Roman<br />

conquest, i.e., 323 B.C.) is merely symbolic. In fact, as we will see below,


154 Milanino<br />

the Greek culture developed and survived much longer, and is still largely<br />

present in the roots of our contemporary occidental civilization.<br />

Archaic Greek: Circa 1300–500 B.C.<br />

Although the ancient Greeks attributed the beginnings of their medicine to<br />

the probably mythical Thessalonian prince Asclepios (about 1200 B.C.), earlier<br />

documented Greek medical records came down from the Mediterranean-<br />

Greek colonies, especially the Sicilian one (11,14).<br />

Actually, significant medical theories were derived from observations<br />

of ancient Greek scholars. For instance, Alcmeon (circa 560 B.C.) postulated<br />

that the brain was the center of human sensorial and intellectual life, and<br />

Empedocles (an Alcmeon’s contemporary) speculated that respiration occurred<br />

both at the pulmonary and skin pore levels (11).<br />

It was probably in those times that the use of copper preparations,<br />

most likely borrowed from the Mesopotamian, Egyptian, and Minoan medical<br />

traditions, was introduced as a nonspecific remedy (11,14).<br />

Classic Greek: Circa 500–323 B.C.<br />

About 100 years after Alcmeon’s and Empedocles’ age, when classic Greek<br />

culture developed under the influence of the thoughts of the great philosophers<br />

(such as Socrates and Plato), Hippocrates (460 B.C.) was probably the<br />

first physician to invoke the separation of medicine, religion, and myth, stating,<br />

‘‘No disease is more divine or more human than any other, since every<br />

illness is due to a natural cause, in the absence of which it cannot take place’’<br />

(11). The Hippocratic perspective on human brain functions was also very<br />

modern; redefining the previous hypothesis of Alcmeon, Hippocrates wrote,<br />

‘‘I say that the brain is the most powerful organ of the human body ...The<br />

eyes, ears, tongue, hands and feet all act under the control of the brain.<br />

The brain transmits its messages to the human conscience’’ (11,14). Nevertheless,<br />

this does not necessarily mean that Hippocrates’ attitude had been a sort<br />

of ‘‘revolutionary’’ one. In fact, Hippocrates was especially careful not to<br />

express open dispute with the contemporary religious opinions and traditions,<br />

and in the prologue to his famous ‘‘Hippocrates’ swearing,’’ Hippocrates<br />

wrote, ‘‘I swear in the name of Asclepios, the Physician, Igea, Panacea, and<br />

all the gods and goddesses, calling them to witness, that I will follow ...’’ (11).<br />

Hippocrates’ major work is the Corpus Hippocraticum, which consists<br />

of 72 volumes, of which only 17 are sure to have been written by Hippocrates<br />

himself. The other 55 are ‘‘contaminations of ’’ and ‘‘additions to’’<br />

the original texts by numerous respectable physicians (but also by some<br />

quack doctors) in the course of following centuries (11,14).<br />

In Hippocrates’ original books, the use of copper preparations was<br />

clearly prescribed for the therapy of many such diseases as cutaneous and<br />

eye diseases, vaginal disorders (using an irrigation with copper solutions or suspensions),<br />

and hemorrhoids (using ‘‘suppositories’’ containing copper) (11,14).


Copper in Medicine and Personal Care 155<br />

Finally, ancient Greek medicine was the first to suggest the wearing of<br />

the copper bracelet as an antiarthritic remedy, which brings us directly to<br />

our present debate on the effectiveness of topical copper preparations in<br />

the treatment of rheumatic conditions (14,15).<br />

THE ANCIENT ROMAN CULTURE: CIRCA 600 B.C. TO 476 A.D.<br />

From the beginning, ancient Romans were primarily engaged in bringing<br />

their ‘‘Pax Romana’’ (the archetype of our contemporary globalization process)<br />

to all societies living in the known world. Thus, the Roman age based<br />

both the origin and development of its knowledge, the medical one included,<br />

on Greek thought and tradition, although initially some Etrurian and later<br />

Arab contributions were also present (11). Therefore, in spite of the much<br />

older origin of Roman society and culture, the major works describing<br />

Roman medicine and pharmacology were all written between the end of<br />

the first century B.C. and the second century A.D., and are attributable to<br />

three prominent names, i.e., Pliny ‘‘the elder,’’ Celsus, and Galen (11).<br />

Caius Pliny ‘‘the elder’’ (23–79 A.D.) wrote the 37 volumes of Naturalis<br />

Historia, in which Pliny discourses not only upon the subjects of astronomy,<br />

geography, zoology, and botany, but also on medicine, physiology, and<br />

pharmacology. Pliny’s pharmacopoeia, along with medicinal plant and<br />

animal derivatives, devotes significant attention to minerals, copper in particular.<br />

This metal is frequently mentioned in many preparations in which it is<br />

present in the form of pharmacologically active compounds such as copper<br />

oxides, copper sulfide, copper sulfate, basic cupric carbonate, and basic cupric<br />

acetate (C.P.E., Nat. Hist. XXX and XXXI) (16). Their practical use was<br />

particularly recommended for the treatment of eye and skin diseases, tonsillitis,<br />

throat inflammation, etc. (C.P.E., Nat. Hist. XXX and XXXI) (16).<br />

Aulus Cornelius Celsus (probably, 30 B.C. to 38 A.D.) was the author of<br />

De Medicina, an eight-volume encyclopedia, in the third tome of which,<br />

treating on ‘‘the fevers,’’ Celsus codified his famous ‘‘four cardinal signs<br />

of inflammation,’’ rubor, tumor, calor et dolor, to which a fifth sign, the fuctio<br />

laesa, was then added by Galeno (17,18).<br />

It was again Celsus who, probably first, formally recognized copper as<br />

useful antiseptic and, especially, anti-inflammatory agent; notably, the antiinflammatory<br />

potential of basic cupric carbonate (i.e., malachite) has been<br />

confirmed by our contemporary research (19). Copper compounds were<br />

indeed frequently employed in Celsus’ very elaborate prescriptions such as<br />

those suggested for the treatment of anal rhagades, hemorrhoids, tonsillitis,<br />

wound disinfection, etc. We like to report as an example, Celsus’ recipe for<br />

curing anal rhagades:<br />

Verdigris (basic cupric acetate): parts 2<br />

Myrrh: parts 12<br />

Antimony: parts 16


156 Milanino<br />

Poppy ‘‘tears’’: parts 16<br />

Acacia: parts 16<br />

To be suspended in wine before local application (17)<br />

In the writings of Celsus, a great deal of attention is also dedicated<br />

to surgery. In fact, Celsus first introduced in orthopedics the application<br />

of copper plates (which was thought to be appropriate for those conditions,<br />

since they are also antiseptic and anti-inflammatory) to promote the recovery<br />

of disassembled fractures (17). Interestingly, this practice was still in use<br />

more than 1400 years later, as exemplified by a human skeleton recently<br />

discovered in Belgium (cathedral of Vrasene) (17).<br />

Claudius Galeno (129–200 A.D.) wrote many medical books such as the<br />

Ars magna and the Ars parva, as well as works of pharmacology collected in<br />

the treatises De Methodus Medendi and De simplicium medicamentorum<br />

temperamentis et facultatibus (11). Galeno, a Civis romanus (although fully<br />

Greek by origin, culture, and thought), was an eminent and innovative<br />

anatomist, physiologist, and pathologist, but a less pioneering therapist<br />

(18). In fact, while Galeno’s ‘‘general medicine’’ writings describe some sort<br />

of interesting experimental work, Galeno’s pharmacology books are mainly<br />

a summary of therapeutic experiences gathered by the ancient Mediterranean<br />

and Eastern cultures (the Greek one in particular), skillfully mixed<br />

with the pharmacological reports of Celsus as well as Pliny ‘‘the elder’’ (18).<br />

Thus, copper was also frequently used in Galeno’s therapeutics, but<br />

still on the basis of an empirical methodology, using essentially the same<br />

copper preparations, and being addressed to the same pharmacological targets<br />

as in the previous Mesopotamian, Egyptian, Chinese, Indian, Greek,<br />

and, of course, Pliny’s and Celsus’ heritages. Galeno’s pharmacopoeia is,<br />

therefore, devoid of any original ‘‘scientific’’ approach, perhaps with one<br />

exception: occasionally, Galeno’s ‘‘new’’ drug mixtures were tried to test<br />

in self-experimentation (11,17,18).<br />

Nevertheless, Galeno’s works were destined to play a significant role in<br />

the future trends of medical and pharmacological scientific ‘‘development’’<br />

over the following 1500 years, especially in our European ‘‘world,’’ and also<br />

in the Arabian one (18).<br />

FROM THE HIGH-MEDIEVAL AGE TO THE EARLY<br />

20TH CENTURY<br />

The classic Greek and Roman ‘‘biomedical’’ background deeply affected<br />

biological, medical, and pharmacological thought at least up until the<br />

‘‘Galilean revolution,’’ which generated a totally new way of seeing and<br />

doing science (second half of 16th, first half of 17th centuries) (11,18,20).<br />

Thus, different fragments of Hippocrates’, Pliny’s, Celsus’, and Galeno’s<br />

writings were blended according to current ideas and prejudices, mainly in


Copper in Medicine and Personal Care 157<br />

consequence of the dogmatic and actually prevailing Catholic culture, which<br />

found its major justification in Scholastic philosophy (a re-reading of<br />

Aristotelian thought), as elaborated by Thomas Aquinas (13th century A.D.)<br />

(20,21). Notably, in this discouraging context, remarkable attention was dedicated<br />

to Galeno’s works, since Galeno possibly believed, and certainly stated,<br />

‘‘the human body is merely an instrument of man’s soul’’ (11,18,20).<br />

Nevertheless, the pharmacological use of copper survived during those<br />

dark centuries, although essentially based more on the imaginative superstitions<br />

derived by the ‘‘innovative discoveries’’ of contemporary quack<br />

doctors and alchemists, than on the ‘‘good old’’ empiricism that characterized<br />

the progress in medicine and pharmacology made by scholars of the<br />

ancient cultures, particularly classic Greek–Roman.<br />

Later on, the ‘‘Galilean revolution’’ also began to strongly influence<br />

the world of biology and medicine and the related biomedical disciplines<br />

slowly evolved into our modern experimental development process of scientific<br />

learning. Pharmacology, and especially copper, however, were destined<br />

to wait still a while longer to see their ‘‘renaissance.’’<br />

In fact, focusing on copper, it was only in the first half of the 19th century<br />

that Rademacher, introducing the so-called ‘‘medicine of experience,’’<br />

deduced, after a clinical trial lasting 25 years, ‘‘the body has a strange predisposition<br />

for a great number of diseases which disappeared, or could be<br />

healed, in the presence of copper. Healthy subject did not show any response<br />

to copper’’ (18,22). At the end of the 19th century, copper was administered<br />

internally and was found to be effective against numerous illnesses such as<br />

skin diseases and the infections of tuberculosis and syphilis (22). The roots<br />

of these clinical uses of copper, however, were again to be found in a sort of<br />

‘‘renewed empiricism,’’ which, according to Classic Age traditions, considered<br />

copper a valuable antiseptic and anti-inflammatory agent.<br />

Between the end of the 19th and the beginning of the 20th centuries,<br />

mounting consideration for the discoveries of modern microbiology, physiology,<br />

and biochemistry, as well as the appearance of chemistry as science<br />

in the field of pharmacology (e.g., the synthesis of arsenic derivatives, salicylates,<br />

sulfanilamides, etc.) (18,22), led to the abandonment of the use of<br />

copper in medicine. It is true that during the 1940s some copper complexes,<br />

such as Cupralene and Dicuprene, were still employed for treating arthritic<br />

diseases, but the advent of the therapy with exogenous cortisone extracts or<br />

derivatives rapidly led also to their disuse.<br />

BEGINNING OF THE SCIENTIFIC AGE FOR COPPER: 1928–1976<br />

Curiously, it was only in the first half of the past century, while copper therapy<br />

was gradually being neglected by a majority of clinicians, that the<br />

importance of copper in biology and medicine finally found its experimental<br />

justification.


158 Milanino<br />

In fact, in 1928 copper was first shown to be essential for life (23) since<br />

it is required for the synthesis of hemoglobin. Subsequently, its involvement<br />

in inflammation was demonstrated both in humans (1938) and laboratory<br />

animals (1953) (24,25). Almost 30 years after the scientific reports above,<br />

in 1967, a dramatic increase of the concentration in ceruloplasmin (the<br />

major multifunctional copper protein present in the serum, synthesized by<br />

the liver and deeply involved in iron metabolism and copper delivery to the<br />

extrahepatic tissues) was observed in the serum and periodontal tissue<br />

specimens of patients suffering from periodontal disease (an acute inflammatory<br />

process) (26,27). In 1968, a relevant increase of serum copper and<br />

ceruloplasmin was also reported in rheumatoid arthritic patients (28). Subsequently,<br />

the same evidence was found to characterize the adjuvant arthritis<br />

of the rat, an experimental systemic disease still the best existing model<br />

of human rheumatoid arthritis (29,30).<br />

All the above were only occasional reports, however. The leading<br />

breakthroughs that characterized the beginning of scientific development<br />

of ‘‘copper and inflammation’’ research came in 1976, when Sorenson’s (31)<br />

classic paper indicated many copper complexes as active acute and chronic<br />

anti-inflammatory agents, and when Whitehouse (32) posed the question<br />

about a possible ‘‘ambivalent,’’ i.e., pro- and anti-inflammatory, role of<br />

copper in the development and control of inflammation.<br />

CONCLUSIONS<br />

Today, the fact that ‘‘endogenous’’ copper exerts a key role in the development<br />

and control of inflammation is a well-recognized biological<br />

phenomenon (33). Moreover, the anti-inflammatory/antiarthritic potential<br />

of ‘‘exogenous’’ copper in curing experimental models of inflammation has<br />

also been demonstrated repeatedly by many research groups (34,35). Recently,<br />

great progress has been made, at the molecular level, in understanding<br />

how copper ‘‘traffics’’ in the organism and within the cells. Although this<br />

basic topic of research is still in its infancy, in the near future it may yield<br />

essential information for the understanding of the mechanism(s) by which<br />

this metal, both ‘‘endogenous’’ and ‘‘exogenous,’’ can keep under appropriate<br />

control the development of the inflammatory process.<br />

Therefore, the present state of the art appears most encouraging for<br />

the continuation of research to ascertain whether copper could represent a<br />

truly innovative approach to the curing of human inflammatory diseases,<br />

rheumatoid arthritis in primis.<br />

Nevertheless, few serious attempts have been made, and none currently,<br />

to study the use of copper in the therapy of such diseases. There<br />

are several prejudicial barriers that keep copper out of both experimental<br />

laboratories and the clinic: copper is generally thought to be far more toxic


Copper in Medicine and Personal Care 159<br />

than it actually is (36) and, especially, the marketing of copper compounds<br />

offers profits too low to be advantageous to business.<br />

Nonetheless, we would like to stress that the pharmacological exploitation<br />

of the anti-inflammatory and antiarthritic copper potential is<br />

anything but an obsolete idea. On the other hand, rheumatoid arthritis<br />

and related degenerative diseases that are found worldwide and are the<br />

cause of great disability with high social and economic costs, still wait for<br />

a pharmacological therapy that is both effective and safe. One can hope<br />

for the continuation of research in this field.<br />

ABBREVIATIONS<br />

L taxonomic classification by Linné<br />

R & P taxonomic classification by Ruiz and Pavon<br />

REFERENCES<br />

1. Carpanetto D, Bianchini P. l’Enciclopedia: Atlante Storico. Vol. 31. Chap. 1.<br />

Roma: Gruppo Editoriale l’Espresso, 2004.<br />

2. Sterpellone L. Dagli dei al DNA. Vol. 1. Chapters 1–6. Roma: Antonio Delfino<br />

Editore, 1988.<br />

3. Baines J, Málek J. Atlante dell’Antico Egitto. Novara: Istituto Geografico De<br />

Agostini, 1985:36.<br />

4. von Beckerath J. Handbuch der ägyptischen Königsnamen. MÄS 1984; 20:158.<br />

5. David RA. The pyramid builders of ancient Egypt. In: Chapters V and VI.<br />

London: Routledge and Kegan, 1986.<br />

6. Rossi Osmida G. La scoperta della vanità. Archeo 1989; 58:62.<br />

7. Guy E, Rachet MF. Dictionnaire de la Civilisation égyptienne. Paris: Librairie<br />

Larousse, 1972:100,192.<br />

8. Sorenson JRJ. Pharmacological activities of copper compounds. In: Berthon G, ed.<br />

Handbook of metal–ligand interactions in biological fluids. Vol. 2. New York:<br />

Marcel Dekker Inc., 1995:1128.<br />

9. Windholz M, ed. The Merk index. 10th ed. Rahway: Merck & Co. Inc.,<br />

1983:379.<br />

10. Radicchi R. Civiltà sumero-akkadica. Chapters 4 and 5. Pisa: Giardini Editore,<br />

1968.<br />

11. Sterpellone L. Dagli dei al DNA. Vol. 2. Chapters 1–4. Roma: Antonio Delfino<br />

Editore, 1988.<br />

12. Capasso L. Aria al cervello. Archeo 1990; 63:121.<br />

13. Cuory C. Mèdècine de l’Amerique precolombienne. Chapters 2,3, and 6. Paris:<br />

Roger Dacosta, 1969.<br />

14. Pravega D. La terapia nella medicina greca. Chapters 1–7. Pisa: Giardini<br />

Editore, 1963.<br />

15. Walker WR, Beveridge SJ, Whitehouse MW. Dermal copper drugs: the copper<br />

bracelet and Cu(II) salicylate complexes. In: Rainsford KD, Brune K,


160 Milanino<br />

Whitehouse MW, eds. Trace elements in the pathogenesis and treatment of<br />

inflammation. Basel: Birkhäuser Verlag, 1981:359.<br />

16. Lotz L-O, Weser U. Biological chemistry of copper compounds. In: Rainsford KD,<br />

et al, eds. Copper and zinc in inflammatory and degenerative diseases. Chap. 3.<br />

Dordrecht: Kluwer Academic Publisher, 1998.<br />

17. Capasso L. I romani in farmacia. Archeo 1989; 57:54.<br />

18. Ackerknecht EH. Therapeutics from the primitives to the 20th century. Chapters<br />

I–VI. New York: Hafner Press, 1973.<br />

19. Bonta IL. Microvascular lesions as target of anti-inflammatory and certain other<br />

drugs. Acta Physiol Neerl 1969; 15:188.<br />

20. Bynum WF, Browne EJ, Porter R, eds. Macmillan dictionary of the history of<br />

science. New York: The Macmillan Press, 1981:449.<br />

21. Vattimo G, Ferraris M, Marconi D (consultant authors). Le Garzantine: Filosofia.<br />

2nd ed. Milano: Garzanti Libri s.p.a., 1999:1036.<br />

22. Deuschle U, Weser U. Copper and inflammation. Prog Clin Biochem Med 1985;<br />

2:97.<br />

23. Hart EB, Steembock H, Waddel J, et al. Iron in nutrition. Vol. VII. Copper as a<br />

supplement to iron for haemoglobin building in the rat. J Biol Chem 1928; 77:797.<br />

24. Heilmeyer L, Stuwe G. Der Eisen-kupferantagonismus im Blutplasma beim<br />

Infektionsgeschehen. Klin Wochenschr 1938; 17:925.<br />

25. Wintrobe MM, Cartwright CE, Gubler CJ. Studies on the function and metabolism<br />

of copper. J Nutr 1953; 50:395.<br />

26. Linder MC. Biochemistry of copper. Chap. 4. New York: Plenum Press, 1991.<br />

27. Sweeney SC. Alterations in tissues and serum ceruloplasmin concentration associated<br />

with inflammation. J Dent Res 1967; 46:1171.<br />

28. Lorber A, Cutler LS, Chang CC. Serum copper levels in rheumatoid arthritis: relationship<br />

of elevated copper to protein alteration. Arthritis Rheum 1968; 11:65.<br />

29. Karabelas DS. Copper metabolism in the adjuvant induced arthritic rat. Ph.D.<br />

thesis. Ann Arbor: Michigan State University, 1972.<br />

30. Rainsford KD. Adjuvant polyarthritis in rats: is this a satisfactory model for<br />

screening anti-arthritic drugs? Agents Actions 1982; 12:452.<br />

31. Sorenson JRJ. Copper chelates as possible active forms of the anti-arthritic<br />

agents. J Med Chem 1976; 19:135.<br />

32. Whitehouse MW. Ambivalent role of copper in inflammatory disorders. Agents<br />

Actions 1976; 6:201.<br />

33. Milanino R, Velo GP, Marrella M. Copper and zinc in the pathophysiology and<br />

treatment of inflammatory disorders. In: Nève J, Chappuis P, Lamand M, eds.<br />

Therapeutic uses of trace elements. New York: Plenum Publishing Corporation,<br />

1996:115.<br />

34. Milanino R, Moretti U, Marrella M, et al. Copper and zinc in the development<br />

and control of inflammation. In: Handbook of metal–ligand interactions in biological<br />

fluids. Berthon G, ed. Vol. 2. New York: Marcel Dekker Inc., 1995:886.<br />

35. Sorenson JRJ. Copper complexes offer a physiological approach to treatment of<br />

chronic diseases. In: Ellis GP, West GB, eds. Progress in medicinal chemistry.<br />

Vol. 26. Amsterdam: Elsevier, 1989:437.<br />

36. Medeiros DM, Wildman R, Liebes R. Metal metabolism and toxicities. In:<br />

Massaro EJ, ed. Handbook of human toxicology. Chap. 3. Boca Raton: CRC<br />

Press LLC, 1997.


9<br />

The Role of Copper in Onset,<br />

Development, and Control of<br />

Acute and Chronic Inflammation<br />

Roberto Milanino<br />

Facoltà di Medicina e Chirurgia, Sezione di Farmacologia,<br />

Dipartimento di Medicina e Salute Pubblica, Università di Verona,<br />

Verona, Italy<br />

INTRODUCTION<br />

The involvement of ‘‘endogenous’’ copper in inflammation was discovered<br />

in the second half of the last century when evidence was published that:<br />

(i) total serum copper markedly increased in aseptic and septic acute inflammation,<br />

such as turpentine edema, Staphylococcus aureus abscesses, and<br />

typhoid vaccine injection (1953); (ii) total serum copper and ceruloplasmin<br />

as well as the ceruloplasmin measured within the inflamed tissue significantly<br />

increased in patients suffering from acute periodontal disease (1967);<br />

(iii) total serum copper was found significantly elevated in rheumatoid<br />

arthritic patients (1968), and in adjuvant-induced arthritis (AA) in the rat<br />

(1972), still a very useful experimental model for that human disease (1–5).<br />

It was, however, only in 1976, i.e., about two millennia after the first<br />

This chapter, in part, was reprinted from Milanino R. Copper: an overview of the role of endogenous<br />

and exogenous metal in the development and control of the inflammatory process. Rev<br />

Environ Health 2006; 21:1–70, with permission.<br />

161


162 Milanino<br />

documented intuition by Celsus on this issue in pharmacology (6), that the<br />

researchers’ attention again addressed the role of ‘‘exogenous’’ copper as a<br />

potentially effective anti-inflammatory and antiarthritic drug. In fact, in<br />

that year, two major papers were published.<br />

The first one, due to the work of J.R.J. Sorenson (7), not only showed<br />

that many subcutaneously injected copper(II) complexes or salts were very<br />

active anti-inflammatory and antiarthritic agents, but also proposed that the<br />

complexes extemporaneously formed in vivo with ‘‘endogenous’’ copper were<br />

the real active forms of the most common and currently used antiarthritic drugs<br />

(such as salicylate, aspirin, D-penicillamine, etc.). The above theory has actually<br />

been the subject of a lively debate among medicinal chemists, coordination chemists,<br />

and pharmacologists, and it is still an unresolved question.<br />

The second, especially stimulating paper was published by M.W.<br />

Whitehouse (8) in the same year. In that article, the author speculated on<br />

the role of both ‘‘endogenous’’ and ‘‘exogenous’’ copper in inflammation,<br />

based on the somewhat contradictory evidence known at that time:<br />

In some circumstances, soluble copper preparations behave as<br />

acute or chronic inflammatory (or irritant) agents.<br />

Acute and chronic inflammations are both characterized by a significant<br />

increase of total serum copper, which, in light of its irritating<br />

potential, may be even regarded as a factor capable of worsening<br />

the pathological process.<br />

D-Penicillamine, a very well-known de-coppering agent that is still<br />

now successfully used in the therapy of Wilson’s disease (9), is also<br />

a valuable antiarthritic drug.<br />

Pathological (jaundice and other liver diseases) or physiological<br />

(pregnancy) events that cause total serum copper to remarkably<br />

increase above normal levels, on the other hand, often brought<br />

about spontaneous remission of rheumatoid arthritis.<br />

Supplementation with copper using a number of folk remedies such as<br />

Cu bangles, cider vinegar, shellfish, nuts, etc., as well as the treatment<br />

with copper complexes or salts reported by Sorenson (7), seem to be or<br />

actually are remarkably good anti-inflammatory/antiarthritic agents.<br />

Thus, Whitehouse proposed that copper, either ‘‘endogenous’’ or<br />

‘‘exogenous,’’ can be both injurious and beneficial, even at one and the same<br />

time, acting in the organism as ‘‘inert,’’ ‘‘toxic,’’ or ‘‘pharmacoactive,’’<br />

depending on the different physiological or pathological conditions characterizing<br />

the organism itself.<br />

The actual state of things was then clearly far from being settled, and<br />

this intriguing issue induced a number of research teams to begin studying in<br />

more detail the roles that ‘‘endogenous’’ and ‘‘exogenous’’ copper could<br />

really have on the onset, development, and control of either acute or chronic<br />

inflammation.


The Role of Copper in Inflammation 163<br />

STUDIES ON COPPER-DEFICIENT, EXPERIMENTALLY<br />

INFLAMED ANIMALS<br />

In 1977, our research group set out the idea to study the development of<br />

acute and chronic inflammatory processes in experimental animals made<br />

deficient of copper by feeding them with a diet containing very low amounts<br />

of this essential metallo-element. The rationale of the approach was based<br />

on the assumption that inflammation developing in a copper-deprived<br />

animal could perhaps give some valuable indication about what the prevailing<br />

effect of copper on this kind of diseases was, i.e., a pro- or an<br />

anti-inflammatory one. Before beginning the project, normal female and<br />

male rats were kept on diets that, according to Owen and Hazelrig (10), were<br />

either sufficient (10 ppm) or deficient (0.4 ppm) in copper for three months,<br />

and then a complete toxicological examination of the animals was performed.<br />

It resulted that the copper-deprived female rats had a reduced<br />

ponderal growth compared with control animals. However, the major blood<br />

cell and hematochemical serum markers were within normal range; moreover,<br />

normal was also the condition of the numerous tissues, organs, and<br />

apparatus, examined both macro- and microscopically (11). On the other<br />

hand, the same copper-deficient dietary regimen was severely impairing<br />

for male rats’ survival, and marked deviations from normality were found<br />

evaluating many of the parameters mentioned above (unpublished observation).<br />

This latter finding was subsequently confirmed by Fields and<br />

coworkers (12), although it is still an unexplained phenomenon.<br />

Acute Inflammation<br />

First, the effect of copper depletion on the development of an aseptic acute<br />

inflammatory process was examined in female rats kept on a 0.4 ppm Cucontaining<br />

diet for 30 or 90 days, using the carrageenan-induced paw edema<br />

(CPE) model (13). Between the 15th and the 20th day from the beginning of<br />

the copper-deficient feeding, the concentration of serum copper was found dramatically<br />

reduced, to about a mean 6% of the control values, and this level<br />

remained constant throughout the whole experiment (Table 1, zero time).<br />

The effects of the treatment on the development of the paw edema were, nonetheless,<br />

totally different. In fact, after 30 days of copper deprivation the mean<br />

swelling measured in the deficient group was comparable to that of the control,<br />

whereas 90 days after the beginning of the experiment, the metallo-elementdeficient<br />

animals developed an inflammatory reaction significantly greater than<br />

that measured in the normally fed controls, which reached its maximum difference<br />

(þ58%) about 20 hours after the subplantar irritant injection (Table 1).<br />

The above proinflammatory effect of copper depletion was confirmed<br />

in male rats by Denko and coworkers (14,15), who employed the same<br />

experimental model (maximum increase of the foot edema þ31%), although<br />

the deficient diet used was less severe in both copper content (0.6 ppm) and


164 Milanino<br />

Table 1 Serum Copper Concentrations and Paw Edema Volumes After 30 and<br />

90 Days of Normal (Cu ¼ 10 ppm) or Copper-Deficient (Cu ¼ 0.4 ppm) Feeding<br />

Days of<br />

diet<br />

Hours after<br />

challenge<br />

Total serum copper<br />

(mg/dL SD)<br />

Paw edema volume<br />

(plethysmographic<br />

units SD)<br />

Normal Deficient Normal Deficient<br />

30 0 193.2 12.1 10.7 — —<br />

3 185.9 20.3 11.9 14.1 22.2 3.4 21.3 2.6<br />

5 179.2 20.6 10.1 13.8 24.1 3.5 22.9 3.9<br />

20 270.3 22.3 a<br />

91.7 18.1 a<br />

15.4 3.6 14.5 3.8<br />

90 0 190.4 21.0 12.9 11.4 — —<br />

3 197.1 20.4 10.5 15.9 22.1 3.0 28.6 4.1 b<br />

5 187.6 19.1 26.1 19.2 25.3 3.5 30.8 5.1 c<br />

20 276.2 22.1 a<br />

102.2 19.0 a<br />

15.1 3.6 23.8 5.7 b<br />

Note: Statistics—Student’s t test (data standard deviations; N ¼ 13 rats per group and per hour).<br />

a P < 0.001; comparison versus the zero time values of either normally fed or deficient rats.<br />

b P < 0.001; comparison versus the time-matched, normally fed animals.<br />

c P < 0.010; comparison versus the time-matched, normally fed animals.<br />

Source: The data reported in this table are the mean values of those obtained in an unpublished<br />

preliminary experiment (N ¼ 3 rats per group) and those published in Ref. 13.<br />

length of feeding (30 days). However, the data reported in Table 1 provided<br />

other, relevant evidence. Firstly, the proinflammatory effect observed was<br />

independent of the amount of total copper present in serum, but dependent<br />

on the metallo-element-deficient length of the dietary intake. Secondly, at the<br />

20th hour and in all tested groups, the acute inflammatory challenge promoted<br />

a statistically significant increase of total serum copper concentration<br />

that, later on, we demonstrated to be very highly correlated to the serum<br />

ceruloplasmin level, as measured by the oxidase activity method (16). Interestingly,<br />

this total serum copper increase was found to be far more dramatic in<br />

the deficient rats (þ671% at day 30, and þ873% at day 90) when compared<br />

with that measured in the control animals (þ45% at day 30, and þ40% at<br />

day 90). Ceruloplasmin being a copper protein essentially synthesized and<br />

secreted by the liver as an acute-phase reactant, and considering that it was<br />

proposed to exert a protective role in inflammation, one could speculate that<br />

this increase of circulating ceruloplasmin may represent the first evidence of a<br />

main role for ‘‘endogenous’’ copper as a physiological agent acting in the<br />

control of the development and remission of inflammatory process (14,17).<br />

To better understand the soundness of the latter hypothesis, as well as<br />

the fact that the proinflammatory effect of copper deficiency was independent<br />

of total serum copper concentrations and, conversely, linked to the<br />

amount of copper contained in the depleted diet and to the length of time<br />

that the animals were exposed to it, further experiments were done. Two


The Role of Copper in Inflammation 165<br />

different copper-deficient diets were considered (containing either 0.2 or<br />

0.7 ppm of copper), fed to female rats for two different periods of time<br />

(respectively, 30 and 150 days), in two models of aseptic carrageenaninduced<br />

acute inflammation, i.e., the relatively mild CPE, and the more<br />

distressing carrageenan-induced pleurisy (CP) (18).<br />

The results obtained in the CPE test following the 0.2 ppm copperdepleted<br />

diet regimen showed that the proinflammatory effect observed after<br />

30 days of preliminary deficient feeding was not only very obvious (Table 2)<br />

but also remarkably higher than that obtained in the previous experiment<br />

(Table 1) (13). In fact, the average paw swellings measured three, five, and<br />

20 hours after the challenge were of þ36% (0.4 ppm Cu diet for 90 days,<br />

Table 1) versus a þ51% (0.2 ppm Cu diet for 30 days, Table 2); moreover,<br />

the maximum enhancement of the local inflammatory reaction, which was<br />

gauged at the 20th hour also in the 0.2 ppm diet-treated group, was of<br />

about 95% (Table 2). A similar dramatic proinflammatory effect was promoted<br />

as well by the 0.2 ppm copper-deficient diet given for 30 days in<br />

the carrageenan-induced pleurisy model.<br />

In the CP experimental disease, the intrathoracic injection of a sterile<br />

carrageenan suspension caused the leakage, from the blood into the thoracic<br />

cavity itself, of an inflammatory exudate composed by a fluid fraction in<br />

which a large amount (tens of millions) of ‘‘inflammatory’’ cells (mainly<br />

Table 2 Serum Copper Concentrations and Paw Edema (CPE) or Exudate (CP)<br />

Volumes After 30 Days of 0.2 ppm Copper-Deficient Feeding (Control Diet:<br />

Cu ¼ 10 ppm)<br />

Inflammatory<br />

model<br />

Hours after<br />

challenge<br />

Total serum copper<br />

(md/dL SD)<br />

Paw edema or<br />

exudate volumes<br />

(plethysmografic<br />

units or mL SD)<br />

Normal Deficient Normal Deficient<br />

CPE 0 171.2 22.5 4.6 7.2 0 0<br />

3 167.2 29.6 7.3 7.6 18.2 1.6 25.4 3.7 b<br />

5 163.5 30.1 5.8 4.3 24.3 2.1 28.9 1.5 b<br />

22 291.3 29.4 a<br />

24.9 26.8 10.1 3.3 19.7 3.1 b<br />

CP 0 171.4 29.1 7.9 5.9 0 0<br />

6 173.8 26.1 6.7 3.7 0.83 0.3 1.64 0.4 b<br />

22 248.8 24.2 a<br />

12.1 9.4 — —<br />

Note: Statistics—Student’s t test (data standard deviations; N ¼ 13 rats per group and per hour).<br />

a<br />

P < 0.001; comparison versus zero time values (CPE or CP), of either normally fed or<br />

deficient rats.<br />

b<br />

P < 0.001; comparison versus time-matched, normally fed animals.<br />

Source: The data reported in this table are the mean values of those obtained in an unpublished<br />

preliminary experiment (N ¼ 3 rats per group) and those published in Ref. 18.


166 Milanino<br />

neutrophils) were present. This inflammatory reaction (that is routinely<br />

scored six hours after the challenge) was found to be remarkably higher in<br />

the copper-deprived rats compared with the response of the normally fed<br />

pleuritic animals (þ98%, Table 2). Nevertheless, the increase of the total<br />

exudate volume was accompanied by a proportional rise in its cellular component,<br />

which, in turn, seemed to suggest that the anti-inflammatory activity<br />

of ‘‘endogenous’’ copper might have little effect on the overall process of<br />

leukocyte migration, at least in conditions of copper deficiency and, in this<br />

particular model, of acute inflammation. However, another evidence needs to<br />

be stressed. The severity of the 0.2 ppm-induced copper deficiency seemed<br />

to have reduced the body stores of the metal to such an extent as to prevent<br />

the inflammatory process from triggering the statistically significant increase<br />

of total serum copper (Table 2). Such an increase is seen, as a rule, in<br />

normally fed rats, and was reported also in the copper-depleted, inflamed<br />

animals examined in the previous experiment (Tables 1 and 2) (13).<br />

When the above acute inflammations (i.e., CPE and CP) were induced<br />

in rats fed a 0.7 ppm Cu diet for about five months, no significant differences<br />

were measured in either the paw edema or the total exudate volumes<br />

between normally fed and copper-deficient animals (Table 3) (18).<br />

Table 3 Serum Copper Concentrations and Paw Edema (CPE) or Exudate (CP)<br />

Volumes After 150 Days of 0.7 ppm Copper-Deficient Feeding (Control Diet:<br />

Cu ¼ 10 ppm)<br />

Inflammatory<br />

model<br />

Hours after<br />

challenge<br />

Total serum copper<br />

(md/dL SD)<br />

Paw edema or<br />

exudate volumes<br />

(plethysmografic units<br />

or mL SD)<br />

Normal Deficient Normal Deficient<br />

CPE 0 172.3 27.9 21.5 20.8 0 0<br />

3 — — 18.9 6.0 20.3 5.7<br />

5 — — 29.3 8.2 27.9 8.4<br />

22 250.4 31.8 a<br />

70.1 89.9 13.8 5.1 12.7 4.9<br />

CP 0 171.4 29.1 21.3 29.1 0 0<br />

6 — — 1.0 0.2 1.1 0.4<br />

22 285.3 33.9 a<br />

89.6 86.4 b<br />

— —<br />

Note: Statistics—Student’s t test (data standard deviations; N ¼ 13 rats per group and per hour).<br />

a<br />

P < 0.001; comparison versus the zero time values (CPE or CP), of either normally fed or deficient<br />

rats.<br />

b<br />

P < 0.050; comparison versus the zero time values (CPE or CP), of either normally fed or deficient<br />

rats.<br />

Source: The data reported in this table are the mean values of those obtained in an unpublished<br />

preliminary experiment (N ¼ 3 rats per group) and those published in Ref. 18.


The Role of Copper in Inflammation 167<br />

The determination of total serum copper levels (made at zero time<br />

and 20 hours after the challenge) showed that this parameter increased, as<br />

expected, in the inflamed controls, but its rise was far less significant in<br />

the inflamed-depleted animals. However, this latter and unpredicted result<br />

is not considered to be a biologically significant one since the single means<br />

measured were characterized by extremely high standard deviation values<br />

(Table 3) (18).<br />

The above-summarized data lead to suggest the following proposals:<br />

In rats, the induction of a copper-deficiency status is a condition<br />

that promotes a very significant enhancement of the acute inflammatory<br />

reaction, which, in turn, speaks in favor of an anti- rather<br />

than a proinflammatory role of ‘‘endogenous’’ copper.<br />

The observation that the amount of copper left in the diet and/<br />

or the length of the deprived feeding are able to influence the<br />

reported phenomenon seems to indicate a sort of dose- and<br />

time-dependent effect of copper deficiency, testifying that this<br />

phenomenon is a real and not a casual event. It must be stressed<br />

that this conclusion has been confirmed in a subsequent study by<br />

Kishore and coworkers (19).<br />

The rats not dramatically depleted of copper (i.e., those that underwent<br />

feeding with a diet containing 0.4 ppm of the metallo-element)<br />

react to the challenge by secreting relatively striking amounts of<br />

ceruloplasmin into the blood from the liver. Considering that this<br />

protein appears to exert an anti-inflammatory action in vivo (14,20),<br />

this evidence seems to further support the hypothesis of a natural<br />

defensive role of ‘‘endogenous’’ copper in inflammation.<br />

Finally, the findings just mentioned led us to speculate that the<br />

appearance of the enhanced acute inflammatory reaction promoted<br />

by the deficiency of copper could strictly depend on the amount of<br />

copper still present in the liver (21). Later on, this hypothesis found<br />

its experimental demonstration in male rats fed a 0.5 ppm Cu for<br />

20, 40, or 60 days and then tested in the CPE model (19). Interestingly,<br />

in the same paper the author showed that not only the paw<br />

edema in copper-deficient animals was highly and negatively correlated<br />

to the concentration of copper in the liver, but also that<br />

the correlation with liver Cu,Zn superoxide dismutase (SOD 1)<br />

activity was inconsistent (19). Conversely, the activity of SOD 1<br />

in total blood cells of copper-deprived rats (fed a diet containing<br />

0.2 ppm of copper, for one month) was found to be reduced to<br />

about 50% of the control values; similar results were also obtained<br />

in copper-deficient pigs (22,23). These two last observations directed<br />

the attention to the role that superoxide radicals play in the<br />

development of the inflammatory process in vivo, and, in turn, in


168 Milanino<br />

the scavenging activity of many copper compounds other than the<br />

SOD 1 itself (24,25). Thus, the above information led us to speculate<br />

that ‘‘endogenous’’ copper could control the inflammation also by<br />

regulating the superoxide metabolism, and, as a consequence, its<br />

deficiency may, at least in part, justify the exacerbation of the acute<br />

inflammatory reaction observed in the laboratory animal (18).<br />

Chronic Inflammation<br />

The effects of dietary copper deficiency on experimental chronic inflammation<br />

were first studied in the adjuvant arthritis of the female rat, after<br />

60 days of feeding the animals with either a copper-depleted (Cu 0.4 ppm)<br />

or a control (Cu 10 ppm) diet, before carrying out a tail-intradermal injection<br />

of heat-killed Mycobacterium butyricum finely suspended in liquid<br />

paraffin (complete adjuvant) (26). Note that the copper-deficient feeding<br />

was continued throughout the experiment (26).<br />

As expected, in the normally fed rats the systemic reaction to the challenge<br />

began to become evident macroscopically at about 12–14 days after<br />

the inoculum and reached its maximum development between the 18th<br />

and the 28th day (Fig. 1, upper panel). Together with a marked loss of<br />

body weight, the other major pathological signs [that represent the basis<br />

of the arthritic score routinely used to assess disease severity, the maximum<br />

theoretical value of which is 31 (5,26)] showed: (i) a dramatic swelling of the<br />

paws, particularly the hind ones; (ii) the appearance of numerous arthritic<br />

nodules randomly disseminated in the tail; and, especially, (iii) a very deep<br />

bone and cartilage degradation at the joint levels (chiefly the hind tibiotarsals),<br />

very clearly documented at both X-ray and histological examination,<br />

of which a severe joint stiffness and, in turn, a dramatic impairment<br />

of the deambulation ability were the consequence. In our experiment, in this<br />

group of normally fed animals, the highest arthritic score (29.4) was measured<br />

at day 21 (Fig. 1, upper panel) (26). At the same time, a progressive<br />

increase of total serum copper concentration was observed, which also<br />

reached its maximum (þ82%) 21 days after complete adjuvant injection<br />

(Fig. 1, lower panel) (26). On the contrary and rather surprisingly, the<br />

copper-deficient animals did not react to complete adjuvant injection, and<br />

the systemic response expected was almost totally abolished by dietary<br />

copper depletion. In fact, only a barely visible swelling of the hind paws<br />

was observed 21 days after the challenge (highest arthritic score 4.3; Fig. 1,<br />

upper panel), and the X-ray and histological examinations showed that the<br />

joint status of these copper-deficient and complete adjuvant-injected rats<br />

appeared to be almost fully normal (26). Moreover, throughout the experiment<br />

the total serum copper concentration, albeit extremely low, remained<br />

constant ranging between 10 and 15 mg/dL (Fig. 1, lower panel) (26). The<br />

fact that the diet-induced copper deficiency was a condition able to strongly


The Role of Copper in Inflammation 169<br />

Arthritic<br />

score<br />

Serum Cu<br />

(µg/dL)<br />

35<br />

28<br />

21<br />

14<br />

7<br />

0<br />

350<br />

280<br />

210<br />

140<br />

70<br />

0<br />

*<br />

0 7 14 21 28<br />

Days after the inoculum<br />

*<br />

* * * *<br />

0 7 14 21 28<br />

Days after the inoculum<br />

Cu-normal<br />

Cu-deficient<br />

Figure 1 Development of the adjuvant arthritis (arthritic score) and increases of<br />

total serum copper concentrations in normally fed and dietary Cu-depleted rats.<br />

Arthritic score (upper panel) and total serum copper (lower panel) in normally fed<br />

and copper-deficient adjuvant-arthritic rats. Source: From Refs. 11 and 26.<br />

inhibit the normal development of the AA of the rat was confirmed by other<br />

authors (27).<br />

Previous reports have shown that copper is involved in the prostaglandin<br />

biosynthesis, and, in particular, that it seems to increase the conversion<br />

of arachidonic acid to prostaglandin F2a (an anti-inflammatory mediator),<br />

and to reduce the production of the prostaglandin E2 (a proinflammatory<br />

product of the arachidonic acid cascade) (28–31). In our experiment (26),<br />

we showed that the lung prostaglandin-synthetase activity was the same in<br />

*<br />

Cu-normal<br />

Cu-deficient


170 Milanino<br />

normal and copper-depleted nonarthritic animals. Moreover, no difference<br />

was observed in the reactivity to exogenous prostaglandin E2 or F2a added<br />

to strips of rat stomach fundus or colon from nonarthritic copper-deficient,<br />

compared with nonarthritic normally fed animals. Thus, the above data<br />

seemed to exclude the influence of copper depletion on prostaglandin<br />

synthesis pathway as a possible mechanism explaining the observed phenomenon.<br />

On the other hand, at that time it was already known that a lower<br />

immunological reactivity (as measured by cytotoxic anaphylactic reaction,<br />

phagocytic, and serum bactericidial activities) was promoted by copper<br />

deficiency in the rat (32). Consequently, considering that in the development<br />

of the adjuvant-arthritis model a normal response of the immune system is<br />

strictly required, we proposed that the protective effect of copper deficiency<br />

on the ‘‘normal’’ development of the AA was an epiphenomenon secondary<br />

to an impairment of the immune function (5,26). This hypothesis was<br />

confirmed later on by Kishore and coworkers (33), who showed that the<br />

copper-deficient male rats (Cu in the diet 0.5 ppm; length of feeding 40 days)<br />

were actually in a state of apparent immunosuppression as demonstrated by<br />

impaired ex vivo responsiveness to the T-cell-dependent contact-sensitizing<br />

antigen oxazolone and diminished capacity to respond to the T-cellindependent<br />

antigen Type III pneumococcal polysaccharide stimulation.<br />

LABORATORY ANIMALS: STUDIES ON ‘‘ENDOGENOUS’’<br />

COPPER METABOLISM IN ACUTE AND<br />

CHRONIC INFLAMMATION<br />

Illnesses always promote more or less remarkable, and sometimes dramatic,<br />

changes of the whole-body homeostatic mechanisms that come into play<br />

with the aim of self-defense, or may be ‘‘side effects’’ of the disease itself,<br />

which could even contribute to worsening the condition. This was, actually,<br />

the theoretical question posed by Whitehouse (8) speculating on the possible<br />

‘‘inert,’’ ‘‘toxic,’’ or ‘‘pharmacoactive’’ role of ‘‘endogenous’’ copper in the<br />

development and control of inflammation. Thus, in light of the ascertained<br />

proinflammatory effect of nutritional copper deficiency on the development<br />

of the acute processes, it seemed reasonable to conduct more detailed studies<br />

on the changes in ‘‘endogenous’’ copper metabolism induced by the inflammatory<br />

process in laboratory animals. Some of the most significant results<br />

obtained are summarized in Tables 4 (and references therein; acute inflammation)<br />

and 5 (and references therein; chronic inflammation).<br />

Status of Copper in Blood and Urine<br />

Since the early reports, many different studies carried out in different animal<br />

species have shown that the acute inflammatory process, regardless of the<br />

noxa used to promote it, is characterized by a significant increase of total


The Role of Copper in Inflammation 171<br />

Table 4 Experimental Animals: Examples of the Changes in Copper Status<br />

Induced by Acute Inflammation in Some Relevant Body Compartments<br />

Species Disease<br />

Total<br />

serum<br />

Blood<br />

cells Liver<br />

Kidney<br />

Inflamed<br />

area References<br />

Rat Turpentine edema I U — — — 1<br />

Radiation injury U — — — — 34<br />

Turpentine abscesses I — I — — 35<br />

Femur fracture — — D — — 36<br />

CP I I U U — 37<br />

CP I U U — P 38<br />

CPO I U U — I 38<br />

Rabbit Turpentine edema I — — — — 39<br />

Ocular inflammation I — — — I 40<br />

Ocular inflammation — — — — I 41<br />

Dog Turpentine abscesses I — — — — 42<br />

Guinea pig Turpentine abscesses I — — — — 43<br />

Mouse Collagenase injection I — — — — 2<br />

Abbreviations: I, significantly increased; U, unchanged; D, significantly decreased; P, present in<br />

the inflammatory exudate.<br />

serum (or plasma) copper concentration (Table 4) (2,3,34–43). An exception<br />

was apparently represented by the effect of radiation injury in the rat (34).<br />

However, this result failed to be confirmed by more recent work, which<br />

showed a remarkable increase of serum copper, even in that experimental<br />

model of inflammation (44,45). Also, chronic inflammation induced in<br />

the experimental animals typically brings forth a remarkable rise of total<br />

serum (or plasma) copper (Table 5), albeit an occasional report showed<br />

that, in dogs chronically suffering from nonexperimentally induced dermatitis<br />

or anal gland fistula, these diseases did not cause significant changes in<br />

metallo-element status in the serum (4,33,42,46–51).<br />

As a general rule, the inflammation-induced increase of total serum<br />

copper is accompanied by a parallel increase of serum ceruloplasmin<br />

concentration, and, as noted before, these two parameters characterizing<br />

serum copper status are highly and statistically significantly correlated in<br />

normal as well as acutely and chronically inflamed animals (4,16,52,53).<br />

Moreover, it has been shown in hamsters that transcription of ceruloplasmin<br />

mRNA increases within three hours of induction of inflammation by turpentine<br />

injection, reaching a peak 2.5-fold above normal at 12–18 hours (54).<br />

Thus, at least in animals fed a diet containing standard amounts of copper,<br />

the total serum (or plasma) copper measured during inflammation appears<br />

to represent an extremely reliable index of circulating ceruloplasmin levels,<br />

which, in turn, belies an old report according to which the human inflammatory<br />

process (rheumatoid arthritis) is distinguished by a selective rise of the


172 Milanino<br />

Table 5 Experimental Animals: Examples of the Changes in Copper Status Induced<br />

by Chronic Inflammation in Some Relevant Body Compartments<br />

Species Disease<br />

Total<br />

serum<br />

Blood<br />

cells Liver Kidney<br />

Inflamed<br />

area References<br />

Rat AA I — I D — 4<br />

AA I — I — — 33<br />

AA I — I — — 46<br />

Chronic<br />

I — I — — 47<br />

inflammations<br />

AA I I I D I 48<br />

AA I U I — — 49<br />

AA I — I U — 50<br />

Dog S. aureus-induced<br />

arthritis<br />

I — — — — 51<br />

Chronic otitis I — — — — 49<br />

Abbreviations: AA, adjuvant-induced arthritis; I, significantly increased; U, unchanged; D, decreased.<br />

non-ceruloplasmin-bound fraction of serum copper (1). This latter claim was<br />

subsequently further disproved by Freeman and O’Callaghan (55), who,<br />

directly measuring (following column fractionation) the non-ceruloplasmin-bound<br />

copper in the serum of normal and inflamed animals and<br />

humans, showed that this fraction of circulating metal undergoes only negligible<br />

variations during both adjuvant arthritis in the rat as well as human<br />

rheumatoid arthritis.<br />

Another issue is worth emphasizing in view of its relevance in the<br />

studies on ‘‘endogenous’’ copper involvement in human inflammatory diseases.<br />

Although the above-reported increases of circulating copper and/or<br />

ceruloplasmin are important markers in revealing the existence of an inflammatory<br />

state [unless, for example, pregnancy is present, or the subject is<br />

undergoing steroid therapy (56)], the data obtained during the past years<br />

in our laboratory, studying over 1200 inflamed rats, show that serum or<br />

plasma copper as well as ceruloplasmin concentrations are not directly correlated<br />

with the actual severity of the pathological condition examined, and<br />

cannot even discriminate between acute and chronic processes. In fact, it<br />

was found that different acute nonseptic inflammations (e.g., rat CPE and<br />

CP), which developed in very similar manner, caused remarkably variable<br />

increases in the above parameters (57,58). A great variation in increased<br />

total plasma copper concentration was also reported among adjuvantarthritic<br />

rats regardless of illness severity level (26,48). In particular, the<br />

average increase in total plasma copper in animals, which manifested experimental<br />

disease to a mild degree, was of about 54%, and that measured in<br />

rats bearing a fully expressed pathology was approximately 60% (46). Therefore,<br />

hypercupremia that characterizes any inflammatory reactions appears


The Role of Copper in Inflammation 173<br />

to be a sort of ‘‘all or nothing’’ phenomenon that has indeed little value as a<br />

marker of disease seriousness.<br />

To conclude the analysis on copper status in blood of acutely or<br />

chronically inflamed animals, the erythrocyte compartment remains to be<br />

considered. A number of contradictory data have been reported on this<br />

issue. In fact, according to some authors neither acute nor chronic inflammatory<br />

processes changed the concentration of copper in the erythrocytes,<br />

whereas an increase of this fraction of total blood copper was reported in<br />

rats with carrageenan-induced pleurisy and adjuvant arthritis (3,37,38,<br />

48,49). However, the increase of erythrocyte copper measured in the arthritic<br />

rats (48), although statistically significant at day 3 and 21 after challenge,<br />

was small and probably did not have major biological relevance. Moreover,<br />

as ascertained later on studying the same parameter in human rheumatoid<br />

arthritis (59), the rise of copper in the red cells of chronically inflamed subjects<br />

was, most likely, a secondary phenomenon on which the significant<br />

decrease in erythrocyte volume had a not negligible influence.<br />

Finally, very few and contradictory data exist on the urinary copper<br />

excretion in acutely and chronically inflamed rats. For example, the metal<br />

concentration in the urine was found to increase in femur-fractured animals,<br />

whereas the challenge with complete adjuvant did not modify this<br />

parameter (36,49). Therefore, at least as the experimental models are concerned,<br />

it is not possible to draw any conclusion on this topic on the basis<br />

of existing evidence.<br />

Status of Copper in Solid Tissues and Inflamed Areas<br />

Before getting into a detailed discussion of inflammation-induced changes<br />

in copper status in some solid tissue involved in onset, development, and<br />

control of the inflammatory processes, a preliminary issue deserves attention.<br />

In most, if not all, papers dealing with copper and inflammation, the<br />

amount of the metallo-element measured in the different body compartments<br />

is exclusively reported as concentration values. Nevertheless, in some<br />

instances this could be a misleading index. As a matter of fact, we studied<br />

the concentration and total amount of liver copper during a daily ninehour<br />

period in normal rats, i.e., between nine hours and 30 minutes and<br />

18 hours and 30 minutes (Table 6) (60).<br />

The results reported in Table 6 clearly show that during the period of<br />

time considered, an apparent statistically significant increase in liver copper<br />

concentration occurred. However, this phenomenon was essentially due to a<br />

parallel and progressive decrease of liver weight (possibly caused to different<br />

states of liver hydration and/or glycogen content); conversely, the total<br />

amount of the metal measured in this compartment remained constant<br />

throughout the experiment (60). This physiological phenomenon strongly<br />

suggests: (i) the evaluation of copper concentration and total amount, as well


174 Milanino<br />

Table 6 Daily Changes in Hepatic Copper Concentration and Total Amount in<br />

Normal Female Rats<br />

Time<br />

(hours)<br />

Total serum<br />

Cu (mg/dL)<br />

Liver weight<br />

(g)<br />

Liver copper<br />

Cu (mg/g)<br />

Total liver<br />

Cu (mg)<br />

0 151.3 6.6 4.31 33.03<br />

3 þ8% 4% þ5% þ2%<br />

6 þ0% 12% a<br />

þ16% b<br />

þ5%<br />

9 þ3 21% b<br />

þ25% b<br />

þ3%<br />

Note: Statistics—Student’s t test.<br />

a<br />

P < 0.05.<br />

b<br />

P < 0.01.<br />

Source: From Ref. 60.<br />

as (ii) the routine use of a control group (‘‘time controls’’) should be utilized<br />

in every experiment, and these rats should be killed at the same time as those<br />

of each treated group, thus ensuring that the results obtained and their discussion<br />

could have a better chance of being fully reliable (60). Actually, we<br />

strictly followed this rule in all the experiments carried out in our laboratory.<br />

As shown in Tables 4 and 5, the liver is the tissue in which copper<br />

status has been most frequently studied in inflamed animals. We already<br />

know that the liver is the major organ responsible for ceruloplasmin synthesis<br />

and secretion into blood (17,56). Then, in the acute processes, the first<br />

striking evidence is that, in spite of the remarkably increased production and<br />

release of this protein induced by inflammation, the level of total hepatic<br />

copper does not decrease (37,38). The femur fracture in the rat is, perhaps,<br />

the only model of an acute inflammatory process in which a reduced liver<br />

copper concentration was reported (36); whether or not this isolated result<br />

depends on the peculiarity of this important traumatic condition is unknown.<br />

On the other hand, when chronically inflamed animals were studied, the<br />

hepatic level of copper was always found to be significantly increased, both<br />

as concentration and total amount values (4,33,46–50). Moreover, we observed<br />

in the adjuvant-arthritic rat that a rise of hepatic copper level preceded<br />

the appearance of any visible signs of the disease, and, afterwards was found<br />

to be directly proportional to the manifested severity of the pathology<br />

(46,61). In greater detail, liver copper total amounts were statistically significantly<br />

increased above control values three days after the challenge (þ15%),<br />

i.e., during the asymptomatic phase (46). Furthermore, when the disease<br />

reached its complete development (i.e., at day 14, 21, and 30), the average<br />

hepatic copper rise was of about þ61% in the severely arthritic rats, but only<br />

of about þ31% in those animals that manifested the pathology to a mild<br />

degree, these two data being significantly different in the cross Student’s t test<br />

evaluation (48). Thus, according to our results and in contrast with total<br />

serum copper and ceruloplasmin concentrations, the hepatic metal level


The Role of Copper in Inflammation 175<br />

status appears to be a consistent index of the overall importance of the<br />

inflammatory process, being able to clearly discriminate among acutely,<br />

and chronically weakly or seriously affected rats.<br />

The kidneys, together with the red blood cells, the liver, and to some<br />

extent the plasma, are compartments in which copper seems to be easily<br />

exchangeable and thus available to be moved to other tissues according to<br />

actual body needs (61). As a consequence, the study of copper status in<br />

the kidneys could provide useful information to better understand the effect<br />

of inflammation on this ‘‘endogenous’’ metal metabolism. Unfortunately,<br />

very few data are available, especially on the acutely inflamed animals, in<br />

which the renal copper level was found not to be influenced by the development<br />

of the carrageenan-induced pleurisy of the rat (37). On the contrary,<br />

the chronic inflammatory process appeared to cause a significant decrease<br />

of kidney copper levels, although other authors failed to confirm this evidence<br />

(4,48–50). One of the above papers described that, like hepatic copper<br />

increases, the decreases of both concentration and total amount of copper in<br />

kidneys were not only statistically significant during the asymptomatic<br />

phase of the rat AA (day 3), but also dependent on the seriousness of disease<br />

development, and reached their maximum level in the severely arthritic rats<br />

30 days after complete-adjuvant injection ( 48% compared to the total<br />

metal amount measured in the time- and age-matched, healthy controls)<br />

(48). Whether or not the decrease in total copper amount observed in the<br />

kidneys (which, differently from the liver, cannot be promptly supplied by<br />

metallo-element from the digestive tract) may be the expression of the need<br />

for a physiological mobilization of copper towards extrarenal biological<br />

pathways more directly involved in the control of the inflammatory process<br />

is presently unknown.<br />

Occasionally, but limited to the rat AA model, the concentration of<br />

copper in compartments other than blood, liver, and kidney has been studied.<br />

Thus, copper levels were found to be increased in the brain, stomach,<br />

bone, and pancreas, while, according to the same paper, they were unchanged<br />

in the heart and skeletal muscle (50). Other authors, however, found<br />

the concentration of this metal unaffected in the femur and in the brain<br />

(49,62,63). An increase of copper concentration and total amount was measured<br />

in the spleen of the adjuvant-arthritic rat, being once again directly<br />

related to the seriousness in the development of pathology (46). However,<br />

these increases actually may bear, per se, an uncertain biological meaning,<br />

since they were found to be mainly dependent on the increases of spleen<br />

weight and total serum copper levels, both of which were induced by the<br />

experimental pathology itself (48).<br />

Although very rarely evaluated, the analysis of copper status within<br />

inflamed fluids and tissues certainly deserves special attention. The evidence<br />

so far available shows that copper concentration in acute inflammations<br />

(Table 4) was found to be higher, compared with the noninflamed fluids


176 Milanino<br />

or tissues, in the inflamed and untreated aqueous humor of endotoxininjected<br />

rabbits, in the intrathoracic exudate caused by the injection of a<br />

sterile carrageenan suspension in the rat (in this instance, due to technical<br />

reasons, the comparison with the extremely small amount of fluid present<br />

in the thoracic cavity of noninjected animals was not done), and in the<br />

carrageenan-injected, inflamed rat paw (sectioned at the level of the tibiotarsal<br />

joint) (38,40,41); in the last case a significant inflammation-induced<br />

copper increase was observed, considering both the concentration and the<br />

total amount of metallo-element. Copper concentration was, instead, found<br />

unchanged in the colon of rats in which an experimental untreated colitis<br />

was induced (64). On the other hand, the intravenous (IV) injection of 67 Culabeled<br />

porphyrins in male rats showed a very remarkable uptake of the<br />

radioactive compound not only in the liver and kidneys but also in neoplastic<br />

tissue, when present and wherever localized in the animal body (65). In<br />

addition, the subcutaneous (sc) treatment with 64 Cu-labeled D-penicillamine<br />

of rats with an experimental sponge granuloma evidenced a selective accumulation<br />

of the radioactive metal within both the implanted sponge and<br />

the capsule that had surrounded it as a consequence of the organism’s reaction<br />

(66). Also in the AA rat model (Table 5), the inflamed hind paws (again<br />

sectioned at the tibio-tarsal joint) showed a statistically significant rise in<br />

copper concentration (and total amount), already evident during the asymptomatic<br />

phase (i.e., seven days after the challenge), and became dramatic<br />

when the disease reached its symptomatic stage (48,62,63). Like previously<br />

described for the liver and the kidneys, when the symptomatic phase was<br />

evaluated specifically, the copper status of the hind paws was clearly able<br />

to discriminate between slightly and severely affected animals (Table 7).<br />

Concerning the increased amounts of copper in the carrageenaninjected<br />

rat paw, a further point is worth emphasizing. The onset of the<br />

Table 7 Hind Paws Copper Status During the Symptomatic Phase of the<br />

Complete-Adjuvant-Induced Arthritis of the Rat<br />

Days after the<br />

inoculum<br />

14 þ26% a<br />

21 þ38% a<br />

30 þ32% a<br />

Mild disease Severe disease<br />

Cu (mg/g) Cu total (mg) Cu (mg/g) Cu total (mg)<br />

þ92% a<br />

þ50% a<br />

þ47% a<br />

þ39% a,b<br />

þ136% a,b<br />

þ106% a,b<br />

þ160% a,c<br />

þ183% a,c<br />

þ260% a,c<br />

Note: Statistics—Student’s t test.<br />

a P < 0.01 versus time- and age-matched noninflamed controls.<br />

b P < 0.01 versus copper concentration of time- and age-matched mild-disease group.<br />

c P < 0.01 versus copper total micrograms of time- and age-matched mild-disease group.<br />

Source: The data reported in the table are converted to percentage increases from the original<br />

values published in Ref. 48.


The Role of Copper in Inflammation 177<br />

acute inflammation is characterized by progressive leakage from the local<br />

vascular bed, of an inflammatory exudate made by a plasma-proteins-rich<br />

fluid (containing also ceruloplasmin) in which a great number of inflammatory<br />

cells (mainly leukocytes) are present (67). This phenomenon induces<br />

the formation of an edema that, when carrageenan is used as inflammatory<br />

agent, is already measurable one hour after the challenge, and, in most<br />

instances, reaches its highest values between three and about 4–8 hours later<br />

(68,69). Subsequently, the liquid fraction of the exudate is rapidly reabsorbed,<br />

whereas the cell fraction remains in situ for several days to carry out the<br />

repair and remission processes (67). If the data reported in Table 8 are taken<br />

into account, it clearly emerges that the statistically significant increase of<br />

paw-copper concentration precedes the hours in which the characteristic rise<br />

of total plasma copper (and ceruloplasmin) is measured. Moreover, the paw<br />

copper is still significantly elevated above control values 96 and 144 hours<br />

after the challenge, i.e., when the edema has almost entirely disappeared<br />

and the total plasma copper returned to normal. The same is also observed<br />

if the total amounts of paw copper are considered (data not shown).<br />

In the chronic processes, the edema present in the affected animals is<br />

mainly formed by a solid tissue made of different inflammatory cell types<br />

(such as leukocytes, lymphocytes, macrophages, fibroblasts, epithelioid cells,<br />

etc.) together with local necrotic cells and newly formed fibrous as well as<br />

synovial tissues; but, in contrast with acute inflammation, the serum-plasmaderived<br />

fluid component is much less prominent (5,67). Evaluating the status<br />

of copper in the hind inflamed paws of the arthritic rats, a scenario is observed<br />

Table 8 Percentages of Concentration Changes in Plasma and Paw Tissue Copper<br />

in Carrageenan-Inflamed Rats<br />

Time after the<br />

challenge (hours)<br />

Edema<br />

weight (g)<br />

Total plasma<br />

copper (mg/g)<br />

Paw copper<br />

(mg/g)<br />

0 0 0 0<br />

1 0.38 a<br />

4 þ12 a<br />

3 1.04 b<br />

2 þ20 b<br />

5 1.19 b<br />

3 þ19 b<br />

24 0.51 b<br />

þ74 b<br />

þ21 b<br />

48 0.44 a<br />

þ63 b<br />

þ26 b<br />

72 0.41 a<br />

þ55 b<br />

þ25 b<br />

96 0.23 þ8 þ25 b<br />

144 0.16 2 þ21 b<br />

Note: Statistics—Student’s t test versus the time- and age-matched controls.<br />

a P < 0.05.<br />

b P < 0.01.<br />

Source: From Refs. 37, 38, and 60.


178 Milanino<br />

similar to that described in the case of the acutely inflamed animals. In<br />

particular, as pointed out before, a significant increase of copper concentration<br />

and total amount are measured during the asymptomatic phase of the<br />

experimental disease (day 7; þ14%, P < 0.01), and, later on, these same parameters<br />

appeared to be very reliable markers of the severity in experimental illness<br />

(Table 7) (48,62,63). Interestingly, the whole blood copper amounts<br />

(calculated on the basis of body weight, according to the Altman and Dittmer<br />

formula) were found comparably higher throughout the experiment, being<br />

insignificantly different among them in the asymptomatic, slightly and seriously<br />

affected rats (48,70). Thus, all the evidence summarized above seems<br />

to suggest that the increase of copper, quantified in the acutely and chronically<br />

inflamed rat paw tissue, is not merely a consequence of the increased<br />

plasma copper levels, but, rather, the expression of a true and selective metal<br />

accumulation in this area. This event can, in turn, be seen as a signal revealing<br />

an increased local need for copper, which could have been induced by the<br />

reaction that the body promoted to counteract the inflammatory noxa.<br />

Finally, taking into account all the organism’s compartments in which<br />

acute or chronic inflammation-induced significant changes of the total copper<br />

amount were measured [i.e., the whole blood, liver, kidney, spleen, inflamed<br />

pleural exudates, and inflame paw (37,38,48,60,62,63)], we recalculated the<br />

comprehensive variations of the rat body copper content brought about by<br />

the inflammatory processes examined. The data obtained (Fig. 2) clearly show<br />

that: (i) both acute and chronic inflammatory processes induced an overall<br />

increase amount of the metallo-element in the organism; (ii) although the<br />

total whole blood copper content (which we know to be raised to comparable<br />

extents as response to any sort of inflammation) and the total content of kidney<br />

copper (that, as previously reported, was reduced in the AA of the rat)<br />

were taken into account in the calculation, the net accumulation of body<br />

copper reported in Figure 2 was found directly proportional to the severity of<br />

the experimental pathology studied. Importantly, the above-mentioned accumulation<br />

appeared to occur without depleting other compartments of the<br />

metal, such as the bone and the muscle, which are known to hold high<br />

amounts of copper (49,50,61). Finally, considering that the healthy adult rat<br />

contains about 2 mg/g of body weight of copper (71), the total copper content<br />

of the control animals that we used for our experiments ranged between 250<br />

and 420 mg. Thus, the statistically and biologically significant inflammationinduced<br />

increases of the metal reported in Figure 2 ranged between 5% and<br />

7% in the acutely inflamed rats, and may even approximate 25% in the case<br />

of the severely affected arthritic animals. In conclusion, as commented for<br />

the inflamed tissue, the above observation seems to suggest that the inflammatory<br />

challenge induced also a whole-body increased demand for copper,<br />

to be used in the onset and development of the physiological regulatory<br />

mechanisms devoted to keep the inflammatory process under a<br />

proper control.


The Role of Copper in Inflammation 179<br />

Accumulation<br />

of body copper<br />

(Total µg)<br />

56<br />

42<br />

28<br />

14<br />

0<br />

a<br />

Acute<br />

inflamm.<br />

a<br />

Chronic<br />

inflamm.<br />

Asymptom.<br />

Figure 2 Comprehensive copper accumulation in some rat body compartments relevant<br />

to the inflammatory process. Accumulation (expressed as the difference of total<br />

micrograms between inflamed and noninflamed animals) of total copper measured in<br />

the body compartments in which significant inflammation-induced variations of<br />

copper status were measured. The bars represent the net body increments of the<br />

metal promoted by the different experimental acute and chronic inflammatory processes<br />

studied in various rat models. Statistics (Student’s t test): a, P < 0.001 versus<br />

age-matched non-inflamed controls; b, P < 0.001 versus age-matched asymptomatic<br />

arthritic rats; c, P < 0.001 versus age-matched mild-disease arthritic rats. Source:<br />

From Refs. 37, 38, 48, 62, and 63.<br />

HUMAN SUBJECTS: STUDIES ON ‘‘ENDOGENOUS’’<br />

COPPER METABOLISM IN ACUTE AND CHRONIC<br />

INFLAMMATIONS, WITH A PARTICULAR REFERENCE<br />

TO RHEUMATOID ARTHRITIS<br />

The inflammation-induced changes of ‘‘endogenous’’ copper metabolism<br />

were also frequently studied in humans, focusing especially, though not<br />

exclusively, on rheumatoid arthritis (RA) (Table 9, and references therein).<br />

Due to obvious ethical reasons, the experimental approach to this problem<br />

was restricted to the body compartments in which an examination by nonor<br />

mini-invasive techniques was possible, i.e., in the plasma, red blood cells,<br />

urine, and, in few instances, the inflamed areas. The foremost aims of these<br />

a,b<br />

Chronic<br />

inflamm.<br />

Mild dis.<br />

a,b,c<br />

Chronic<br />

inflamm.<br />

Severe dis.


180 Milanino<br />

Table 9 Examples of the Changes of Copper Status Induced by Some Acute (A)<br />

and Chronic Inflammatory Diseases in Man (Cp ¼ Ceruloplasmin)<br />

Disease<br />

Serum<br />

total Cu<br />

Serum<br />

Cp<br />

Blood<br />

cells<br />

24 hours<br />

urine<br />

Inflamed<br />

area References<br />

Pneumonia (A) I — U — — 72<br />

Periodontal disease (A) I I — — I 2<br />

Tonsillitis (A) I — — — — 73<br />

Sandfly fever virus (A) I — — D — 74<br />

RA I U — — — 3<br />

RA I — — — I 75<br />

RA U — — — U 76<br />

RA U — — — — 77<br />

Ankylosing spondylitis I — — — — 78<br />

RA I — — I — 79<br />

RA I I — — I 80<br />

RA I I U — — 81<br />

RA I I — — — 82<br />

RA (juvenile) I I — — — 83<br />

RA I — — — I 84<br />

RA I — — — — 85<br />

RA I — I U — 86<br />

Psoriatic arthritis I — U — — 87<br />

RA I I U U — 88<br />

RA (juvenile) I — — — — 89<br />

RA I I I U — 59<br />

Abbreviations: I, significantly increased; U, unchanged; D, decreased; RA, human rheumatoid<br />

arthritis.<br />

researchers were: (i) to verify the hypothesis that a marginal copper deficiency<br />

may be a contributory factor in the etiology of rheumatoid arthritis;<br />

(ii) to validate the assumption that the development of a rheumatoid process<br />

could, in time, induce a depletion of the body copper stores; and (iii) to<br />

evaluate whether or not the copper status in serum could be taken as a<br />

reliable index of the severity of the chronic pathology studied (90–93). In<br />

particular, an experimental support to either or both the above (i) and (ii) theoretical<br />

propositions would offer a rational justification for the use of the<br />

‘‘exogenous’’ copper administration as an important metal re-equilibrating<br />

treatment, able to favor the amelioration, if not the resolution, of the<br />

rheumatoid condition.<br />

In humans, the existence of infectious, immune (or autoimmune), and,<br />

in general, any acute or chronic disease with a relevant inflammatory component<br />

(cancer included), is typically characterized by a significant increase of<br />

total serum (or plasma) copper and ceruloplasmin concentrations, these<br />

two parameters being highly and significantly correlated (59,80–83,94). As


The Role of Copper in Inflammation 181<br />

previously noted while discussing the animal models of inflammation, in the<br />

case of rheumatoid arthritis, in spite of the very high correlation existing<br />

between serum copper and ceruloplasmin levels, some authors found that<br />

the increase of the serum metal was essentially determined by a selective rise<br />

of the non-Cp-bound fraction of this parameter (3). This claim was proposed<br />

on the basis of the data obtained indirectly measuring the non-Cp-bound<br />

copper, i.e., calculating this value by subtracting from the total serum copper<br />

concentration the amount of the metal supposed to be carried by the ceruloplasmin<br />

itself. However, applying the same methodology, other authors<br />

confirmed that the serum metal increases were basically due to the rise of circulating<br />

ceruloplasmin (59,82). As it was already reported in this paper, a<br />

further and, perhaps, conclusive support for the latter evidence was obtained<br />

directly quantifying the actual amounts of non-Cp-bound copper in the serum<br />

of rheumatoid patients, in whom only minor variations of this circulating<br />

fraction of the metal were observed (55). Finally, considering as a whole the<br />

data summarized in Table 9, it appears that only very few studies reported<br />

the total serum copper or ceruloplasmin concentrations as unchanged, and<br />

none noted a decrease in the level of the above two parameters (3,76,77).<br />

Total serum copper and ceruloplasmin do not change significantly in<br />

relation to the dietary supply of this metal, unless a biologically significant<br />

copper-deficient alimentary regimen is adopted, or relevant amounts of<br />

intestinal copper-absorption inhibitors, such as zinc, iron, calcium, ascorbic<br />

acid, phitate, etc., are present in the food (56,71,95). Conversely, the ceruloplasmin<br />

concentration in the serum, and thus the total serum Cu, is strongly<br />

influenced by a great number of physiological factors, (e.g., hormonal levels)<br />

and pathological conditions (such as stress, inflammation, etc.) (17,56).<br />

On the other hand, the erythrocytes are considered to be a blood compartment<br />

in which copper is less susceptible to the hormonal as well as<br />

pathological stimuli, unless the pathology considered is directly concerning<br />

the erythrocyte itself (71,95). For this reason, the red blood cell copper level<br />

is taken to be a relatively more reliable index of the actual overall body copper<br />

status, at least as far as moderate deficiencies have to be identified (see<br />

below for further details) (71,95). In general, the erythrocyte copper concentration<br />

was found unchanged in this compartment of blood in RA<br />

patients (Table 9), with the exception of two studies carried out by the same<br />

research team that reported a significant increase of this parameter (59,86).<br />

However, this last claim turned out to be a misleading result since, as<br />

previously recalled in this paper, the apparent increase of the erythrocyte<br />

copper level (measured as mg/mL of metal in the packed cells) was actually<br />

due to the RA-induced significant decrease in the individual volume of red<br />

blood cell, and the overall amount of copper contained in this total fraction<br />

of blood [calculated according to the formula of Altman and Dittmer (70)]<br />

resulted not to be significantly different from that of the age-matched<br />

healthy controls (59).


182 Milanino<br />

Another compartment that may be considered a somewhat good index<br />

of a possible copper deficiency status is the 24-hour urine, albeit, as for the<br />

total serum copper, the level of urinary copper is sensitive to biologically significant<br />

body metal depletion, but not predictive of a marginal deficiency<br />

condition (71,95). This index was found to be decreased only in one case<br />

of acute inflammation [i.e., in the sandfly fever virus infection, carried out<br />

on healthy volunteers (74)]. In the rheumatoid arthritis patients, an early<br />

report of increased copper urinary excretion was published, whereas more<br />

recent works found this parameter unchanged, thus convincingly suggesting<br />

that RA does not promote a significant increase of body copper losses via<br />

this excretory pathway (59,79,86,88).<br />

To our knowledge, the copper status in an inflamed human solid tissue<br />

was studied only in patients suffering from acute periodontal disease, in which<br />

a 15-fold increase of the metal (measured as Cp levels) was found in the inflamed<br />

as compared to the normal periodontal tissue explants (2). A few more<br />

data exist on the copper content in the synovial effusions withdrawn from the<br />

actively inflamed joints of RA patients, compared with the fluids taken from<br />

the human osteoarthritic or traumatic knees used as control values<br />

(75,76,80,84). In one single case, the copper concentration of the rheumatoid<br />

synovial fluid was found to be insignificantly different from that measured in<br />

the osteoarthritic subjects, whereas in the other instances a dramatic increase<br />

of the metallo-element levels was measured in this particular compartment<br />

(75,76,80,84). We would like to stress that this evidence, together with that<br />

obtained evaluating the status of copper in human inflamed blood,<br />

closely recalls the behavior of the metal metabolism previously described when<br />

studying the development of acute and chronic inflammations in the<br />

laboratory animal.<br />

The data summarized in the above section of the paper, together with<br />

other published information, may help in the attempt to propose some<br />

reasonable answers to the questions with which this survey on the human’s<br />

‘‘endogenous’’ copper metabolism in inflammation (especially RA) has been<br />

introduced. The hypothesis that a marginal copper deficiency could contribute<br />

to the development of the rheumatoid arthritis was detailed by<br />

Rainsford and Sorenson (90–92). These authors reported that there was<br />

evidence suggesting that the dietary intake of copper in some Western countries<br />

might be well below that required for natural physiological functions.<br />

Moreover, it was pointed out that high levels of environmental pollutants,<br />

toxic metals such as lead and especially cadmium, might act as antagonists<br />

of copper absorption or affect the normal development of its biological<br />

pathways (91). However, although at least in the United States, the average<br />

intakes of copper may even be below the FDA’s recommended dietary<br />

allowances, there are still conflicting opinions about the practical need for<br />

concern over health in relation to copper status in humans (96). Moreover,<br />

this issue cannot be directly solved using the techniques available today for


The Role of Copper in Inflammation 183<br />

defining the existence of a marginal copper deficiency. In fact, the serum<br />

copper and Cp concentrations may well reveal a moderate or severe metal<br />

deficiency but are not at all responsive to marginal deficiencies (97,98). The<br />

same lack in the capacity to reveal a marginal copper-deficiency condition is<br />

true for both the 24-hour-urine metal excretion and the total copper red blood<br />

cell level (98). Conversely, the evaluation of a body copper marginal deficiency<br />

could, perhaps, be achieved by studying erythrocyte SOD 1 activity, although<br />

an early work reported that this parameter might increase in conditions in<br />

which an oxidative stress of any origin has been caused (97,99,100). More<br />

promising seems to be the measure of the cytochrome-c oxidase activity in<br />

the platelets. Actually, studies with rats show that this enzymatic marker is<br />

a sensitive sign of copper stores (101,102); the cytochrome-c oxidase<br />

activity in platelets highly correlates with copper concentration in the<br />

liver (r ¼ 0.99, P < 0.0001), an established indicator of copper status in<br />

animals (97). In conclusion, the hypothesis that a marginal copper deficiency<br />

status could favor the onset of the human rheumatoid arthritis cannot currently<br />

be either confirmed or denied. On the contrary, according to data<br />

available today, it seems reasonable to suggest that adequately nourished<br />

RA subjects do not develop a significant body copper depletion over time<br />

(59,85). In particular, studying the copper content of plasma, blood cells,<br />

and 24-hour urine in rheumatoid arthritis patients divided into four groups<br />

according to their disease duration, i.e., 0–1, 1–5, 5–10, and over 10 years,<br />

we have clearly shown that all the above-mentioned parameters were not significantly<br />

influenced by the duration of the disease itself (59). It seems worthwhile<br />

to stress that, at least for the urinary copper excretion and copper<br />

erythrocyte levels, a progressive decrease of these indices would have to be<br />

observed if the disease were responsible for a time-dependent biologically significant<br />

depletion of body copper stores (71,95). Consequently, it appears reasonable<br />

to exclude the need for a dietary copper replenishing therapy in<br />

response to the hypothesized RA-induced copper deficiency in humans suffering<br />

from rheumatoid arthritis. Finally, focusing on the possibility that the total<br />

serum (or plasma) copper could be taken as a predictive clinical marker in the<br />

rheumatoid patient, this speculation has been belied by recent work (59,85). For<br />

instance, plasma copper was found to correlate significantly with some other<br />

plasma indices of RA, such as the iron concentration (r ¼ 0.20, P < 0.050), total<br />

proteins (r ¼ 0.33, P < 0.010), a2-globulins (r ¼ 0.46, P < 0.010), c-globulins<br />

(r ¼ 0.22, P < 0.050), and ceruloplasmin (r ¼ 0.77, P < 0.001), but no significant<br />

correlations were found versus the well-established major clinical<br />

markers of the disease, i.e., the number of swollen joints, the grip strength,<br />

the functional class, the anatomical stage, and the physician’s assessment<br />

index (59). Interestingly enough, the above data confirm, adding more<br />

sophisticated details, the evidence already reported in this review when discussing<br />

the acute and chronic animal models of inflammation, which led us<br />

to propose that the increase of total serum (or plasma) copper is most


184 Milanino<br />

likely an ‘‘all or nothing’’ phenomenon, unable to discriminate between<br />

mild and severe inflammatory conditions.<br />

EFFECTS OF ‘‘EXOGENOUS’’ COPPER ADMINISTRATION<br />

ON THE INFLAMMATORY PROCESS<br />

The data summarized so far may be briefly outlined as follows:<br />

Dietary copper deficiency acts as a proinflammatory stimulus, at<br />

least in animal models of acute inflammation.<br />

In rats on a normal Cu-containing diet, the induction of acute and<br />

chronic inflammatory processes clearly produces a condition in<br />

which more copper is required by some compartments of the<br />

organism. In view of the fact that no major redistribution of ‘‘endogenous’’<br />

copper occurs, it seems reasonable to suggest that this<br />

increased demand for the metal is mainly fulfilled by enhanced<br />

intestinal absorption and/or decreased intestinal excretion of copper<br />

(37,57). Interestingly, it has been shown that turpentine edema<br />

increases the amount of dietary copper required to maintain hepatic<br />

SOD 1 levels equal to those of nonstressed rats (103). Moreover,<br />

albeit limited to blood, urine, acutely inflamed periodontal tissue,<br />

and rheumatoid synovial fluids, the data reported in humans would<br />

appear to give further support to the above hypothesis (57). Due to<br />

reasons that are not yet understood, however, the natural antiinflammatory<br />

effect promoted by increased intestinal absorption<br />

and/or decreased intestinal excretion of the metallo-element appears<br />

to be susceptible to significant improvement by the therapeutic<br />

administration of copper compounds (7).<br />

Thus, the use of ‘‘exogenous’’ copper preparations as remedies to counteract<br />

the inflammatory pathologies appears to consistently find its rationale in<br />

the above-summarized remarks. In turn, the study of anti-inflammatory potential<br />

of administered copper would help to better understand the overall network<br />

of relationships existing between this metal, both ‘‘endogenous’’ and ‘‘exogenous,’’<br />

and the onset, development, and remission of the inflammatory process.<br />

Problems Related to Routes of Copper Administration<br />

The final therapeutic goal regarding the use of copper preparations as<br />

anti-inflammatory agents is to identify effective and safe drugs to keep<br />

human rheumatoid arthritis, an important deabilitating disease for which<br />

a fully satisfactory treatment has not yet been found, under adequate control,<br />

if not to cure it (104). To reach this goal, the physician should not only<br />

consider an effective and safe copper compound, but also be able to administer<br />

this potential drug (which could be possibly used for long periods of<br />

time) by a nontraumatic route like oral or topical.


The Role of Copper in Inflammation 185<br />

Many reviews have been published summarizing the anti-inflammatory<br />

properties of over 140 different copper(II) complexes with a wide variety of<br />

ligands (92,105–108). Examination of the literature reveals that for the<br />

large majority of these copper-containing molecules the anti-inflammatory<br />

and/or antiarthritic activity was evaluated in animal models, following sc<br />

or intraperitoneal (ip) administration. In fact, sc and ip treatments are preferred<br />

to demonstrate and reproduce in vivo biological activity of copper.<br />

Copper(II) preparations given by any parenteral route tend to disappear<br />

rapidly from plasma, being mainly accumulated in the liver and, to<br />

some extent, in the kidneys (109). However, when these substances are<br />

administered orally, they may not enter the organism unchanged, and their<br />

copper content may not become bioavailable, unless very special Cu(II)<br />

ligands or particular vehicles and adequately high amounts of complexes<br />

are used. Most copper compounds, when exposed to the acidic pH in the<br />

stomach, actually undergo nearly complete dissociation, followed by formation<br />

and absorption of new, in situ formed Cu(II) complexes (110,111).<br />

Copper is then transferred from the portal circulation to the liver, which<br />

regulates its subsequent metabolism by:<br />

Storing the metal as Cu-thionein in the hepatic tissue, thus creating<br />

a physiological reservoir of the metal to be used according to<br />

actual physiological needs (56).<br />

Completing the synthesis of the copper-dependent proteins, a<br />

process in which a membrane copper P-type ATPase, the Wilsondisease<br />

protein (WND) (112), seems to be involved in delivering<br />

Cu(I) ions into the Golgi’s vesicles. In particular, the WND protein<br />

acts co-operatively with the ClC-4 chlorine channel (113) to assemble<br />

the olo-ceruloplasmin in this subcellular apparatus before the<br />

multifunctional protein is secreted into the general circulation.<br />

Finally, excreting the excess copper via the bile, once again<br />

utilizing the activity of the WND protein assisted by that of the<br />

Cu-chaperon Murr 1 (114).<br />

As previously stressed the Cp synthesis is not induced, during conditions<br />

of adequate dietary copper intake, by increasing the amount of the<br />

metal present in the diet, nor by supplementing the organism with copper<br />

given by any parenteral route (17,56,62,109). On the other hand, the<br />

homeostatic mechanisms regulating the Cp production and secretion appear<br />

to react exclusively to endogenous stimulations (17,56). Therefore, if the<br />

orally administered copper complex is broken down by gastric juice, the extra<br />

copper that enters the liver will, most likely, undergo hepatic first-pass<br />

clearance, becoming not bioavailable for any therapeutic purpose.<br />

Theoretically, topical administration of copper is the most convenient<br />

method of treating rheumatoid arthritis, because it may avoid or minimize<br />

the risks of systemic toxicity, and it may direct the drug straight to the


186 Milanino<br />

affected area, i.e., the inflamed joints. The main problem with such treatment<br />

is to identify both ligands and formulations that allow to carry therapeutically<br />

significant amounts of the active principle through the skin. This problem is<br />

made worse by the poor lipophilic nature of most Cu(II) compounds. Thus,<br />

the goal to get any important clinical success using topically applied copper<br />

preparations in the treatment of RA patients has not yet been accomplished.<br />

Last but not least, the iv route of administration has to be considered,<br />

although in recent times it only has been used in a few cases. Nevertheless,<br />

two of those deserve mention. In the first case, a number of copper-containing<br />

proteins was tested for their anti-inflammatory potential, and found to be<br />

active in an acute animal model of inflammation. In the second, and certainly<br />

in a more significant one, the antirheumatic action of a copper(II) complex<br />

with salicylic acid has been clinically evaluated in man. This latter study<br />

appears to bear even nowadays great interest, both theoretical and practical.<br />

We would like to open our survey on the effects of ‘‘exogenous’’ copper on the<br />

development and control of inflammation, reporting on iv administration of<br />

these metal-containing molecules.<br />

Intravenous Administration of Copper Compounds<br />

Laroche and coworkers (20) isolated a number of copper-dependent enzymes<br />

from different animal and vegetable sources, and iv-tested them for their<br />

acute anti-inflammatory activity in the laboratory animal, using yeastinduced<br />

paw edema model in the mouse (Table 10).<br />

As reported in the table, the copper proteins ceruloplasmin (isolated<br />

from beef erythrocytes) and laccase [isolated from a fungus belonging to<br />

the genus Polyporus (70)], which are both biochemically classified as ‘‘bluecopper<br />

oxidases,’’ appear to be the more active among the tested molecules.<br />

It is relevant to note that, as stressed by the authors to validate their results,<br />

Table 10 Examples of Anti-inflammatory Activity of Some Cu-Dependent<br />

Enzymes Administered iv in the Mouse, Challenged with a Subplantar Injection of<br />

a Yeast Suspension<br />

Copper-dependent enzyme Source Edema inhibition (%)<br />

Cu-serum albumin Human serum None<br />

Ceruloplasmin Beef serum 73<br />

SOD 1 Beef erythrocytes 44<br />

Diamine oxidase Pig kidney 35<br />

Cytochrome oxidase Pseudomonas aeruginosa 53<br />

Ascorbate oxidase Cucumis sativus 37<br />

Laccase Polyporus versicolor 72<br />

Abbreviations: SOD 1, cytoplasmatic Cu,Zn superoxide dismutase; iv, intravenous.<br />

Source: From Ref. 20.


The Role of Copper in Inflammation 187<br />

the anti-inflammatory activity was measured three hours after the challenge<br />

to which the iv treatment followed immediately. This makes it possible to<br />

use heterologous proteins without inducing any immune reaction in the<br />

experimental animal (20).<br />

As mentioned regarding intravenous use of copper complexes in<br />

humans, a 20-year-long clinical study demands attention. Hangarter (115)<br />

reported that in the treatment of arthritic diseases the therapeutic results<br />

with iv copper alone were comparable to those of chrysotherapy, although<br />

copper treatment did not give rise to the considerable side effects associated<br />

with gold-salts treatment. However, Hangater’s major efforts were dedicated<br />

to clinical evaluation of a copper complex marketed as Permalon TM .<br />

Chemically, the antirheumatic drug Permalon is a complex of one or two<br />

cupric ions coordinated by two or four salicylate molecules, i.e., Cu(II)-<br />

(salicylate)2 or Cu(II)2-(salicylate)4 (105,116). In the period between 1950<br />

and 1971, Hangarter (115,116) studied the activity of Permalon in 1147<br />

patients suffering from different rheumatic-degenerative diseases such as the<br />

arthrosis deformans, sciatica, erythema nodosum, lumbar spine syndrome,<br />

cervical spine–shoulder syndrome, acute rheumatic fever, and, particularly,<br />

rheumatoid arthritis. Focusing on the RA patients, the treatments with slow<br />

iv infusions of cupric salicylate followed the protocol reported in Table 11,<br />

where the clinical results obtained are also summarized.<br />

Commenting on the above results the author stated that, according<br />

to own experience, the antirheumatic effects obtained with intravenous<br />

cupric salicylate were considerably superior to those achieved with the<br />

antirheumatic agents commonly used at that time, such as nonsteroidal<br />

anti-inflammatory drugs (NSAIDs), antimalarial agents, gold salts, as well<br />

as cortisone preparations (116). Interestingly, during the open discussion<br />

that followed the presentation of that paper at an international symposium<br />

held in Little Rock (Arkansas) in 1981, it emerged that the RA patients<br />

who responded to the therapy with remission of the disease maintained<br />

Table 11 Protocol of Permalon Treatment Within 620 Rheumatoid Arthritis<br />

Patients, and Results Obtained by the Therapy—A 20-Year Study<br />

Treatment 6–10 infusions at intervals of 2–4 days<br />

Composition of each infusion Salicylate 6–8 g; copper 7.5–10.0 mg<br />

Salicylate: maximum dose for a full cycle 80 g, i.e., average of 2.5 g/die<br />

Copper: maximum dose for a full cycle 100 mg, i.e., average of 3.1 mg/die<br />

Therapeutic effect reported:<br />

Remissions 403 cases (65%)<br />

Significant improvements 143 cases (23%)<br />

No effects 74 cases (12%)<br />

Source: From Ref. 116.


188 Milanino<br />

their completely symptom-free status for a subsequent average period of<br />

three years (116). The therapy was occasionally accompanied by a transient<br />

nausea and tinnitus, both of which are very well-known minor side effects<br />

due to the administration of high salicylate doses (117). However, no other<br />

adverse reactions were observed during the treatments as well as the subsequent<br />

follow-up. In particular: (i) no disturbances were reported at the<br />

level of the gastrointestinal tract; (ii) the blood sugar, electrolyte levels, and<br />

the classical serum electrophoresis markers were maintained within the<br />

normal ranges; and finally, (iii) no evidence of cardiac, circulatory, renal, hepatic,<br />

respiratory, or central nervous system toxicities were found (115,116).<br />

Unfortunately, the above-reported results were obtained following the criteria<br />

of an open trial (i.e., in the absence of a double-blind comparison versus both<br />

sodium salicylate- and placebo-treated control groups); consequently, they<br />

cannot be taken as a valid report on the basis of clinical protocols of evaluation<br />

accepted today. In 1971, Prof. Hangarter retired; the production of<br />

Permalon was discontinued by the manufacturer for economic reasons<br />

(116), and since then no other research team decided to submit cupric salicylate<br />

to a currently officially approved clinical trial.<br />

Subcutaneous and Intraperitoneal Administration<br />

of Copper Compounds<br />

Table 12 shows a few representative examples of copper compounds, such as<br />

simple copper salts, copper-containing proteins, and copper complexes with<br />

widely different inorganic and organic ligands, which have been shown to<br />

possess significant anti-inflammatory and/or antiarthritic properties following<br />

sc or ip injections in the experimental animals. Notably, a number of<br />

these copper complexes are formed using NSAIDs as complexing agents.<br />

Although today well established at least in the animal models, the<br />

anti-inflammatory activity of copper compounds subcutaneously or intraperitoneally<br />

injected, but also given orally, has been the subject of a dispute<br />

originated by the actual irritating potential that copper ions could exert at<br />

the site of exposure (110,129). In the classical models of experimental acute<br />

inflammation, the compounds to be tested are administered one hour before<br />

the inflammatory challenge, and their activity is evaluated between three<br />

and seven hours after the challenge itself (68,69). Bonta and Noordhoek<br />

(130) showed that a preventive ip injection with a well-known promoter<br />

of acute inflammation, i.e., a carrageenan suspension, was able to strongly<br />

inhibit the normal course of the subsequent inflammatory reaction induced<br />

by subplantar injection of a kaolin preparation. The authors proposed<br />

that the challenge with an irritant (or potentially irritant) substance done<br />

at one site of the body could trigger the organism’s physiological antiinflammatory<br />

response, thus enabling the living system to reduce the degree<br />

of the inflammation later on induced by the same, or any other irritant,


The Role of Copper in Inflammation 189<br />

Table 12 Representative Examples of Copper Compounds Active as In Vivo<br />

Anti-inflammatory and/or Antiarthritic Agents, Following sc or ip Administration<br />

in the Animal (Rodents) Models of Inflammation<br />

Compound Administration route Reference<br />

Cu(II)Cl2 sc 118<br />

Cu(I)2O sc 119<br />

Cu(II) 2-(acetate) 4 sc 7<br />

Cu(OH)2CuCO3 [basic cupric<br />

sc 120<br />

carbonate (malachite)]<br />

Cu(I)-thiomalate sc 121<br />

Cu(II)-ascorbate sc 7<br />

Cu(II)-(anthranilate)2 sc 7<br />

Cu(II)2-(3,5-DIPS)4 sc 122<br />

Cu(II)-diaqutetrakis(p-cresotate)-(H2O) 2 ip 123<br />

Cu(II)-diaqutetrakis(o-cresotate)-(H2O) ip 124<br />

Cu(II)-histidine ip 125<br />

Cu(II)-tryptophan ip 125<br />

Cu(II)-cysteine ip 125<br />

Cu,Zn-SOD (from bovine liver) sc 126<br />

Cu(I)-thionein ip 127<br />

Cu(II)2-(salicylate)4 a<br />

sc 122<br />

Cu(II)2-(aspirinate)4 a<br />

sc 122<br />

Cu(II)n-(niflumate)2n-(H2O)n a<br />

sc 122<br />

a<br />

Cu(I)-D-penicillamine-(H2O) 1.5 sc 122<br />

Cu(II)n-(fenamole)n-(acetate)2n a<br />

sc 122<br />

Cu(II)-piroxicam a<br />

ip 128<br />

a<br />

The anti-inflammatory activity of the copper complex is significantly greater than that of the<br />

parent ligand used in the experiment.<br />

Abbreviations: sc, subcutaneous; ip, intraperitoneal.<br />

injected at a remote site of the same organism; this phenomenon was labeled<br />

‘‘counter-irritancy’’ (130). Thus, the parenteral and oral irritation usually<br />

ascribed to copper suggested that the anti-inflammatory activity seen with<br />

the copper compounds could be explained on the basis of their ability to<br />

promote a ‘‘counter-irritant’’ reaction (129,131). However, this hypothesis<br />

was consistently disputed by other published data, according to which the<br />

‘‘counter-irritancy’’ of copper compounds was only negligibly responsible<br />

for their anti-inflammatory activity (7,132,133). Recently, we carried out<br />

an experiment in the attempt to better clear up this issue (the paper has been<br />

submitted for publication). Noninflamed female rats were subcutaneously<br />

injected with aseptic isotonic solutions of Cu(II)-(acetate)2 containing<br />

either 3.0 [i.e., a fully therapeutic dose (7,105)] or 0.3 mg/kg of copper.<br />

The animals were sacrificed three, seven, or 24 hours after treatment. Skin<br />

specimens of the injected area were removed to perform a full macroscopic


190 Milanino<br />

and microscopic histological examination, and the blood was collected to<br />

determine its plasma total copper concentration. The results obtained<br />

unequivocally pointed out that, at all considered times, the histology did<br />

not showed any significant sign of a local inflammatory response. Moreover,<br />

the plasma total copper remained unchanged throughout the experiment,<br />

compared with that measured in the saline-treated controls. In particular,<br />

this significant marker of inflammation did not increase 24 hours after<br />

the sc injection of either copper acetate solutions. The latter evidence seems<br />

to convincingly suggest that the treatments did not induce a systemic antiinflammatory<br />

reaction capable of interfering with the own anti-inflammatory<br />

potential of copper, further sustaining that the pharmacological action of<br />

the metal is real, and that it does not depend on any biologically relevant<br />

‘‘counter-irritancy’’ phenomenon.<br />

As mentioned in the introduction, Sorenson (7), in his classical paper,<br />

not only showed that many sc given copper-containing molecules are very<br />

active anti-inflammatory and/or antiarthritic agents, but also proposed a<br />

theory that was destined to open an active debate. Sorenson’s (92,107) idea<br />

was that the coordination compounds formed in vivo between the surplus<br />

of copper present in the plasma of inflamed animals or humans and the clinically<br />

used nonsteroidal anti-inflammatory agents could represent the active<br />

metabolites of these drugs, and moreover, Sorenson suggested that<br />

the superoxide radical disproportionation (i.e., SOD 1 mimetic activity)<br />

accounted for the anti-inflammatory action of these copper(II) complexes.<br />

Sorenson’s hypothesis was, however, challenged by coordination chemists<br />

who, mainly, if not exclusively, using a computer simulation approach,<br />

claimed that the complexes between copper(II) and salicylate, acetylsalicylate,<br />

as well as many other NSAID-type counter-anions, could not exist<br />

under plasma conditions since the ligands are incapable of effectively competing<br />

for the metal (108,134–136). Nevertheless, the original observations<br />

made over the years that copper chelates of anti-inflammatory drugs have<br />

greater potency as anti-inflammatory agents can still be demonstrated with<br />

a considerable number of compounds (137). In particular, in a variety of<br />

laboratory animal models, parenterally administered copper aspirinate<br />

showed to posses relevant anti-inflammatory activity beyond that provided<br />

by an equimolar amount of aspirin (137). Referring to aspirin, it seems appropriate<br />

to recall that oil–water partition coefficient studies have shown<br />

that copper aspirinate, likely in the form Cu(II)2(aspirinate)4, is 10 times<br />

more lipophylic than aspirin at comparable pH values of about 4.0 (108,138).<br />

Obviously, this implies that copper aspirinate, once entered into the organism,<br />

is facilitated in diffusing through the lipophilic cellular barriers and,<br />

thus, more bioavailable than aspirin. Another interesting observation was<br />

the result of the studies carried out with a compound closely related to the<br />

salicylate, i.e., the 3,5-di-isopropylsalicylate (3,5-DIPS), which, although<br />

has no anti-inflammatory activity as such, was shown to possess potent


The Role of Copper in Inflammation 191<br />

anti-inflammatory activity when complexed with Cu(II) ions (105). It was<br />

found that in plasma Cu(II)(3,5-DIPS)2/Cu(II)2(3,5-DIPS)4 forms stable<br />

complexes with human serum albumin (139,140). This, in turn, led researchers<br />

to suppose that other, either known or unknown endogenous copper<br />

ligands, could allow the existence of some Cu(II)-NSAID or non-NSAID complexes<br />

in vivo, by coordinating them in sort of a multimolecular adduct able to<br />

survive as such in plasma or in interstitial fluids. Finally, to further sustain the<br />

soundness of the Sorenson’s hypothesis, two other in vivo studies deserve to be<br />

mentioned. In the first, Korolkiewicz and coworkers (141) showed that copper<br />

aspirinate is significantly more active than an equimolar mixture of copper and<br />

aspirin, when given orally to inflamed rats. This observation seems to indicate<br />

that, in spite of any theoretical prevision obtained from computer simulation<br />

models, some copper aspirinate complex should survive intact at the acid pH<br />

of the stomach, and be absorbed by the animals treated. In the second study,<br />

Milanino and coworkers (142) demonstrated that a standard oral dose of<br />

indomethacin (i.e., 2 mg/kg) was remarkably active in reducing the carrageenan-induced<br />

rat paw swelling (average inhibition: 49%). However, the drug lost<br />

about 60% of its potency when administered to inflamed copper-deficient rats.<br />

Oral Administration of Copper Compounds<br />

As noted before, the oral route of administration of copper compounds<br />

appears to be inconvenient, in general it being improbable that these molecules<br />

would survive as intact Cu-complexes in gastric juice. Nevertheless,<br />

there are examples that show that this problem could be overcome (Table 13).<br />

Table 13 Some Representative Examples of Copper Compounds Active as In Vivo<br />

Anti-inflammatory and/or Antiarthritic Agents, Following Oral Administration in<br />

the Animal (Rodents) Models of Inflammation<br />

Compound Vehicle Reference<br />

Cu(I) 2O Mulgofen a<br />

131<br />

Cu(II)-(acetate)2 Isotonic solution 143<br />

Cu(II)-(diaminoethane)2-Cl2O Mulgofen a<br />

144<br />

Cu(II)-D-alanine Mulgofen a<br />

144<br />

Cu(II)2-(3,5-diisopropylsalicylate)4 Propylene glycol 92<br />

Cu(II)-bis(2-benzimidazolyl)thioethers Gum Arabic 145<br />

b<br />

Cu(II) 2-(aspirinate) 4 Starch mucilage 141<br />

Cu(II)2-(aspirinate)4 b<br />

Sunflower oil 146<br />

Cu(II)2-(benoxaprofen)2 b<br />

Sunflower oil 146<br />

Cu(II)-carbonate Diet 62<br />

a<br />

Polyoxyethylated vegetable oil.<br />

b<br />

The anti-inflammatory activity of the copper complex is significantly greater than that of the<br />

parent ligand used in the experiment.


192 Milanino<br />

One possibility is to use as ligand a structure able to form stable complexes<br />

with Cu(II) ions, thus obtaining a significant degree of resistance to<br />

acid attack in the stomach. As a matter of fact, that is the case for the<br />

unsubstituted bis(2-benzimidazolyl)thioether (NSN), which is able to bind<br />

Cu(II), forming (both in the solid state and in solution) an adduct in which<br />

the metal is penta-coordinated by the two benzimidazole N donors and the<br />

thioether moiety, all belonging to the ligand, as well as by one water molecule<br />

and one perchlorate group (145,147). Significantly, the Cu(II)-NSN<br />

is stable enough in vitro to be intact at over 90% of its original amount,<br />

at a pH ranging between 4.5 and 4.0 (145). Moreover, about 10% of the<br />

complex is still present as such, well above the in vitro addition of the stoichiometric<br />

quantities of HCl (i.e., two equivalents) required for the complete<br />

protonation of the ligand (pH values near 3.0) (145,147). Thus, as can be<br />

reasonably expected to be seen in the above evidence and the intrinsic<br />

anti-inflammatory potential of copper, the Cu(II)-NSN complex was shown<br />

to be orally active in both the acute (carrageenan paw edema; maximum<br />

inhibition 57%, P < 0.001) and chronic (adjuvant-induced arthritis; maximum<br />

inhibition 46%, P < 0.01) models of rat inflammation, whereas NSN<br />

alone was virtually devoid of any anti-inflammatory activity (145). Moreover,<br />

in the acute model the pharmacological response of Cu(II)-NSN<br />

showed to be clearly dose-dependent, thus validating the biological meaning<br />

of the anti-inflammatory (and antiarthritic) effect observed (145). Unfortunately,<br />

it was not possible to carry out further studies on this interesting<br />

molecule. Consequently, it remains unknown whether in vivo it acts as<br />

intact Cu(II)-NSN or it expresses its pharmacological activity upon transformation,<br />

by the organism’s metabolism, into a chemically different active<br />

molecule. Moreover, there is no information about its possible mechanism(s)<br />

of action as an anti-inflammatory agent, in vivo or in vitro. Conversely,<br />

another potentially strong copper-complexing molecule, i.e., the D-penicillamine<br />

(a drug still commonly used for the treatment of Wilson’s disease) (9), has been<br />

given orally, as Cu(II)-D-penicillamine complex to rats affected by the kaolininduced<br />

paw edema, but it remained inactive also at the very high dose of<br />

300 mg/kg (144).<br />

An alternative way to overcome the gastric barrier action is to use vehicles<br />

able to protect the integrity of the orally administered copper complex,<br />

thus assuring both its absorption and bioavailability. One such a vehicle<br />

appears to be sunflower oil. In fact, suspending the Cu(II)2(aspirinate)4 complex<br />

in sunflower oil, Rainsford (146) was able to show that 150 mg/kg of<br />

orally administered complex caused a 38% inhibition (P < 0.05) of the rat<br />

paw swelling (carrageenan-induced inflammation), whereas the same dose<br />

of aspirin alone was totally inactive. Similar results were also obtained studying<br />

the copper complex with another NSAID, i.e., the benoxaprofen (146).<br />

Thus, 100 mg/kg of Cu(II)2(benoxaprofen)4 exhibited a 53% inhibition of<br />

the edema, whereas 100 mg/kg of the ligand alone were fully inactive.


The Role of Copper in Inflammation 193<br />

Interestingly, both NSAID-copper complexes not only showed a significantly<br />

greater potency compared with the parent drugs, but they were also virtually<br />

devoid of the gastric ulcerogenic activity that characterized the benoxaprofen<br />

(upon repeated daily oral dosing), and in particular aspirin (146). Evidence<br />

that Cu-NSAIDs complexes have a much lower gastric toxicity than the<br />

parent compounds has been first reported and then repeatedly confirmed<br />

by Sorenson (92,105,107,122). Another vehicle that could favor the absorption<br />

of intact copper complexes following their oral administration is a<br />

polyoxyethylated vegetable oil named mulgofen. For instance, some copper<br />

complexes, such as the Cu(II)-(diaminoethane)2 and Cu(II)-D-alanine,<br />

resulted to be active inhibitors of the kaolin-induced rat paw edema model<br />

when suspend in mulgofen and given orally at 100 mg/kg (144). However,<br />

using the same vehicle, oral aspirin and copper aspirinate were both found<br />

to be inactive at the dose of 100 mg/kg (131). Nevertheless, regardless of<br />

the vehicle used, it seems reasonable to point out that a significant<br />

number of studies, which have been reviewed recently (92,148), showed that<br />

a remarkable number of copper complexes, and those formed using many<br />

NSAIDs as ligands, in particular, are active anti-inflammatory and/or antiarthritic<br />

agents not only after iv, sc, and ip dosing but also following oral<br />

administration, provided that sufficiently high amounts of the compounds<br />

are given.<br />

Since it was not possible to show a proinflammatory effect of dietary-induced<br />

copper deficiency on the adjuvant-arthritic rat (26,27,33), the<br />

behavior of the metal in this experimental chronic pathology was studied<br />

by carrying out an ‘‘opposite’’ experiment, i.e., examining the development<br />

of the AA in rats fed a copper-supplemented diet. Using this simple ‘‘dietary<br />

trick,’’ it was possible to examine the effects of the oral copper administration<br />

on the inflammatory process, excluding, at the same time, any possible<br />

biological actions of the ‘‘exogenous’’ ligands used to form the conventional<br />

copper complexes commonly studied as anti-inflammatory/antiarthritic<br />

drugs. According to a preliminary observation, a putative anti-inflammatory<br />

diet containing 200 ppm of copper (added as carbonate; control diet,<br />

Cu ¼5 ppm) was actually able to inhibit the development of the adjuvant<br />

arthritis ( 28%, P < 0.01) in the copper-supplemented rats after one month<br />

of ‘‘prophylactic’’ feeding (149). Thus, the approach of studying the pharmacotoxicology<br />

of the 200-ppm copper-supplemented animals was continued,<br />

evaluating the development of the AA in rats preliminarily treated with<br />

experimental diets containing 50, 100, or 200 ppm of copper (i.e., respectively,<br />

10, 20, and 40 times the standard copper requirements) (62,63,150).<br />

The results obtained from the toxicological analyses on noninflamed rats<br />

after 30 and 58 days of 200 ppm copper-supplemented feeding are reported<br />

in Figure 3 (relative to the changes of copper status in the plasma, liver,<br />

and kidneys) and in Table 14. The data plotted in Figure 3 show that the<br />

200 ppm Cu-supplemented diet was able to progressively and significantly


194 Milanino<br />

Figure 3 Status of copper in plasma, liver, and kidney of noninflamed female rats<br />

following 30 and 58 days of feeding with either a normal (Cu ¼ 5 ppm) or supplemented<br />

(Cu ¼ 200 ppm) copper diet. On the right side of each column referring to the<br />

dietary ‘‘copper-supplemented’’ rats (Cu ¼ 200 ppm), the percent increases of copper<br />

versus the time-matched control diet (Cu ¼ 5 ppm) are reported in absolute values.<br />

Statistics (Student’s t test): P < 0.01. Source: From Refs. 62, 63, and 150.<br />

increase the total copper content in the liver and in the kidneys above the control<br />

values, following 30 and 58 days of treatment. Conversely, as expected and<br />

repeatedly stated above, such pronounced copper supplementation did not at<br />

all influence the metal status in the plasma of the animals. It is worth stressing<br />

that, although the same trend described above was observed in the 50 and<br />

100 ppm Cu-supplemented rats, the increases in hepatic- and renal-copper<br />

measured were remarkably lower compared to those obtained with the<br />

200 ppm copper-treated animals, and throughout the experiment were not<br />

significantly different from the control values (61). Table 14 shows that the<br />

200 ppm copper-containing diet notably increased the total amount of<br />

copper present in the hind paws also, albeit this effect was remarkably more<br />

evident at day 30 than at day 58.<br />

However, possibly more interesting are the following evidences:<br />

Copper supplementation did not modify the status of the metal in<br />

the cell fraction of blood, as well as in the brain of the treated rats.<br />

Massive dietary copper supplementation did not change the zinc<br />

status in any of the compartments examined. This observation is<br />

very relevant since, on the one hand, the excess of alimentary


The Role of Copper in Inflammation 195<br />

Table 14 The Toxicology of the Non-inflamed Copper-Supplemented Rats<br />

Parameter<br />

Changes versus the<br />

normally fed rats<br />

(Cu ¼ 5 ppm)<br />

30 days 58 days<br />

Hind paw copper þ36% a<br />

þ17% a<br />

Blood cells and brain copper None None<br />

Plasma, blood cells, liver, kidney, brain,<br />

and hind paws zinc<br />

None None<br />

Hematocrit None þ3% b<br />

White cells (count, differential count) None None<br />

Red cells (count, mean erythrocyte volume) None None<br />

Platelets (count, mean platelet volume) None None<br />

Serum aspartate aminotransferase None None<br />

Serum alanine aminotransferase 16% b<br />

None<br />

Serum alkaline phosphatase None 33% b<br />

Serum creatinine None None<br />

Serum urea None None<br />

Hemoglobin 2% b<br />

None<br />

Sodium, potassium, chlorine, calcium, and CO2 None None<br />

Blood proteins (total and fractionated, albumin,<br />

a/c ratio)<br />

None None<br />

Macroscopic and microscopic examination of liver,<br />

kidneys, adrenals, hind paws, eyes, thymus,<br />

lymph nodes (cervical, mesenteric), salivary glands,<br />

esophagus, stomach, duodenum, jejunum, ileum,<br />

cecum, colon, rectum, spleen, pancreas, trachea,<br />

lungs, heart, aorta, skin, mammary glands,<br />

urinary bladder, prostate, ovary, thyroid,<br />

parathyroids, brain, pituitary, spinal cord,<br />

sciatic nerve, skeletal muscle, femur, and sternum<br />

None None<br />

Note: A summary of the parameters (other than those reported in Fig. 3, i.e., the plasma, liver,<br />

and kidney copper values) evaluated after 30 and 58 days of feeding with the ‘‘anti-inflammatory’’<br />

200 ppm copper-containing diet.<br />

a P < 0.01.<br />

b P < 0.05 (Students’ t test).<br />

Source: From Refs. 62 and 150.<br />

copper might induce an inhibition of the intestinal zinc absorption,<br />

and, on the other, zinc has been shown to be an ‘‘endogenous’’ as<br />

well as an ‘‘exogenous’’ anti-inflammatory agent (56,151).<br />

The parameters related to the main toxicological markers of the<br />

blood cell, blood proteins, and hematochemical conditions,<br />

revealed that they were not affected by the treatment.


196 Milanino<br />

The major indices of the hepatic and renal functions were found<br />

to be within normal ranges. This evidence bears particular significance,<br />

since both liver and kidney (together with the brain and, to<br />

some extent, the erythrocytes and bones), are well-known target<br />

organs for copper toxicity (9,152).<br />

Finally, the macro- and microscopic examination showed that all tissues<br />

listed in Table 14 have fully normal anatomical characteristics.<br />

As far as the antiarthritic activity is concerned, it should be pointed<br />

out that the tail injection of the complete adjuvant was done after 30 days<br />

of preliminary feeding with either the control (Cu ¼ 5 ppm) or any of the<br />

three copper-supplemented diets. The development of the chronic pathology<br />

was then followed for 28 days, during which the dietary regimen of each<br />

of the four groups of animals was kept unchanged (62,63,149). The results<br />

shown in Figure 4 indicate that the 50 and the 100 ppm copper-containing<br />

diets were not able to reduce the arthritic score at any considered time<br />

point (i.e., 14, 21, and 28 days after the inoculum). Conversely, the 200 ppm<br />

copper-supplemented diet, although ineffective at day 14 and 21, showed<br />

a noteworthy and biologically significant inhibition of the arthritic score<br />

% inhibition<br />

of arthritic score<br />

35<br />

28<br />

21<br />

14<br />

7<br />

0<br />

Cu = 200 ppm<br />

Cu = 100 ppm<br />

Cu = 50 ppm<br />

14 21 28<br />

Days after complete-adjuvant tail injection<br />

Figure 4 Effects of three different copper-supplemented diets on the development<br />

of adjuvant-induced arthritis in the rat. The plotted data represent the percent inhibition<br />

of adjuvant-arthritis scores measured in arthritic rats fed with diets containing<br />

10, 20, or 40 times the amount of dietary copper (Cu ¼ 5 ppm) present in the standard<br />

diet given to the adjuvant-arthritic control p < 0.001. Source: From Ref. 62.<br />

*


The Role of Copper in Inflammation 197<br />

measured at the end of the experiment (day 28). The pharmacological importance<br />

of the above results was confirmed when evaluating the total plasma<br />

copper concentration in the arthritic rats fed the control diet versus those<br />

kept on the 200 ppm copper-added one during the symptomatic phase of<br />

the experimental disease (day 14 and day 28). Figure 5 shows that, different<br />

from the AA-controls, the total plasma copper concentration in the 28-daysarthritic<br />

copper-supplemented animals was dramatically reduced, which, in<br />

turn, suggests that an amelioration of the overall clinical status of affected<br />

rats may have occurred. The above speculation is further sustained when<br />

% copper<br />

increase<br />

% copper<br />

increase<br />

150<br />

120<br />

90<br />

60<br />

30<br />

0<br />

150<br />

120<br />

90<br />

60<br />

30<br />

0<br />

AA day 14<br />

plasma<br />

AA day 28<br />

*<br />

hind paws<br />

plasma hind paws<br />

AA rats on copper normal diet AA rats on copper 40 times supplemented diet<br />

Figure 5 Percent increases of total copper content in the plasma and hind paws of<br />

adjuvant-arthritic rats, fed with either a standard (Cu ¼ 5 ppm) or copper-supplemented<br />

(Cu ¼ 200 ppm) diet. Each column represents the percent increase in total<br />

copper induced by chronic disease in affected rats, compared with their time- and<br />

diet-matched controls, at 14 (upper panel) or28(lower panel) days after challenge.<br />

Statistics (Student’s t test): all the values are statistically significant versus the respective<br />

control groups; however, emphasis has been put on the comparison between total<br />

plasma copper of arthritic copper-supplemented rats at day 14 and 28, P < 0.0001.<br />

Source: From Ref. 63.


198 Milanino<br />

taking into account other significant markers of the seriousness of the<br />

experimental disease (Table 15).<br />

For a correct understanding of the data reported in Table 15, it should<br />

be recalled that the paw weight and the circulating white blood-cell and<br />

blood-platelet numbers significantly increase in the AA animals; conversely,<br />

a remarkable decrease of total kidney-copper amount and total plasma-zinc<br />

concentration are brought about by the pathology, being inversely correlated<br />

to its gravity (5,48,57). It thus becomes evident that the tendency<br />

towards normalization shown by all the above-mentioned disease-induced<br />

metabolic and physical changes (the arthritic score and the total plasma<br />

copper concentration included) seems to testify in favor of the efficacy of<br />

the 200 ppm copper supplementation in counteracting the development<br />

of rat adjuvant arthritis. Finally, differently from plasma copper, the total<br />

amount of the metal contained in the hind paws of the arthritic animals<br />

(expressed as percent increases), does not seem to show biologically significant<br />

changes comparing the data of day 14 with those of day 28, in both<br />

copper-normal and -supplemented rats (Fig. 5). The maintenance of high<br />

level of copper in the inflamed area might be simply due to a slow clearance<br />

rate of the metal from this body compartment. However, considering that<br />

28 days after the complete-adjuvant injection the experimental pathology<br />

remains unresolved, it could be more likely that copper is kept within the<br />

affected site in order to favor the local processes of tissue repair. This<br />

hypothesis is supported by the evidence that the physiological equilibrium<br />

of connective tissue depends upon a correct functioning of the copperdependent<br />

enzyme lysyl oxidase (153–156). Moreover, a tripeptide-copper<br />

complex, i.e., the glycyl-L-histidyl-L-lysine-Cu(II) (GHK-Cu), which is<br />

naturally formed within the damaged tissue, appears to be also intimately<br />

involved in tissue regeneration and remodelling process (155,157–160).<br />

Table 15 Percent Differences of Some Markers of Adjuvant-Arthritis Severity in<br />

Normally Fed and 200 ppm Cu-Supplemented AA Rats<br />

Parameter<br />

AA in the 5 ppm<br />

copper-fed rats (%)<br />

AA in the 200 ppm<br />

copper-fed rats (%)<br />

Mean weight of hinds paws (g) þ58 þ30 a<br />

Kidney copper (total mg) 53 34 a<br />

Total plasma zinc (mg/mL) 47 16 a<br />

White blood cells (10 3 /mm 3 ) þ41 þ12 a<br />

Blood platelets (10 3 /mm 3 ) þ46 þ13 a<br />

Note: The values were measured at the end of the experiments (day 28).<br />

Total number of rats: 30–40 per group.<br />

a<br />

P < 0.001 (Student’s t test).<br />

Source: From Refs. 62 and 63.


The Role of Copper in Inflammation 199<br />

Percutaneous Administration of Copper Compounds<br />

Reports related to the concerns and merits of copper preparations topically<br />

applied as anti-inflammatory/antiarthritic agents are thoroughly discussed<br />

in other chapters of this book. Nevertheless, the issue of the treatment of<br />

systemic inflammatory pathologies with percutaneous copper merits concise<br />

mention also in this review. The efficacy of the cutaneous treatment with<br />

copper-containing preparations in the therapy of wounds and some skin diseases<br />

has been empirically known for more than 3500 years ago [papyri of<br />

Ebers and Smith, circa 1550–1450 B.C. (6)]. Nowadays, the fact that tissue<br />

repair requires the local presence of copper-dependent molecules is an<br />

experimentally well-established evidence (155). The copper-dependent<br />

enzyme lysyl oxidase belongs to these molecules, which catalyses the formation<br />

of the cross-linking compounds that bind together the peptide chain of<br />

collagen and elastin, and in so doing impart both support and elastic properties<br />

to the tissues (153). Furthermore, when the extracellular-matrix macromolecules<br />

are degraded by different pathological stimuli, the protein<br />

cleavage leads to a virtuous feedback mechanism, yielding a number of peptides,<br />

some of which act as potent signals for the repair and remodelling of<br />

the damaged tissue (161). In particular, one of these peptides, the GHK-Cu,<br />

appears to be extremely active in promoting the healing of excision wounds<br />

and the skin damages caused by scald-burns (both experimentally induced),<br />

probably by means of a multifaceted way of action (155,158). Notably, the<br />

Cu(II)-tripeptide mentioned above is also fully active following topical treatments<br />

(159). At least theoretically, it appears to be easy to obtain relevant<br />

therapeutic results giving selected copper-containing preparations by cutaneous<br />

route, in the case of open wounds and, perhaps, skin pathologies.<br />

However, it is much more difficult to obtain similar effects using the same<br />

route of administration on intact skin, as when one has to deal with chronic<br />

systemic pathologies, such as adjuvant-arthritis of the rat and the human<br />

rheumatoid arthritis. As reported above, in both chronic diseases the untreated<br />

organism tends to accumulate copper within the inflamed tissues and<br />

inflammatory exudates (48,75,80,84). According to the most currently accepted<br />

interpretation, this particular event, together with the increases of the<br />

total copper measured in some other body compartments, is likely devoted<br />

to contrasting and eventually defeating the chronic pathology (151). This<br />

hypothesis appears to be also supported by the observation that the concentrations<br />

of copper in the paws of the 28-days-arthritic rats is increased by about<br />

50% to 60% in both copper-normal and copper-supplemented animals, compared<br />

to their respective time-matched controls (62,63). However, only in the<br />

copper-supplemented arthritic rat, in which the absolute amounts of the metal<br />

contained in the hinds paws is about 1.5 times greater than that measured in<br />

the arthritic controls, a significant therapeutic effect was observed at the end<br />

of the experiment (62,63). Thus, achieving an increased copper concentration


200 Milanino<br />

within the inflamed site seems to be an important therapeutic goal using any<br />

potential anti-inflammatory and/or antiarthritic copper-containing molecule.<br />

The attempts at using percutaneous administration of copper in the<br />

treatment of inflammatory conditions began with the studies of Walker<br />

and coworkers (162,163), who claimed that wearing the copper bracelet<br />

could have some beneficial effects on rheumatoid arthritis patients, and also<br />

showed that the metallic copper might be actually dissolved by some sweat<br />

components and permeate the intact cat skin. However, the research on the<br />

anti-inflammatory potential of topically applied copper preparations was<br />

quickly reoriented toward the use of nonphysiological cupriphores, as soon<br />

as it became evident that nonlipophilic NSAIDs can form much more lipophilic<br />

molecules by complexing with copper, and at the same time, they<br />

proved to be more potent anti-inflammatory/antiarthritic agents compared<br />

to the parent compounds (7,107,108,138). A number of studies have been<br />

carried out using salicylic acid as cupriphore, although according to the<br />

authors, at least one other NSAID, phenylbutazone, has also been, successfully<br />

tested as a Cu(II) complex following percutaneous administration in<br />

inflamed rats and horses (164,165). Focusing on Cu(II) salicylate, an ethanolic<br />

preparation of this complex, registered as Alcusal 1 , has been the<br />

subject of active research, done both in the laboratory animals and humans.<br />

For instance, it was reported that topically applied Alcusal caused: (i) a significant<br />

relief of swelling and joint stiffness in patients with rheumatoid<br />

arthritis; (ii) suppression of established polyarthritis in rats; (iii) prevention<br />

of the inflammatory response to the injection into the rat hind paw of carrageenan,<br />

histamine, or hydroxyl apatite; (iv) minimal toxicity of the applied<br />

compound in treated rats (166,167). Similar anti-inflammatory results were<br />

obtained in laboratory animals, with Dermcusal 1 , a Cu(II) salicylate complex<br />

in dimethyl sulfoxide/glycerol, and the choice of this vehicle was<br />

claimed to favor the cutaneous absorption of the copper salicylate in comparison<br />

with its parent, ethanol-based preparation (Alcusal) (168). Further<br />

studies were carried out dissolving 64 Cu(II)-labeled salicylate in ethanol/<br />

dimethyl sulfoxide/glycerol (66). This preparation was topically applied to<br />

rats affected by two substantially different models of inflammation, i.e.,<br />

the sponge granuloma (of about six days duration and of proliferative character),<br />

and the carrageenan-induced foot edema (which lasts for a much<br />

shorter time, and is mostly edemic in nature) (69). This kind of copper salicylate<br />

preparations being already known for its anti-inflammatory activity<br />

(167–169), the authors focused attention on the body labeled copper distribution<br />

in the two models of inflammation. The data reported showed an<br />

increase of radioactivity in the kidneys, liver, spleen, adrenals, thymus,<br />

and serum from animals with long-lasting granulomatous inflammation. On<br />

the other hand, no biologically significant percent changes of 64 Cu-relative<br />

specific activity were observed in the tissues of rats with carrageenaninduced<br />

paw edema (66). Thus, especially in the case of the rats affected


The Role of Copper in Inflammation 201<br />

by sponge granuloma, it appeared that copper was actually absorbed from<br />

the skin and then distributed systemically. Although promising on the basis<br />

of the studies preformed with experimental animals, the anti-inflammatory/<br />

antiarthritic potential of the above copper salicylate preparations topically<br />

applied actually failed to be confirmed in humans. Their beneficial clinical<br />

activity in osteoarthritic patients and in subjects affected by sports injuries<br />

has been recently reviewed by Beveridge (164). The author based his claim<br />

quoting only two unpublished studies with the Alcusal gel, which were, moreover,<br />

carried out on a very limited number of patients (i.e., 20 and 19,<br />

respectively). Conversely, a recent randomized, double-blind, placebo-controlled<br />

trial done with topically applied copper salicylate gel on 93<br />

patients with osteoarthritis of the hip and knee (170) was unsuccessful in<br />

showing significant differences of the copper salicylate treatment compared<br />

with controls that received placebo. Furthermore, significantly more patients<br />

in the copper salicylate group reported adverse reactions, particularly<br />

skin rashes (17.0% vs. 1.7% of the placebo group), which caused the exclusion<br />

of these subjects from the trial (170).<br />

Nonetheless, the contradictory data so far reported on the percutaneous<br />

efficacy of copper in the therapy of human inflammatory conditions<br />

should, by no means, discourage the pursuit of studies aimed at finding a<br />

copper preparation able to efficiently penetrate the cutaneous barrier, and<br />

to show a significant anti-inflammatory and/or antiarthritic activity in<br />

humans. A recent study, in particular, continues in this effort. In fact,<br />

Hostynek et al. (171) examined, by sequential tape stripping, the diffusion<br />

of metallic pulverized copper through the human stratum corneum of intact<br />

skin in vivo. Copper content was determined (inductively coupled plasmamass<br />

spectroscopy) in 20 sequential strips taken from treated and control<br />

subjects, either under occlusive (air exclusion) or semiocclusive (access of<br />

air) copper applications, for periods of up to 72 hours. Focusing on 72-hour<br />

exposure, and there on strips from no. 15 to 20 (that reach the glistening<br />

epidermal layer, i.e., the vital layers of epidermis in intimate contact with<br />

the derma postcapillary vascular bed), the following evidence was obtained:<br />

(i) occlusive copper application let copper permeate the epidermis to a limited<br />

extent only; (ii) conversely, under semiocclusive conditions, the presence<br />

of both oxygen and some unidentified molecules contained in skin exudates<br />

oxidizes Cu(0), and the in situ formed complexes entered the skin accumulating<br />

within the stratum corneum in biologically significant amounts; and<br />

(iii) in particular, at 72 hours, an average of about 0.6 mg/cm 2 of copper<br />

was measured in each of layers from 15 to 20. Thus, assuming application<br />

of copper on a surface of circa 100 cm 2 (which is a reasonable area to be<br />

topically treated in the case of an inflamed knee joint), 72 hours after a single<br />

treatment the total amount of metal that will accumulate in the inner<br />

layer of epidermis (strip no. 20) will be equivalent to 60 mg of copper. Actual<br />

bioavailability of such a relevant quantity of the metallo-element is


202 Milanino<br />

unknown; however the evidence stated is a first, coming from direct measurements<br />

showing potential, biologically significant concentrations of ionic<br />

copper reaching selected anatomical locations in the body following percutaneous<br />

treatment. Although in all likelihood most of the metal that<br />

permeated the superficial skin layers under such experimental conditions<br />

will be lost due to the effect of epidermal desquamation, since it is complexed<br />

with molecules that are per se subject to the natural process of<br />

excretion, the above results underscore the possibility of successfully using<br />

the topical route of administration for copper.<br />

In our opinion, the main issues related to the possibility of effectively<br />

exploiting the topical route of administration for copper compounds may be<br />

briefly outlined as follows:<br />

The metal should be complexed by a ligand able to form a stable<br />

and ‘‘shielded’’ lipophilic chelate, thus in vivo allowing its penetration<br />

into the skin, and protecting the ‘‘exogenous’’ Cu(II) ions<br />

from the competition with the numerous potentially complexing<br />

molecules present in the tissue, at least during the diffusion of the<br />

complexes through the skin. This kind of approach could, at<br />

the same time, reduce the risk that ‘‘free’’ copper ions released<br />

from the complex may come into direct contact with the tissues,<br />

causing occurrence of adverse dermatological reactions (170,172).<br />

The ligand also has to be able to coordinate the metal by bonds strong<br />

enough to allow the distribution of the complex in the organism, without<br />

being so stable as to end up with a molecular structure actually<br />

containing copper that is a non-bioavailable. In fact, too stable<br />

chelates, such as those in vivo formed with tetrathiomolybdate<br />

(173,174), are not capable of exerting any pharmacological action<br />

except that of seriously depriving the treated organism of copper.<br />

Both at the local and systemic levels, the ligand has to be nontoxic.<br />

To this end, the natural amino acids and a number of di- or tripeptides,<br />

but also simpler structures such as Cu(II) acetate, appear to<br />

be the favorites, disregarding whether or not they are endowed<br />

with their own anti-inflammatory potential.<br />

Even though the choice of the ligand would have completely fulfilled<br />

the above-mentioned suggestions, it appears possible that many<br />

copper complexes could turn out to be insufficiently lipophilic in<br />

order to permeate intact skin and supply the target tissues with<br />

pharmacologically adequate amounts of the metal. Consequently,<br />

the use of proper vehicles in which to dissolve or suspend the complex<br />

is most likely an important part of the overall task. Aside from<br />

the excipients typically used (i.e., ethanol, dimethyl sulfoxide, etc.),<br />

a very attractive and new approach could be that of embedding the<br />

copper complex in liposomal vesicles (175). As a matter of fact,


The Role of Copper in Inflammation 203<br />

recent papers have shown the high efficiency of such carriers for the<br />

percutaneous administration of a variety of drugs, such as asthma<br />

medications, kanamycin, and indomethacin (176–178).<br />

COPPER ANTI-INFLAMMATORY ACTIVITY: HYPOTHESES<br />

EXPLAINING THE POSSIBLE MECHANISMS OF ACTION<br />

The essentiality of copper for humans was first recognized early in the past<br />

century, when Hart and coworkers (179) showed copper to be critical for<br />

erythropoiesis in the rat. Since then, it became apparent that, during their<br />

evolution, all the aerobic living systems have recruited specialized copper<br />

sites to provide an adequate electron transfer reactivity to the proteins destined<br />

to cope with oxygen (180,181). The facts that in vivo this transition<br />

metal so easily shifts from the Cu(II) to the Cu(I) oxidation state and vice<br />

versa as well as its ability to form stable complexes with electron-donor biomolecules<br />

are the basic reasons for the utilization of copper in the living<br />

oxygen-dependent organisms (182). Thus, copper is a crucial constituent<br />

of many redox enzymes as well as nonenzymatic biologically active molecules,<br />

the importance of which in maintaining physiological homeostasis<br />

of biochemical, cellular, and tissue functions is a well-established evidence.<br />

Table 16 lists some examples of copper-molecules, separated into two<br />

groups, depending on their prevailing enzymatic or nonenzymatic activity.<br />

Inflammation is a physiological reaction, aimed at defending the<br />

organism against exogenous as well as endogenous attack, and copper is<br />

deeply involved in this process. However, inflammation develops through an<br />

extremely complex and strictly interdependent network of single responses,<br />

in which, step by step, different cell types (such as mast cells, polymorphonuclear<br />

leukocytes, monocytes, endothelial cells, macrophages, lymphocytes,<br />

fibroblasts, etc.), and different mediators (such as, histamine, serotonin, arachidonic<br />

acid derivatives, nitric oxide, cytokines, oxygen free radicals,<br />

chemokines, complement factors, etc.) come progressively into play (200).<br />

Copper was shown to be intimately involved in this overall inflammation<br />

network, influencing the behavior of many inflammatory cells, as well as the<br />

metabolism and/or activity of numerous inflammatory mediators (200).<br />

However, providing a comprehensive treatise on this issue would go beyond<br />

the actual purposes of this review; consequently, comments are brief, outlining<br />

some representative examples of the possible mechanisms of action<br />

on which the anti-inflammatory activity of endogenous and/or exogenous<br />

copper may be based.<br />

Histamine Release and Activity<br />

It is generally accepted that histamine is the first chemical mediator to be<br />

released after the inflammatory stimulus, and it is also well established


204 Milanino<br />

Table 16 Examples of Copper-Dependent Molecules Involved in Maintaining<br />

Physiological Homeostasis and Ensuring Adequate Copper Management<br />

Copper molecules In vivo main functions References<br />

Enzymatic proteins<br />

Cytochrome c oxidase Terminal enzyme of the<br />

mitochondrial respiratory<br />

chain<br />

17, 183<br />

Cu,Zn superoxide<br />

Dismutation of superoxide 17, 184<br />

dismutase (SOD 1)<br />

anions<br />

Ceruloplasmin Oxidation of Fe(II) in plasma;<br />

distribution of copper to<br />

extra-hepatic tissues;<br />

antioxidant activity<br />

17, 185–187<br />

Lysyl oxidase Cross-linking of collagen and<br />

elastin in tissue regeneration<br />

153, 188<br />

Monoamine and<br />

Oxidative deamination of<br />

184, 189<br />

diamine oxidases<br />

histamine, serotonin, etc.<br />

Dopamine b-monooxygenase<br />

Catecholamine biosynthesis 190, 191<br />

Monophenol monooxygenase<br />

(tyrosinase)<br />

Melanin biosynthesis 192<br />

Cell surface monoamine Regulation of glucose uptake 193<br />

oxidase<br />

and cell adhesion<br />

Peptylglycine a- amidating Bioactivation of neuroactive 184, 194<br />

enzyme<br />

Nonenzymatic proteins<br />

peptides<br />

Metallothioneins (MTs) Copper storage and detoxification;<br />

stress proteins<br />

195<br />

Transcription factors (Mac 1, Regulation of gene transcription 182<br />

Amt 1, Ace 1, etc.)<br />

for SOD 1, catalase, MTs, etc.<br />

Copper chaperones (Atox1, Copper handling within<br />

196–198<br />

hCCS, hCox17, etc.), and different pro- and eukaryotic<br />

membrane-copper pumps cell types;<br />

(P-type ATPases)<br />

maintenance of copper<br />

homeostasis<br />

A-domains of the clotting Participation in blood<br />

182, 199<br />

factors V and VIII<br />

coagulation process<br />

that its main role in inflammation is to increase the vascular capillary bed<br />

permeability (201–203). According to some early research, a binuclear,<br />

hydroxyl-bridged Cu(II) complex is the active form of histamine (105). On<br />

the other hand, copper, as cofactor of the diamino oxidases, is essential<br />

in the process of histamine degradation (184,189). Furthermore, the metal<br />

seems to regulate the release of this bioactive molecule from the mast cells,


The Role of Copper in Inflammation 205<br />

after their stimulation by a wide variety of proinflammatory noxa (92).<br />

Nevertheless, other biological pathways, e.g., the triggering of the complement<br />

cascade, and bioactive molecules, e.g., the arachidonic acid products and nitric<br />

oxide, are know to play a very significant role in regulating the inflammationinduced<br />

increase of vascular permeability (31,67,203–205). Note that, in all<br />

the above-mentioned processes, some copper-dependent molecules may be<br />

involved directly or indirectly. Therefore, the ambivalent role of copper in<br />

the processes of histamine release, activity, and catabolism, has a yet unclear<br />

relevance in justifying the mechanism of the anti-inflammatory action of this<br />

transition metal, either endogenous or exogenous.<br />

The Arachidonic Acid Cascade<br />

The products of the cyclo-oxygenase pathways, i.e., prostacyclins, prostaglandins,<br />

and tromboxanes, are, since the late 1970s, well-known mediators<br />

of many different biological reactions (31). As far as inflammation is concerned,<br />

the prostaglandins among these molecules are likely to play an<br />

important role, and the inhibition of cyclo-oxygenase (COX-1 and COX-2)<br />

activities actually represents one of the foremost mechanisms of action of<br />

the large majority of NSAIDs (206,207). As previously noted, copper(II),<br />

used as simple salts such as CuSO4 and Cu(NO3)2, seems to be able to interact<br />

in vitro with the activity of these enzymes, altering the equilibrium in<br />

the production of the prostaglandins E2 and F2a, thus promoting an antiinflammatory<br />

effect (28,29). These early observations were later confirmed<br />

using rabbit kidney medulla slices, and rat peritoneal macrophages (208,209).<br />

Nonetheless, as described studying the severe copper-deficient rat, it was<br />

observed that metal deprivation did not have significant effects on either<br />

the lung COX-1 and/or COX-2 activity(ies), or on the reactivity of the copper-deprived<br />

gastrointestinal tissues to the exogenous administration of<br />

prostaglandin F2a and E2 (26). This, in turn, seems to suggest that endogenous<br />

copper might not play a direct biologically significant role in the<br />

arachidonic acid cascade. Moreover, the administration of some copper(II)<br />

complexes with non-anti-inflammatory ligands, such as the bis-pyridine,<br />

bis(2,4-dimethylpyridine), and bis(2,4,6-trimethylpyridine), showed to have<br />

a remarkable anti-inflammatory activity in vivo, but no effect on the prostaglandin<br />

biosynthetic pathways (210). Conversely, Cu(II)2(aspirinate)4 and<br />

Cu(II)2(indomethacinate)4 were reported to decrease the activity of cycloxygenases<br />

to a greater extent compared to the parent ligands (211), thereby<br />

strongly limiting the production of the arachidonic acid different endproducts,<br />

prostaglandins included (212). Thus, in view of the conflicting results<br />

described above, and considering also that the biosynthesis of the various<br />

arachidonic acid cascade products is qualitatively as well as quantitatively<br />

different in the different body tissues (207), whether or not the endogenous<br />

Cu(II) may exert a significant part of its physiological anti-inflammatory


206 Milanino<br />

effects also by regulating the arachidonic acid pathways, is an issue that<br />

still remains to be better clarified.<br />

Connective Tissue Metabolism<br />

Inflammation can be controlled by stimulating the repair processes that<br />

ultimately replace damaged tissue with new cells and extracellular matrix;<br />

when these processes are impaired, the inflammatory reaction may tend to<br />

perpetuate and worsen, eventually degenerating into chronic disease (153).<br />

The roles of the copper-dependent enzyme lysyl oxidase in the biochemical<br />

pathways of reconstruction of collagen and elastin, as well as the participation<br />

of the GHK-Cu in the process of tissue repair and remodelling, were<br />

already mentioned briefly (153,158). In particular, the above copper(II)<br />

tripeptide complex has been the subject of numerous investigations, which<br />

highlight many interesting features of this molecule: (i) GHK-Cu is normally<br />

present in human plasma, probably being in a dynamic equilibrium with its<br />

‘‘de-coppered’’ form GHK; (ii) both GHK and GHK-Cu are generated, in<br />

situ, by the proteolysis of inflammatory and extracelluar matrix proteins<br />

(due to the action of lytic enzymes released by activated phagocytes) during<br />

episodes of tissue damage; (iii) GHK-Cu has been found to be a potent chemoattractant<br />

selective for macrophages, monocytes, and mast cells, but<br />

has no activity in favoring the migration of other inflammatory cell types,<br />

such as neutrophils; (iv) GHK-Cu has a high superoxide dismutase activity,<br />

which contributes to the detoxification of the superoxide anions produced,<br />

particularly in the initial phases of inflammatory response; and (v) in the<br />

inflammation-damaged area both GHK and, especially, GHK-Cu induce<br />

the removal of tissue debris, capillary growth, differentiation as well as viability<br />

and axon outgrowth of neuronal cells, and, finally, the production of<br />

fibroblast-mRNA for the synthesis of matrix protein, such as collagen, elastin,<br />

proteoglycans, glycosaminoglycans, and decorin (158,213) (see also the<br />

website: http://www.skinbiology.com/copperpeptideregeneration.html). Thus,<br />

the evidence reported above seems not only to stress the remarkable biological<br />

importance of the endogenous copper in regulating the tissue repair<br />

process, but may also stimulate research aimed at finding out other copper<br />

peptides suitable to bind copper and able to act in vivo as new antiinflammatory<br />

and, especially, antiarthritic drugs.<br />

Regulation of Nitric Oxide Synthase Activity<br />

In vivo the enzyme nitric oxide synthase is devoted to the production of the<br />

highly bioactive nitric oxide molecule (N¼O). The physiological roles of<br />

nitric oxide are also expressed in decreasing the vascular tone, decreasing<br />

the platelet adhesion and aggregation, and increasing the cytotoxic activity<br />

of macrophages and neutrophils (205,214). Moreover, it has recently been<br />

shown that, in vitro, most of the noxious effects of interleukin-1 (IL-1) on


The Role of Copper in Inflammation 207<br />

the cartilage metabolism (i.e., decrease synthesis of extracellular matrix<br />

component, abnormal cell renewal, enhanced sensitivity of chondrocytes<br />

to oxidative stress, etc.) are probably mediated by the action of superoxide<br />

on nitric oxide that, in turn, acts on IL-1-stimulated chondrocytes (215).<br />

The authors speculated that a combined therapy with NO synthase inhibitors<br />

and antioxidant, e.g., Cu(II)2(3,5-DIPS)4 that seems to posses both<br />

activities, may be promising for full cartilage protection (215). The inflammatory<br />

reaction is one of the conditions in which an upregulation of nitric<br />

oxide synthase has been reported (205,216). It is noteworthy to remark that<br />

the anti-inflammatory agent Cu(II)2(3,5-DIPS)4 has been shown to have a<br />

significant downregulatory activity on the nitric oxide synthase in vitro,<br />

which would be a mechanism well compatible with the pharmacological actions<br />

of this complex (217,218). Moreover, in vitro copper selectively inhibits the catalytic<br />

activity of the constitutive nitric oxide synthase I in C6 glioma cells (219).<br />

Whether or not a regulatory role of endogenous copper on the activity of the<br />

nitric oxide synthases could also occur in vivo, is, at present, unclear. However,<br />

it has recently been observed that the copper-dependent monoamine oxidases<br />

seem to modulate the expression of the macrophage-inducible isoform II of this<br />

enzyme (220), which leaves this intriguing possibility open.<br />

Leukocyte Activity and Migration<br />

The activity of polymorphonuclear (neutrophils) and mononuclear leukocytes<br />

(simply referred to as PMNLs) plays a pivotal role in the development and<br />

control of both acute and chronic inflammations. Among other functions,<br />

the intense phagocytic activity of these cells is specifically oriented towards<br />

eliminating deleterious materials from injured tissue, such as bacteria and other<br />

microorganisms, neoplastic cells, cell debris, antigen–antibody complexes, nonbiotic<br />

foreign particles, etc. (221). Immediately following the noxa the vascular<br />

permeability increases locally, chemotactic/activating factors are released by<br />

the endothelial vascular and inflammatory cells (that are also resident in situ),<br />

as well as by the invading organisms and/or their degradation products, and,<br />

consequently, impressively high amounts of PMNLs are recruited at the<br />

inflamed site from blood (222). Following activation by the different chemoattractants,<br />

the circulating leukocytes are induced to penetrate into the injured<br />

tissue, where they are further activated by a variety of locally present molecules,<br />

and, consequently, prompted to carry out their typical defense functions,<br />

among which the microcosm of biological pathways expressed by the so-called<br />

‘‘respiratory burst’’ plays a primary role. Actually, both endogenous and exogenous<br />

copper are certainly involved in many different steps of the above chain<br />

of events, as well as in the overall inflammatory process (Table 17).<br />

During the PMNLs phagocytosis that occurs in the inflamed area,<br />

these cells dramatically increase (10–15-fold) their oxygen consumption<br />

within a few seconds after contact with the stimulating substance (respiratory


208 Milanino<br />

Table 17 Some Examples of Copper-Containing Molecules Involved in the<br />

Scavenging of Oxygen-Derived Free Radicals in Chemotaxis, Migration and Oxidative<br />

Burst of Polymorphonuclear (Neutrophils) Leukocytes<br />

Compound<br />

Activity on oxygen radicals<br />

scavenging, and other<br />

PMNLs reactions References<br />

SOD 1 Scavenging of superoxide 223<br />

Cu(I)-thionein Scavenging of superoxide and<br />

hydroxyl radicals<br />

127, 224<br />

Ceruloplasmin Scavenging of oxygen radicals 225<br />

Cu(II)-GHK Scavenging of superoxide,<br />

chemotactic activity, and<br />

stimulation of repair processes<br />

158, 213<br />

Cu(II)SO4; Cu(II)-H-<br />

(1-His-Gly)2OH<br />

Scavenging of superoxide 226<br />

Cu(II)-rutin Scavenging of oxygen radicals 227<br />

Cu(II)-bis-pyridine; Cu(II)- Inhibition of superoxide<br />

210<br />

bis(2,4-dimethylpyridine);<br />

Cu(II)-bis(2,4,<br />

6-trimethylpyridine)<br />

production<br />

Cu(II)2(3,5-DIPS)4-albumin Scavenging of superoxide;<br />

inhibition of superoxide<br />

production<br />

139, 140, 228<br />

Cu(II)-salicylate a ;<br />

Scavenging of superoxide 229<br />

Cu(II)-acetylsalicylate a<br />

Cu(II) (salsalate)2 a<br />

Scavenging of superoxide 230<br />

Cu(II) 2(3,5-DIPS) 4;<br />

Cu(II)2(aspirinate)4 a ;<br />

Cu(II)2(indomethacinate)4 a<br />

Inhibition of chemotaxis,<br />

231, 232<br />

migration, and<br />

superoxide production<br />

Cu(II)-piroxicam a<br />

Scavenging of superoxide;<br />

inhibition of migration<br />

128<br />

Cu(II)2(niflumate)4 a<br />

Inhibition of chemotaxis;<br />

superoxide production<br />

233<br />

Cu(II)2(dimethylsulfoxide)2-<br />

(mu-niflumate)4 a<br />

Inhibition of superoxide<br />

140<br />

production<br />

Cu(II)-carbonate in the diet Inhibition of ex vivo adhesion 63<br />

a<br />

Activity of the copper complex is significantly greater than that of the parent ligand used in the<br />

experiment.<br />

Abbreviations: PMNLs, polymorphonuclear (neutrophil) and mononuclear leukocytes, SOD 1,<br />

cytoplasmic Cu, Zn superoxide dismutase Cu(II)-GHK, glycyl-L-histidyl-L-lysine-Cu(II);<br />

DIPS, diisopropylsalicyclic acid.<br />

burst) (234). Inside the PMNLs phagosomes, the enzyme NAPH oxidase<br />

utilizes oxygen to give rise to large amounts of superoxide anions, which,<br />

in turn, leads to the formation of much more reactive oxyl radicals, such


The Role of Copper in Inflammation 209<br />

as singlet oxygen, hydroxyl radical, and hydroperoxyl radical, as well as<br />

hydrogen peroxide (155,234). This sustained production of noxious oxygen<br />

products (that are collectively called ‘‘reactive oxygen species,’’ ROS) has<br />

the obvious scope of opposing the disease-inducing actions of the ingested<br />

particles (either biotic or nonbiotic) during the physiological process of<br />

inflammation (235). However, the oxygen radicals, which the PMNLs’<br />

respiratory burst has generated, could be cytotoxic not only for the foreign<br />

inflammatory material but also for the PMNLs themselves, as well as for<br />

the adjacent tissue cells and matrix or interstitial- and exudate-fluid<br />

biomolecules (235,236). It has been proposed that the chemically nonspecific<br />

character of the ROS-induced reactions affects the cell membranes and the<br />

soluble or matrix biological structures, damaging them and also leading, at<br />

least theoretically, to the extemporary formation of endogenous antigens<br />

that could, in turn, favor the evolution of the acute inflammatory reaction<br />

toward its chronic phase (237). An early study showed that the attack of<br />

superoxide anions on the phagocytic cell membrane might initiate the<br />

arachidonic acid cascade (238). Furthermore, it was observed that the enzymatic<br />

oxidation of arachidonic acid, which ends with the formation of its<br />

active metabolites, generates free radicals, the hydroxyl radical in particular,<br />

as side products (239). In addition, the enzyme nitric oxide synthase may<br />

function to produce superoxide anions (205,240). Thus, a very relevant goal<br />

to be achieved for keeping inflammation under proper control appears to be<br />

the inactivation of the fairly large excess of ROS that may be released within<br />

the inflamed tissue. The enzyme that in vivo is responsible for scavenging<br />

superoxide anions is the SOD 1 molecule. This enzyme, however, not only<br />

is specific for superoxide but is also largely contained only inside the intact<br />

cells; as a consequence, it cannot counteract the other oxygen radicals, and<br />

its action outside the cell is probably a secondary one. Nevertheless, other<br />

natural copper-containing molecules, such as ceruloplasmin, Cu(I)-thionein,<br />

and Cu(II)-GHK, which may be present in the plasma-exudate and/or<br />

in situ produced during inflammation, could efficiently replace SOD 1 in<br />

scavenging superoxide as well as, at least indirectly, the other ROS (Table<br />

17) (155). Interestingly, when inflammation is localized in the skeletal muscle,<br />

the endogenous carnosine and homocarnosine, which may easily bind<br />

copper ions in situ forming stable complexes, are able to act as oxygen radical<br />

scavengers. This hypothesis was proposed on the basis of the evidence<br />

that both Cu(II)-carnosine and Cu(II)-homocarnosine can dismutate the<br />

superoxide anions released from the neutrophils activated by in vitro contact<br />

with phorbol miristate acetate (PMA) (241). It may be relevant to stress that<br />

all the above-summarized evidence could well explain the reason for the<br />

observed copper accumulation within the inflamed tissues of both acutely<br />

and chronically inflamed laboratory animals and humans, previously<br />

described in this review. Also, many small molecular weight copper complexes<br />

exogenously administered have been shown to disproportionate


210 Milanino<br />

superoxide anions and/or inactivate other oxygen radicals (Table 17)<br />

(218,242). Among these molecules are included structures such as some<br />

oligopeptides (not necessarily of endogenous origin), macrocyclic tetraanhydroaminobenzaldehyde,<br />

ethylenediaminetetra-acetate, as well as the<br />

NSAID piroxicam, which have all been shown to be unable to display<br />

any scavenging activity when tested noncomplexed with copper; the complexes<br />

between Cu(II) and all the above listed molecules are, in contrast,<br />

very active superoxide (or ROS) scavengers (128,227,243–245). Conversely,<br />

many other NSAIDs do show an SOD-mimetic activity of their own; this<br />

activity, however, is significantly and, sometimes, dramatically enhanced by<br />

their complexation with copper(II) ions (Table 17). Another biochemical<br />

trait that a relevant number of NSAIDs share with a group of pyridine<br />

derivatives, and which may be related to their mechanism of action, is<br />

the ability to inhibit the PMNLs respiratory burst, reducing the oxygen uptake<br />

of these cells and decreasing their superoxide and other ROS production; once<br />

again, this effect is significantly increased when the same substances are evaluated<br />

as Cu(II) complexes (Table 17) (218). Notably, a study by Nilsson (246)<br />

has shown that the inhibition of the respiratory burst by two copper compounds,<br />

i.e., the Cu(II)2(3,5-DIPS)4 and the Cu(II)SO4, may be primarily due<br />

to the modulation of the protein kinase C activity. According to Sorenson<br />

(107,218), this observation may be extended to other copper complexes of<br />

NSAIDs, such as Cu(II)2(aspirinate)4, Cu(II)2(indomethacinate)4, etc.; moreover,<br />

interference with the protein kinase C activity pathways of action also<br />

accounts for the inhibition of chemotaxis and/or migration shown by the above<br />

(and possibly other) Cu(II) complexes (Table 17).<br />

The crucial process of migration of circulating leukocytes into the<br />

inflamed site, which is restricted to the postcapillary venules of the affected<br />

area, is a complex network of cellular adhesion molecule interactions that<br />

involves also many inflammatory chemoattractants and mediators (either of<br />

‘‘self’’ or ‘‘non-self’’ origin) such as histamine, cytokines (IL-1, IL-6, tumor<br />

necrosis factor a, etc.), chemokines (IL-8, eotaxin, fractalkine, RANTES<br />

proteins, etc.), leukotriene B 4 and possibly other arachidonic acid derivatives,<br />

Cu(II)-GHK, platelet activating factor, complement factor C 5a, nitric<br />

oxide, ROS and oxidized low-density lipoproteins, bacterial lipopolysaccharides,<br />

bacterial formilated peptides, etc. (213,247,248). All these<br />

substances govern, in a time- and stimulation-mediated expression and<br />

activity, the different endothelial and inflammatory cells responses, which<br />

are aimed at creating the proper conditions for the recruitment and activation<br />

at the inflammatory site of the different cells types actually involved in<br />

the physiological onset, development, and control of inflammation. Summarily,<br />

the PMNLs migration can be divided into three distinct phases:<br />

The first one, which immediately follows the insult by the phlogistic<br />

agent, is the tethering of the flowing leukocytes to the


The Role of Copper in Inflammation 211<br />

endothelial vascular cells. This event is caused by the exposure on<br />

the outer surface of PMNL plasma membrane of the adhesion<br />

molecule L-selectin that strongly, but provisionally, binds to a yet<br />

unidentified receptor located on the endothelial cell surface (249).<br />

After tethering, the leukocyte begins to ‘‘roll’’ along the blood<br />

endothelial vessel, to eventually firmly adhere to the endothelium<br />

itself before migrating into the inflamed site. The process of<br />

rolling appears to be basically mediated by the expression of the<br />

endothelial surface molecules P- and E-selectins that bind to<br />

selectin-specific leukocyte-surface ligands, the best known of which<br />

is the P-selectin glycoprotein ligand-1. Arrival of P- and E-selectins<br />

on the scene has the effect of initiating and continuing the leukocyte<br />

rolling, then gradually slowing down, thus preparing the last phase<br />

of the entire migration process, i.e., the firm adhesion to the endothelial<br />

cell of the PMNLs and their subsequent extravasation (249).<br />

The processes of firm endothelial leukocyte adhesion and leukocyte<br />

transmigration towards the inner tissues of the inflamed site are<br />

also extremely complex and not yet fully understood. They imply<br />

the exposition on the leukocyte plasma membrane of a number of<br />

b-integrin adhesion molecules, in particular the b2-integrins LAF-1<br />

(CD11a/CD18) and Mac-1 (CD11b/CD18), as well as, possibly, of<br />

the L-selectin again (250,251). The structures mentioned above<br />

bind to some counterpart endothelial-expressed adhesion molecules<br />

such as the family of intercellular adhesion molecules (ICAMs),<br />

of which the ICAM-1 is the most relevant member, the vascular<br />

adhesion molecule-1 (VCAM-1), as well as the vascular adhesion<br />

protein-1 (VAP-1) (250–252).<br />

It has been proposed that the expression and activation of VAP-1<br />

could be the initial step in the pathway of firm leukocyte adhesion and<br />

transendothelial migration, at least in conditions of a blood flow velocity<br />

comparable with those that allow the binding of other adhesion molecules<br />

(251). In fact, in the absence or blockade of this protein in vitro, the normal<br />

interactions between the leukocyte adhesion molecules, such as the LAF-1,<br />

Mac-1, etc., with their endothelial counter-receptors, such as the ICAM-1,<br />

VCAM-1, etc., are remarkably prevented (50% inhibition), and the leukocyte<br />

transmigration significantly reduced (251,253). Recently, the above<br />

observations have been confirmed in vivo, showing that the deficiency or<br />

the blocking of VAP-1 remarkably inhibits the PMNLs from migrating into<br />

the inflamed sites, thus preventing the customary development of both acute<br />

and chronic inflammations in the mouse (254,255). A very relevant feature<br />

of the VAP-1 is that this endothelial protein is, at the same time, an<br />

adhesion molecule and a copper-dependent enzyme, i.e., a semicarbazidesensitive<br />

amine oxidase, also known under the acronym AOC3 (193,252,253).


212 Milanino<br />

The above biological activities of VAP-1 are exerted by two distinct extracellular<br />

domains of the protein, and are strictly interdependent since the<br />

specific inhibition of the amine oxidase activity also abolishes the ability of<br />

VAP-1 to bind to its, yet unidentified, leukocyte counter-receptor (193,252).<br />

Apart from the copper dependency of VAP-1 physiological role, it already<br />

was reported that the biological action of some inflammatory chemoattractants<br />

and mediators, such as histamine, arachidonic acid derivatives,<br />

GHK-Cu, nitric oxide, and ROS [that also cause the generation of the oxidized<br />

low-density lipoproteins (247)], was modulated by either endogenous<br />

and/or exogenous copper. Moreover, nontoxic amounts of copper sulfate or<br />

chloride have been shown to induce the expression of ICAM-1 on the<br />

surface of chondrocytes in situ in cartilage explant cultures; however, these<br />

copper-induced ICAM-1 molecules failed to promote the adhesion to IL-1activated<br />

peripheral blood monocytes (256).<br />

Another interesting evidence came from studies on the in vivo<br />

lysozyme secretion and ex vivo adhesion and superoxide anion production,<br />

by neutrophils isolated from rats fed a standard or a 200 ppm coppersupplemented<br />

diet, either healthy or affected by complete-adjuvant-induced<br />

arthritis (63). Notably, the lysozyme concentration was measured directly in<br />

the blood of the experimental animals, whereas the neutrophil adhesion and<br />

superoxide generation were evaluated in an ex vivo assay that utilized fetal<br />

bovine serum-coated plates, and exogenously added PMA as stimulant (257).<br />

The results showed that: (i) the dietary supplementation with copper did not<br />

change the blood lysozyme concentration, as well as the adhesion and superoxide<br />

production by the neutrophils isolated from the noninflamed rats;<br />

(ii) conversely, all the above parameters significantly increased in the adjuvant-arthritic<br />

animals of both dietary-treated groups; and (iii) nevertheless,<br />

albeit lysozyme secretion and superoxide generation were found comparable<br />

to those measured in the nonsupplemented arthritic rats, the ability of the<br />

‘‘inflamed’’ copper-supplemented neutrophils to adhere to the assay plates<br />

was markedly reduced (47%; P < 0.001) (Fig. 6). This last observation seems<br />

to suggest that exogenous copper, orally administered by means of a copper<br />

carbonate-integrated diet, could specifically and significantly impair the expression<br />

and/or functioning of the neutrophils’ adhesion molecules, which, in<br />

turn, may partly account for the antiarthritic activity of the 200 ppm coppersupplemented<br />

diet reported in the same experiment (63).<br />

Immune System Development and Reactivity<br />

Research mostly coming from trace element depletion studies clearly<br />

established the importance of an adequate copper (as well as iron, zinc,<br />

and selenium) dietary intake for protection of animals and humans<br />

in the process of host resistance to the invading pathogens (Table 18). In<br />

fact, a large body of concordant data support the following general


The Role of Copper in Inflammation 213<br />

Neutrophil<br />

adhesion<br />

(%)<br />

40<br />

30<br />

20<br />

10<br />

0<br />

*<br />

Diet 30 d Diet 44 d Diet 58 d<br />

AA inoculum AA 14 d AA 28 d<br />

AA not suppl.<br />

AA Cu suppl.<br />

Healthy Cu suppl.<br />

Healthy not suppl.<br />

Figure 6 Effects of normal (Cu ¼ 5.0 ppm) or supplemented (Cu ¼ 200 ppm) copper<br />

diet on the ex vivo adhesion of neutrophils isolated from either healthy and adjuvantarthritic<br />

rats. Percent of ex vivo adhesion of blood neutrophils isolated after 30 days<br />

(AA inoculum), 44 days (AA 14 days), and 58 days (AA 28 days) of feeding, from rats<br />

kept on normal- (Cu ¼ 5.0 ppm) and copper-supplemented (Cu ¼ 200 ppm) diets. Statistics<br />

(Student’s t test): P < 0.01, comparison between Cu supplemented, AA 14 and<br />

28 days, versus not supplemented, AA 14 and 28 days. Note that the adhesion percentages<br />

of neutrophils isolated from both Cu-normal and Cu-supplemented arthritic rats<br />

are significantly different (P < 0.01) from those measured in the respective control<br />

groups (i.e., normal- and Cu-supplemented healthy animals). Source: From Ref. 63.<br />

conclusions (258): (i) an insufficient copper supply is associated with the<br />

impairment of numerous activities of cells involved in both innate and<br />

acquired immune reactions; (ii) the observed alterations may be due to<br />

decreased activities of individual cells, reduction in the total number of<br />

active cells, or to a combination of the above two effects; (iii) the extent<br />

of the damage to the immune system could be sufficient to increase the host<br />

susceptibility to the exogenous (or endogenous) aggressions; and (iv) all<br />

negative effects of copper deficiency on immune reactivity can be reversed<br />

by restoring the normal copper status in the examined subjects.<br />

The evidence reported in Table 18 shows that an analytically measurable<br />

copper deficiency status impairs the overall inflammatory reactions,<br />

decreasing not only the phagocytic-cell ability to efficiently destroy the<br />

invading hosts but also seriously affecting the physiological activity of the<br />

whole battery of immune-competent organs (such as the thymus, bone<br />

*


214 Milanino<br />

Table 18 Some Representative Examples of Dietary Copper-Deficiency on<br />

Mammalian Immunity Functions<br />

Effects of copper deficiency References<br />

Decreased phagocytic and cytotoxic activity of<br />

32<br />

rodent-competent cells<br />

Reduced superoxide anion production and candidacidal<br />

259<br />

activity of animals neutrophils<br />

Increased susceptibility to transplanted leukemia<br />

260<br />

cells in the mouse<br />

Decreased resistance to the attack of different pathogens<br />

261<br />

in the mouse<br />

Impaired ex vivo activity of rat T cells to different stimulants 33<br />

Alterations of protein and lipid composition of murine<br />

262<br />

plasma membrane<br />

Reduced superoxide anion production and candidacidal<br />

263<br />

activity of peritoneal macrophages<br />

Decreased thymus weight and antibody titer in rodents,<br />

264<br />

characterized by a male versus female increased vulnerability<br />

Decreased rat splenic T-cell reactivity to mitogens, and<br />

265<br />

decreased number of helper and cytotoxic T-cell subset<br />

Decreased killing capacity of steer PMNLs 266<br />

Decreased proliferative capacity of rat splenocytes,<br />

267<br />

restored by interleukin-2 or copper addition<br />

Decreased proliferative ability of monocytes and increased<br />

268<br />

number of B-cells in the peripheral blood, in humans<br />

Decreased production of interleukin-2 by human T-lymphocytes 269<br />

Impaired interleukin-2 gene expression in a human<br />

270<br />

T-lymphocyte line (Jurkat cells)<br />

Abbreviation: PMNLs, polymorphonuclear (neutrophil) and mononuclear leukocytes.<br />

marrow, and spleen) and cells (such as the B- and T-lymphocyte cell lines).<br />

Notably, the above effects of copper depletion were significantly more<br />

pronounced in male versus female rats (264). Also, marginal conditions of<br />

copper deficiency (obtained in male rats fed with a 2.7 ppm coppercontaining<br />

diet) that do not involve evident changes of copper levels and<br />

ceruloplasmin activity in the serum nor significantly modify the organ (liver<br />

in particular) copper status are capable of markedly inhibiting the proliferative<br />

response of the splenic mononuclear cells to challenge by mitogens<br />

(271). In addition, the bioactivity of T-lymphocyte-produced IL-2 (which<br />

plays a central role in the regulation of the acquired immune responses)<br />

was dramatically reduced in those experimental conditions (270,271). More<br />

recently, it has been shown that, at least in the Jurkat cells, the decrease<br />

synthesis of IL-2 appears to be caused by the copper deficiency-induced inhibition<br />

of the expression of the NA-AT transcription factor (258,272). Much


The Role of Copper in Inflammation 215<br />

less data are available on the effects that copper supplementation may have<br />

on normal immune reactions. For example, the subcutaneous injection of<br />

Cu(II)2(3,5-DIPS)4 significantly stimulated the production of the different<br />

immune cell lines by the lymphoid tissues and the spleen, thus favoring an<br />

early recovery of injured mice after their exposure to 8 Gy whole-body<br />

irradiation (273). Furthermore, a significant inhibition of delayed-type hypersensitivity<br />

reactions was reported in mice fed with copper-supplemented<br />

diets, and this effect was also shown to be dependent on the amount of<br />

the metallo-element present in the diet as well as on the duration of the<br />

supplemented-feeding treatment (274).<br />

However, the leukocytes, such as neutrophils and monocytes, as well<br />

as the injured endothelia, actively participate in triggering the immune response,<br />

also by means of the production and coordinated action of numerous<br />

inflammatory mediators, such as ROS, nitric oxide, arachidonic acid derivatives,<br />

interleukins, chemokines, adhesion molecules, etc. (213,247,248,256).<br />

As we already stressed, albeit the production of superoxide and other ROS<br />

by tissue phagocytes has an essential role in counteracting the noxious<br />

actions of the invading pathogens, the persistence of an excess of these active<br />

oxygen products may be itself potentially dangerous for the organism. Thus,<br />

it has been previously proposed that an efficient control of the oxygen radicals<br />

generation could be an important goal in ensuring a proper control of<br />

the overall inflammatory reaction development. However, recent data may<br />

appear to challenge the above hypothesis. In fact, it has been reported<br />

that transgenic mice overexpressing SOD 1 showed a significant increase in<br />

delayed-type hypersensitivity reactions; conversely, the selective blockade<br />

of SOD 1 activity by means of disulfiram resulted in a significant decrease<br />

in the development of both delayed-type hypersensitivity reactions and<br />

adjuvant-induced arthritis in the rat (275). In other words, the above results<br />

would suggest that an overexpression of ROS could inhibit the development<br />

of the immune-mediated chronic phase of the inflammatory process, a final<br />

result comparable to that previously described as an effect of severe copper<br />

deficiency in the laboratory animals (26,27,33). Even so, a great number of<br />

copper complexes, both of endogenous as well as exogenous origin, are<br />

endowed with free-radical scavenging potential; furthermore, they are also<br />

able to inhibit the phagocyte respiratory burst, thus lowering the amount<br />

of ROS generated at the inflamed site (Table 17). These copper complexes<br />

have been unequivocally demonstrated to have significant anti-inflammatory<br />

and antiarthritic roles in vivo (Tables 10–13). Moreover, as previously<br />

detailed, the dietary deficiency of copper not only promotes an enhanced<br />

acute inflammatory reaction but also causes a dramatic decrease in the<br />

SOD 1 activity in the total blood cells (22,23). In addition, the development<br />

of acute- and chronic-inflammatory reactions is also critically influenced by<br />

the processes of chemotaxis, endothelial adhesion, and migration of the circulating<br />

inflammatory cells, the immunocompetent ones included (276).


216 Milanino<br />

Interestingly, a number of the copper complexes listed in Table 17 also have<br />

antichemotactic and/or antimigratory effects. Moreover, it has also been<br />

mentioned that the in vitro addition of copper induces the expression of<br />

nonfunctional ICAM-1 molecules on the chondrocyte surface, and the<br />

dietary-administration of high amounts of copper significantly impairs<br />

the ex vivo neutrophils’ ability to adhere to the assay plates, as well as<br />

remarkably inhibits the AA arthritis development in the treated rats (63,256).<br />

On the other hand, it has also been shown that the neutrophils isolated from<br />

copper-deficient mice expressed ‘‘ex vivo’’ a 50% reduced level of the adhesion<br />

molecule CD11b (i.e., Mac-1) on their surface (277).<br />

In conclusion, the experimentally observed activity of many copper<br />

complexes, which significantly ameliorate the conditions of organisms affected<br />

by chronic pathologies characterized by relevant inflammatory and<br />

immunological components, may depend upon a delicate balance in which<br />

the chemical nature of the copper complex, its actual bioavailability, the site<br />

and, especially, the preferred biological process of intervention, as well as the<br />

phase of development of the disease itself are all concurrently involved.<br />

CONCLUSIONS<br />

The evidence summarized seems to reasonably lead to the following general<br />

conclusions:<br />

The nutritional deficiency of copper significantly impairs the organism’s<br />

ability to develop a normal acute inflammatory reaction to<br />

counteract either biotic or nonbiotic attacks. Notably, this effect is<br />

strictly and simultaneously dependent on the amount of copper<br />

contained in the diet, on the length of treatment, as well as on the<br />

gender of the animals studied, males being remarkably more sensitive<br />

than females to dietary copper depletion.<br />

Both acute and, especially, chronic experimentally induced inflammations<br />

cause a net increase of the total copper in some body<br />

compartments, such as blood, liver, and the inflamed area. This<br />

accumulation of copper seems to be directly correlated with the<br />

severity of the pathology considered; furthermore, with the exception<br />

of the kidneys, it does not appear to involve any comprehensive<br />

and biologically significant redistribution of the metallo-element<br />

between the main body compartments. As a consequence, it is conceivable<br />

that the inflammatory process induces an overall enhanced<br />

requirement for copper, which may be accomplished increasing the<br />

metallo-element absorption/retention and/or decreasing its hepatic<br />

excretion, by the affected organism. It may not be secondary to<br />

underscore that a dramatic accumulation of copper has been reported<br />

also in the inflamed synovial fluid collected from rheumatoid


The Role of Copper in Inflammation 217<br />

arthritic patients, as well as of ceruloplasmin in the human inflamed<br />

periodontal tissue; these observations could imply that the need for<br />

more copper to better cope with the inflammatory pathologies may<br />

also characterize inflammations in humans.<br />

In spite of the previously described inflammation-induced increase<br />

in body copper levels, administration of extra amounts of<br />

copper, either by prophylactic feeding of animals with a metalsupplemented<br />

diet or by therapeutically treating them with<br />

exogenous copper salts or complexes, has clearly shown a significant<br />

anti-inflammatory and anti-arthritic potential in vivo.<br />

In general, copper may be considered an anti-inflammatory trace<br />

metal per se, as evidenced by the active participation of the endogenous<br />

metallo-element in the modulation of inflammatory reaction, by the protective<br />

effect of the dietary-supplemented copper on the development of the rat<br />

adjuvant-induced arthritis, as well as by the activity of therapeutically<br />

administered copper, observed regardless of the counter-anion used to carry<br />

it into the body. Nevertheless, the actual effects (i.e., the real potency) of<br />

exogenous copper as an anti-inflammatory and anti-arthritic drug have been<br />

shown to be significantly dependent on the route of administration chosen,<br />

as well as on the ligand used to form the complex. Focusing on the ligands,<br />

molecules that are endowed with their own anti-inflammatory activity,<br />

i.e., a significant majority of the clinically used NSAIDs, appear to potentiate<br />

their effects when administered complexed with Cu(II) ions. However,<br />

albeit a number of data seem to suggest that the observed anti-inflammatory<br />

activity is mainly due to the in vivo action of the intact complexes,<br />

the possibility that some Cu(II)-NSAIDs may also transfer their copper to<br />

one (or more) endogenous ligand(s) cannot be entirely ruled out. As a consequence,<br />

the final results would be, at least partly, obtained by the distinct<br />

effects of the NSAID given, and that of the complex(es) in vivo formed<br />

between the copper ions carried into the body by the drug and some endogenous<br />

ligand(s) of this essential metallo-element. An accurate survey of the<br />

data, which could explain the mechanisms of copper action in modulating<br />

the phlogosis’ development and remission, does not help much in solving<br />

the above problem, neither can it answer the question whether or not the<br />

endogenous and the exogenous metallo-element may somehow overlap in<br />

their anti-inflammatory actions. Actually, the whole scenery is obscured<br />

by the fact that the copper-dependent defense mechanisms (as well as those<br />

of NSAIDs) may simultaneously address different inflammatory pathways;<br />

their targets would also change according to the type of inflammation developed,<br />

as well as to its specific phase of progress. For instance, it has already<br />

been reported that copper has an unclear and, perhaps, secondary role in<br />

regulating the arachidonic acid cascade in vivo, whereas most of the<br />

NSAIDs can strongly inhibit this chain of reactions. Consequently,


218 Milanino<br />

administering a Cu(II)-NSAID complex, the observed influence on prostaglandins,<br />

prostacyclins, and tromboxanes production is, possibly, mainly<br />

due to the effect of the ligand alone. On the other hand, endogenous copper<br />

in the form of SOD 1, ceruloplasmin, Cu(I)-thionein, Cu(II)-GHK, and<br />

perhaps other in vivo existing copper complexes, has remarkable antioxidant<br />

properties. The same has been reported for some NSAIDs, and their<br />

complexation with copper has been described to potentiate these ROSscavenging<br />

effects. It is also well documented that many NSAIDs inhibit<br />

the respiratory burst and, consequently, the oxygen free radicals generation<br />

by activated phagocytes; once again their copper complexes appear to be<br />

more active, but a possible autonomous role for endogenous copper in this<br />

sequence of PMNLs reactions has not yet been recognized with certainty.<br />

The physiological processes of tissue repair and remodelling are known to<br />

be governed by copper-dependent enzymes in vivo (e.g., the lysyl oxidases),<br />

as well as by copper-oligopeptides complexes formed in situ [e.g., Cu(II)-<br />

GHK]; in this case it may be possible that the role of the endogenous copper<br />

is a predominant one, and the administration of exogenous copper might<br />

have the prevailing function of supplying the damaged tissue with the<br />

metallo-element. Finally, it is well established that the pathway of leukocyte<br />

migration towards target tissues represents a key step in the development<br />

and control of acute and chronic inflammation, and it also eventually<br />

involves the activation of the immune response. Some clinically used<br />

NSAIDs have an inhibitory effect on the inflammatory cells’ chemotaxis<br />

and migration, and their Cu(II) complexes clearly show a significantly<br />

enhanced activity. Recently, endogenous copper has been discovered to play<br />

a very remarkable role in the process of leukocyte rolling, adhesion, and<br />

extravasation, being an essential component of the multifunctional vascular<br />

adhesion protein-1, the activity of which appears to be crucial for normal<br />

leukocyte migration. Moreover, addition of simple Cu(II) salts to the assay<br />

medium in vitro promotes the synovial chondrocyte layers to express nonfunctional<br />

ICAM-1 molecules on their surface. Furthermore, neutrophils<br />

isolated from arthritic rats fed with a 200 ppm copper-supplemented diet<br />

have a significantly decreased ability to adhere to the test plates ex vivo,<br />

whereas the ‘‘parent’’ inflamed cells coming from animals fed a diet containing<br />

standard amounts of copper carry out this process normally. Thus,<br />

the examples summarized above clearly suggest that further research is<br />

needed to better understand which one, if any, of the copper-dependent<br />

processes examined could have a predominant relevance in explaining the<br />

natural anti-inflammatory activity of endogenous copper. Moreover, similar<br />

caution is also advisable in speculating on the possible biological pathways<br />

in which the endogenous and the exogenous copper activities may synergistically<br />

overlap.<br />

Finally, a last issue remains to be mentioned briefly, i.e., the potential<br />

toxicity of administered copper, a concern that, unfortunately, is still raised


The Role of Copper in Inflammation 219<br />

by many clinicians not accustomed with the action of copper compounds in<br />

the therapy of rheumatoid arthritis and other degenerative-inflammatory<br />

human diseases. However, existing data appear to clearly indicate that this<br />

concern is not justified. In fact, copper may have noxious effects only following<br />

the chronic oral (or parenteral) exposure to very large amounts of<br />

the metallo-element; in particular: (i) by chronic oral ingestion of foodstuff<br />

(e.g., water) if it supplies the organism with over 5 mg/kg of copper per day;<br />

and (ii) in the case of prolonged hemodialysis with apparatus that cause the<br />

introduction into the circulation of the metallo-element coming from<br />

copper-containing semipermeable membranes or copper tubing (278,279).<br />

Thus, a real risk of copper toxicity may exist only by administering amounts<br />

of the metallo-element certainly far greater than those that may be actually<br />

used for therapeutic purposes. In this context, it seems very relevant to recall<br />

that rheumatoid arthritis patients treated with a full cycle of Permalon TM<br />

did not show the appearance of significant adverse reactions, in particular<br />

none that could be due to iv administered copper (116). This evidence<br />

is noteworthy in that, although the antiarthritic activity of the above drug<br />

cannot be acknowledged in light of modern clinical evaluation protocols,<br />

the absence of significant adverse reactions reported is reliable, being largely<br />

obtained by means of objective measurements.<br />

In conclusion, the research oriented towards the study of the roles<br />

of copper in inflammation, and the utilization of copper compounds as<br />

anti-inflammatory and antiarthritic remedies, is by no means obsolete. In<br />

particular, the chance of targeting the copper preparations directly to the<br />

inflamed sites deserves notable attention. In fact, the percutaneous route of<br />

administration (using liposomes as copper carriers through the skin barrier)<br />

can both minimize the possible, although remote, toxicological hazards, as<br />

well as, especially, favor the accumulation of the metallo-element where its<br />

therapeutic characteristics may be better expressed.<br />

ACKNOWLEDGMENTS<br />

Review of the manuscript by Mrs. E. M. Hostynek-Welch and Dr. J. J.<br />

Hostynek is gratefully acknowledged.<br />

ABBREVIATIONS<br />

ppm part per million (mg/kg)<br />

CPE carrageenan-induced paw edema<br />

CP carrageenan-induced pleurisy<br />

AA adjuvant-induced arthritis<br />

SOD 1 cytoplasmatic Cu,Zn superoxide dismutase<br />

RA human rheumatoid arthritis<br />

sc subcutaneous<br />

(Continued)


220 Milanino<br />

ip intraperitoneal<br />

iv intravenous<br />

Cp ceruloplasmin<br />

NSAID nonsteroidal anti-inflammatory drug<br />

DIPS di-isopropylsalicylic acid<br />

NSN unsubstituted bis(2-benzimidazolyl)thioether<br />

GHK-Cu glycyl-L-histidyl-L-lysine-Cu(II)<br />

PMNLs polymorphonuclear (neutrophil) and mononuclear<br />

leukocytes<br />

ROS reactive oxygen species<br />

PMA phorbol miristate acetate<br />

IL interleukin<br />

ICAM-1 intercellular adhesion molecule-1<br />

VCAM-1 vascular adhesion molecule-1<br />

VAP-1 vascular adhesion protein-1<br />

REFERENCES<br />

1. Wintrobe MM, Cartwright CE, Gubler CJ. Studies on the function and metabolism<br />

of copper. J Nutr 1953; 50:395.<br />

2. Sweeney SC. Alterations in tissues and serum ceruloplasmin concentration<br />

associated with inflammation. J Dental Res 1967; 46:1171.<br />

3. Lorber A, Cutler LS, Chang CC. Serum copper levels in rheumatoid arthritis:<br />

relationship of elevated copper to protein alteration. Arthritis Rheum 1968;<br />

11:65.<br />

4. Karabelas DS. Copper metabolism in the adjuvant induced arthritic rat. Ph.D.<br />

thesis. Ann Arbor: Michigan State University, 1972.<br />

5. Rainsford KD. Adjuvant polyarthritis in rats: is this a satisfactory model for<br />

screening anti-arthritic drugs? Agents and Actions 1982; 12:452.<br />

6. Milanino R, Marrella M, Velo GP. Copper in the treatment of arthritic conditions:<br />

myth or reality? La chimica e l’Industria (C&I) 1994; 76:138.<br />

7. Sorenson JRJ. Copper chelates as possible active forms of the anti-arthritic<br />

agents. J Med Chem 1976; 19:135.<br />

8. Whitehouse MW. Ambivalent role of copper in inflammatory disorders. Agents<br />

and Actions 1976; 6:201.<br />

9. Scheinberg HI, Sternlieb I. Wilson’s disease. In: Major problems in internal medicine.<br />

Vol. XXIII in the series. Philadelphia: W.B. Saunders Company, 1984:126.<br />

10. Owen CA Jr, Hazelrig JB. Copper deficiency and copper toxicity in the rat. Am<br />

J Physiol 1968; 215:334.<br />

11. Milanino R, Passarella E, Velo GP. Adjuvant arthritis in copper deficient rats.<br />

Joint meeting of German and Italian pharmacologists, Venice, October 4–6,<br />

1977, Abstract 366.<br />

12. Fields M, Lewis CG, Antholine WE, et al. Hepatic iron overload may be responsible<br />

for the severity of copper deficiency in rats fed fructose. In: Anastassopoulou J,


The Role of Copper in Inflammation 221<br />

Collery P, Etienne J-C, Theophanides T, eds. Metal ions in biology and medicine.<br />

Vol. 2. Paris: John Libbey Eurotext, 1992:408.<br />

13. Milanino R, Mazzoli S, Passarella E, et al. Carrageenan oedema in copperdeficient<br />

rats. Agents and Actions 1978; 8:618.<br />

14. Denko CW. Protective role of ceruloplasmin in inflammation. Agents and<br />

Actions 1979; 9:333.<br />

15. Denko CW, Petricevic M, Whitehouse MW. Inflammation in relation to dietary<br />

intake of zinc and copper. Int J Tissue React 1981; 3:73.<br />

16. Conforti A, Franco L, Milanino R, et al. Copper and ceruloplasmin (Cp) concentration<br />

during the acute inflammatory process in the rat. Agents and Actions<br />

1982; 12:303.<br />

17. Linder MC. Biochemistry of copper. In: Chap. 4. New York: Plenum Press, 1991.<br />

18. Milanino R, Conforti A, Fracasso ME, et al. Concerning the role of endogenous<br />

copper in the acute inflammatory process. Agents and Actions 1979; 9:581.<br />

19. Kishore V, Wokocha B, Fourcade L. Effect of nutritional copper deficiency on<br />

carrageenan edema in the rat. A quantitative study. Biol Trace Element Res<br />

1990; 23:97.<br />

20. Laroche MJ, Chappuis P, Henry Y, et al. Ceruloplasmin: experimental antiinflammatory<br />

activity and physicochemical properties. In: Sorenson JRJ, ed.<br />

Inflammatory Diseases and Humana Press. Clifton, 1982:61.<br />

21. Milanino R, Passarella E, Velo GP. Copper and inflammation. In: Weissmann<br />

G, Samuelsson B, Paoletti R, eds. Advances in Inflammation Research. Vol. 1.<br />

New York: Raven Press, 1979:281.<br />

22. Bohnenkamp W, Weser U. 2Cu,2Zn-superoxide dismutase in copper depleted<br />

rats. In: Michelson AM, McCord JM, Fridovich I, eds. Superoxide and superoxide<br />

dismutases. New York: Academic Press, 1977:387.<br />

23. Williams DM, Lynch RE, Lee GR, et al. Superoxide dismutase activity in<br />

copper-deficient swine. Proc Soc Exp Biol Med 1975; 149:534.<br />

24. Brigelius R, Spoettl R, Bors W, et al. Superoxide dismutase activity of low<br />

mulecular weight copper (2þ) ion chelates studied by pulse radiolysis. FEBS<br />

Lett 1974; 47:72.<br />

25. Richardson T. Salicylates, copper complexes, free radicals and arthritis.<br />

J Pharm Pharmacol 1976; 28:666.<br />

26. Milanino R, Passarella E, Velo GP. Adjuvant arthritis in young copperdeficient<br />

rats. Agents and Actions 1978; 8:623.<br />

27. West GB. Diet and adjuvant arthritis in the rat. Int Archs Allergy Appl Immun<br />

1980; 63:347.<br />

28. Lee RE, Lands WEM. Cofactors in the biosynthesis of prostaglandins F 1a and<br />

F2a. Biochem Biophys Acta 1972; 260:203.<br />

29. Maddox IS. The role of copper in prostaglandin synthesis. Biochem Biophys<br />

Acta 1973; 306:74.<br />

30. Velo GP, Dunn CJ, Giroud GP, et al. Distribution of prostaglandins in inflammatory<br />

exudates. J Pathol 1973; 111:149.<br />

31. Giroud JP, Velo GP, Willoughby DA. The role of prostaglandins and cyclic<br />

nucleotides in inflammation. In: Berti F, Samuelsson B, Velo GP, eds. Prostaglandins<br />

and Thromboxanes. New York: Plenum Press, 1977:323.


222 Milanino<br />

32. Treagan L. Metals and the immune response. A review. Res Comm Chem<br />

Pathol Pharmacol 1975; 12:189.<br />

33. Kishore V, Latman N, Roberts DW, et al. Effects of nutritional copper deficiency<br />

on adjuvant arthritis and immuno-competence in the rat. Agents and<br />

Actions 1984; 14:274.<br />

34. Koj A. Acute-phase reactants and lysosomal enzymes in the blood of rats<br />

with experimental inflammation and radiation injury. Folia Biol 1970; 18:274.<br />

35. Powanda MC, Cokerell GL, Pekarek RS. Amino acid and zinc movement in<br />

relation to protein synthesis early in inflammation. Am J Physiol 1973; 225:339.<br />

36. Thompson HJ, Griminger P, Evans JL. Apparent increase in the requirement of<br />

trace minerals subsequent to severe trauma, Fed Proc 1976; 35:343.<br />

37. Milanino R, Cassini A, Conforti A, et al. Copper and zinc status during acute<br />

inflammation: studies on blood, liver and kidneys metal levels in normal and<br />

inflamed rats. Agents and Actions 1986; 19:215.<br />

38. Milanino R, Marrella M, Moretti U, et al. Copper and zinc status in rats with<br />

acute inflammation: focus on the inflamed area. Agents and Actions 1988; 24:356.<br />

39. Podhradska M, Podhradsky D, Andrisina J. Effect of inflammation on serum<br />

glycoproteins in rabbit. Folia Biol 1976; 22:312.<br />

40. McGahan MC. Copper and aspirin treatment increase the antioxidant activity<br />

of plasma. Agents Actions 1990; 31:59.<br />

41. McGahan MC, Fleisher LN, Grimes AM. Effects of copper depletion and<br />

D-penicillamine treatment on the ocular inflammatory response. Agents<br />

Actions 1991; 34:405.<br />

42. Ganrot K. Plasma protein response in experimental inflammation in the dog.<br />

Res Exp Med 1973; 161:251.<br />

43. Bieganski T, Blasinska MZ, Kusche J. Determination of histaminase (diamine<br />

oxidase) activity bt o-dianisine test: interference of ceruloplasmin. Agents<br />

Actions 1977; 7:85.<br />

44. Ward WF, Molteni A, Fitzsimons FJ, et al. Serum copper concentration as an index<br />

of lung injury in rats exposed to hemitorax irradiation. Radiat Res 1988; 114:613.<br />

45. Petryna LH. Copper content in the blood of irradiated animals. Fiziol Zh 2003;<br />

49:97.<br />

46. Cousins RJ, Swerdel MR. Ceruloplasmin and metallothionein induction by<br />

zinc and 13-cisretinoic acid in rats with adjuvant inflammation. Proc Soc Exp<br />

Biol Med 1985; 179:168.<br />

47. Oliva JC, Castell M, Queralt J, et al. Effect of chronic inflammation on copper<br />

and zinc metabolism. Rev Esp Fisiol 1987; 43:25.<br />

48. Milanino R, Moretti U, Concari E, et al. Copper and zinc status in adjuvant<br />

arthritic rats: studies on blood, liver, kidney, spleen and inflamed paws. Agents<br />

Actions 1988; 24:365.<br />

49. Néve J, Fontaine J, Peretz A, et al. Changes in zinc, copper and selenium status<br />

during adjuvant-induced arthritis in rats. Agents Actions 1988; 25:146.<br />

50. Kishore V. Effects of copper aspirinate and aspirin on tissue copper, zinc, and<br />

iron concentrations following chronic oral treatment in the adjuvant arthritic<br />

rat. Biol Trace Element Res 1990; 25:123.<br />

51. Fisher GL. Effects of disease on serum copper and zinc. Values in the beagle.<br />

Am J Vet Res 1977; 38:935.


The Role of Copper in Inflammation 223<br />

52. Feldman RF, Kaneko JJ, Farver TB. Anemia of inflammatory disease in the<br />

dog: clinical characterization. Am J Vet Res 1981; 42:1109.<br />

53. Kishore V, Boutte V, Fourcade L. Nutritional copper deficiency does not affect<br />

sponge granuloma formation in the rat. Biol Trace Element Res 1990; 25:115.<br />

54. Gitlin JD. Ceruloplasmin gene expression during inflammation. J Biol Chem<br />

1988; 263:6281.<br />

55. Freeman PC, O’Callaghan P. Decrease bioavailable copper in rat and human<br />

arthritic plasma. A rationale for copper therapy. Br J Pharmacol 1987; 90(proc<br />

suppl):49P.<br />

56. Cousins RJ. Absorption, transport, and hepatic metabolism of copper and zinc:<br />

special reference to metallothionein and ceruloplasmin. Physiol Rev 1985; 65:238.<br />

57. Milanino R, Marrella M, Gasperini R, et al. Copper and zinc body levels in<br />

inflammation: an overview of the data obtained from animal and human studies.<br />

Agents Actions 1993; 39:195.<br />

58. Milanino R, Marrella M, Pasqualicchio M, et al. Copper and zinc levels in<br />

inflammation. In: Berthon G, ed. Handbook of Metal–Ligand Interactions in<br />

Biological Fluids. Vol. 2. New York: Marcel Dekker Inc., 1995:1077.<br />

59. Milanino R, Frigo A, Barbara M, et al. Copper and zinc status in rheumatoid arthritis:<br />

studies on plasma, erythrocytes, and urine, and relationship with disease activity<br />

markers and pharmacological treatment. Clin Exp Rheumatol 1993; 11:271.<br />

60. Conforti A, Franco L, Milanino R, et al. Copper metabolism during acute<br />

inflammation: studies on liver and serum copper concentrations in normal<br />

and inflamed rats. Br J Pharm 1983; 79:45.<br />

61. Milanino R, Marrella M, Velo GP, Cristofori P, and Terron A. Regulation<br />

by copper of adjuvant-arthritis: a model of chronic inflammation especially<br />

suitable for studying the mechanism of copper anti-inflammatory activity. In:<br />

Rainsford KD, et al., eds. Copper and Zinc in Inflammatory and Degenerative<br />

Diseases. Dordrecht: Kluwer Academic Publishers, 1998:147–159.<br />

62. Albergoni V, Cassini A, Favero N, Rocco GP. Effect of penicillamine on some<br />

metals and metalloproteins in the rat. Biochem Pharmacol 1975; 24:1131–1133.<br />

63. Milanino R, Marrella M, Crivellent F, et al. Nutritional supplementation with<br />

copper in the rat. I. Effects on adjuvant arthritis development and on some in<br />

vivo- and ex vivo-markers of blood neutrophils. Inflamm Res 2000; 49:214.<br />

64. Al-Awadi FM, Khan I, Dashti HM, et al. Colitis-induced changes in the levels<br />

of trace elements in rat colon and other tissues. Ann Nutr Metab 1998; 42:304.<br />

65. Mercer-Smith JA, Cole DA, Roberts JC, et al. The biodistribution of radiocopper-labeled<br />

compounds. Adv Exp Med Biol 1989; 258:103.<br />

66. Beveridge SJ, Garret IR, Whitehouse MW, et al. Biodistribution of 64Cu in<br />

inflamed rats following administration of two anti-inflammatory copper complexes.<br />

Agents Actions 1985; 17:104.<br />

67. Hurley JV. Acute inflammation. Chapters 1, 10, 12. Edinburgh: Churchill<br />

Livingstone, 1972.<br />

68. Arrigoni Martelli E. Aspetti di farmacologia dell’infiammazione. Milano: Tamburini<br />

Editore, 1973:97.<br />

69. Swingle KF. Evaluation for antiinflammatory activity. In: Sherrer RA,<br />

Whitehouse MW, eds. Antiinflammatory Agents: Chemistry and Pharmacology.<br />

Vol. II. New York: Academic Press, 1974:33.


224 Milanino<br />

70. Altman PL, Dittmer DS. Blood volumes. In: Biology data book. Washington:<br />

Federation of American Societies for Experimental Biology, 1964:263, 514.<br />

71. Underwood EJ. Trace Elements in Human and Animal Nutrition. 4th ed. New<br />

York: Academic Press, 1977:56.<br />

72. Lahey ME, Gubler CJ, Cartwright GE, et al. Studies on copper metabolism—<br />

VII—blood copper in pregnancy and various pathological states. J Clin Invest<br />

1953; 32:337.<br />

73. Kryukova NA. Index of inhibition of ceruloplasmin activity under the effect<br />

of ascorbic acid in vitro, in acute inflammatory process. Lab Delo 1972; 31:153.<br />

74. Pekarek RS, Beisel WR. Redistribution and sequestering of essential trace elements<br />

during acute infections. In: Rodriguez JM, ed. Proc IX Int Congr Nutr<br />

Mexico (1972). Vol. 2. Basel: Karger, 1975:193.<br />

75. Niedermeier W, Griggs JH. Trace metal composition of synovial fluid and<br />

blood serum of patients with rheumatoid arthritis. J Chrom Dis 1971; 23:527.<br />

76. Pruzanski W, Russel LM, Gordon DA, et al. Serum and synovial fluid proteins in<br />

rheumatoid arthritis and degenerative joint disease. Am J Med Sci 1973; 265:483.<br />

77. Bajapyee DP. Significance of plasma copper and ceruloplasmin concentrations<br />

in rheumatoid arthritis. Ann Rheum Dis 1975; 34:162.<br />

78. Jayson MIV, Davis P, Whitcher JT, et al. Serum copper and ceruloplasmin in<br />

ankylosing spondylitis, systemic sclerosis, and morphea. Ann Rheum Dis 1976;<br />

35:443.<br />

79. Aaseth J, Munthe E, Forre O, et al. Trace elements in serum and urine of<br />

patients with rheumatoid arthritis. Scand J Rheumatol 1978; 7:237.<br />

80. Scudder PR, McMurray W, White AG, et al. Synovial fluid copper and related<br />

variables in rheumatoid and degenerative arthritis. Ann Rheum Dis 1978; 37:71.<br />

81. Banford JC, Brown DH, Hazelton RA, et al. Serum copper and erythrocyte<br />

superoxide dismutase in rheumatoid arthritis. Ann Rheum Dis 1982; 41:458.<br />

82. Conforti A, Franco L, Menegale G, et al. Serum copper and ceruloplasmin<br />

levels in rheumatoid arthritis and degenerative joint disease and their pharmacological<br />

implications. Pharmacol Res Commun 1983; 15:859.<br />

83. Hyorä H, Makela A-L, Pakarinen P, et al. Trace elements (copper, zinc and<br />

iron) in serum of rheumatic children living in south-western Finland. Acta<br />

Pharmacol Toxicol 1986; 59(suppl 7):403.<br />

84. Peretz A, Néve J, Vertongen F, et al. Synovial fluid copper, zinc and selenium in<br />

relation to inflammatory parameters in rheumatic diseases. In: Sorenson JRJ,<br />

ed. Biology of Copper Complexes. Clifton: Humana Press, 1987:583.<br />

85. Mussalo-Rauhamaa H, Konttinen YT, Letho J, et al. Predictive clinical and<br />

laboratory parameters for serum zinc and copper in rheumatoid arthritis.<br />

Ann Rheum Dis 1988; 47:816.<br />

86. Moretti U, Frigo A, Marrella M, et al. Copper and zinc in plasma, blood cells<br />

and urine of rheumatoid patients. Pharmacol Res Commun 1988; 20(suppl 1):19.<br />

87. Frigo A, Bamabara LM, Concari E, et al. Concerning the potential therapeutic<br />

value of zinc in rheumatoid and psoriatic arthritis. In: Milanino R, Rainsford<br />

KD, Velo GP, eds. Copper and Zinc in Inflammation. Dordrecht: Kluwer Academic<br />

Publishers, 1989:133.


The Role of Copper in Inflammation 225<br />

88. Peretz A, Néve J, Famey JP. Effects of chronic and acute corticosteroid therapy<br />

on zinc and copper status in rheumatoid arthritis patients. J Trace Element<br />

Electrolytes Health Dis 1989; 3:103.<br />

89. Bacon MC, White PH, Raiten DJ, et al. Nutritional status and growth in juvenile<br />

rheumatoid arthritis. Semin Arth Rheum 1990; 20:97.<br />

90. Klevay LM. An appraisal of current human copper nutriture. In: Sorenson JRJ,<br />

ed. Inflammatory Diseases and Copper. Clifton: Humana Press, 1982:123.<br />

91. Rainsford KD. Environmental metal perturbation, especially as they affect copper<br />

status, are factors in the etiology of arthritic conditions: an hypothesis. In:<br />

Sorenson JRJ, ed. Inflammatory Diseases and Copper. Clifton: Humana Press,<br />

1982:137.<br />

92. Sorenson JRJ. Copper complexes offer a physiological approach to treatment<br />

of chronic diseases. In: Ellis GP, West GB, eds. Progress in Medicinal Chemistry.<br />

Vol. 26. Amsterdam: Elsevier, 1989:437.<br />

93. Powanda MC. The role of endogenous mediator (endogenous pyrogen) in<br />

inflammation. In: Sorenson JRJ, ed. Inflammatory Diseases and Copper.<br />

Clifton: Humana Press, 1982:31.<br />

94. Schrauzer GN. Inorganic elements levels in neoplasia. In: Berthon G, ed. Handbook<br />

of Metal–Ligand Interactions in Biological Fluids. Vol. 2. New York:<br />

Marcel Dekker Inc., 1995:1061.<br />

95. Solomons NW. Zinc and copper in human nutrition. In: Kargiouglu ZA,<br />

Sarper RM, eds. Zinc and Copper in Medicine. Springfield: Charles C. Thomas<br />

Publisher, 1980:224.<br />

96. Pennington JA. Intakes of minerals from diets and foods: is there a need for<br />

concern? J Nutr 1996; 126(suppl 9):2304S.<br />

97. Milne DB. Copper intake and assessment of copper status. Am J Clin Nutr<br />

1998; 67(suppl):1041S.<br />

98. Olivares M, Araya M, Uauy R. Copper homeostasis in infant nutrition: deficit<br />

and excess. J Pediatr Gastroenterol Nutr 2000; 31:102.<br />

99. Milne DB, Johnson PE. Assessment of copper status: effect of age and gender<br />

on reference ranges in healthy adults. Clin Chem 1993; 39:883.<br />

100. Nielsen FH, Milne DB, Mullen LM, et al. Dietary sulfur amino acids, and<br />

genetic make-up or interindividual variation affect the response of men to<br />

copper depletion. J Trace Elemen Exp Med 1990; 3:281.<br />

101. Ralston NVC, Milne DB. Effect of dietary copper on platelet volume and cytochrome<br />

c oxidase in platelets. FASEB J 1989; 3:A357.<br />

102. Johnson WT, Dufault SN, Thomas AC. Platelet cytochrome c oxidase is an<br />

indicator of copper status in rats. Nutr Res 1993; 13:1153.<br />

103. Di Silvestro RA, Marten JT. Effects of inflammation and copper intake on rat<br />

liver and erythrocyte Cu–Zn superoxide dismutase activity levels. J Nutr 1990;<br />

120:1223.<br />

104. Pincus T Sokka T. Rheumatoid arthritis. In: Rakel RE, Bope ET, eds. Conn’s<br />

Current Therapy 2004. Philadelphia: Saunders Elsevier Inc., 2004:1021.<br />

105. Sorenson JRJ. Copper complexes. A unique class of anti-arthritic drugs.<br />

In: Ellis GP, West GB, eds. Progress in Medicinal Chemistry. Vol. 15.<br />

Amsterdam: Elsevier, 1978:211.


226 Milanino<br />

106. Crouch RK, Keusler TW, Oberley LW, et al. Possible medicinal uses of copper<br />

complexes. In: Karlin KD, Zubieta J, eds. Biological and Inorganic Copper<br />

Chemistry. New York: Adenine Press, 1985:139.<br />

107. Sorenson JRJ. Copper-potentiation of non-steroidal anti-inflammatory drugs.<br />

In: Berthon G, ed. Handbook of Metal–Ligand Interactions in Biological<br />

Fluids. Vol. 2. New York: Marcel Dekker Inc., 1995:1318.<br />

108. Dillon CT, Hambley TW, Kennedy BJ, et al. Copper and zinc complexes as<br />

anti-inflammatory drugs. Met Ions Biol Syst 2004; 41:253.<br />

109. Linder MC, Wooten L, Cerveza P, et al. Copper transport. Am J Clin Nutr<br />

1998; 67(suppl):965S.<br />

110. Perrin DD, Whitehouse MW. Metal ion therapy: some fundamental considerations.<br />

In: Rainsford KD, Brune K, Whitehouse MW, eds. Trace Elements in the<br />

Pathogenesis and Treatment of Inflammation. Basel: Birkhäuser Verlag, 1981:261.<br />

111. Williams DR, Furnival C, May PM. Computer analysis of low molecular<br />

weight copper complexes in biofluids. In: Sorenson JRJ, ed. Inflammatory<br />

Diseases and Copper. Clifton: Humana Press, 1982:45.<br />

112. Prohaska JR, Gybuna AA. Intracellular copper transport in mammals. J Nutr<br />

2004; 134:1003.<br />

113. Wang T, Weinmann SA. Involvement of chloride channels in hepatic metabolism:<br />

ClC-4 promotes copper incorporation into ceruloplasmin. Gastroenterology 2004;<br />

126:1157.<br />

114. Tao TY, Liu F, Klomp L, et al. The copper toxicosis gene product Murr 1<br />

directly interacts with the Wilson disease protein. J Biol Chem 2003; 278:41,593.<br />

115. Hangarter W. Die Salicylsaure und ihre Abkommlinge-Ursprung. Wiirkung<br />

und Anwendung in der Medizin. New York: Schattauer Verlag, 1974.<br />

116. Hangarter W. Copper-salicylate in rheumatoid arthritis and similar degenerative<br />

disease. In: Sorenson JRJ, ed. Inflammatory Diseases and Copper.<br />

Clifton: Humana Press, 1982:439.<br />

117. Rainsford KD. Side effects and toxicology of the salicylates. In: Rainsford KD,<br />

ed. Aspirin and Related Drugs. London and New York: CRC Press, 2004:367.<br />

118. Jackson GE, May PM, Williams DR. Metal–ligand complexes involved in<br />

rheumatoid arthritis. I. Justification for copper administration. J Inorg Nucl<br />

Chem 1978; 40:1189.<br />

119. Brown DH, Dunlop J, Smith WE, et al. Copper distribution and reactivity in<br />

serum following administration of cuprous oxide to rats and guinea pigs. Agents<br />

Actions 1979; 9:575.<br />

120. Bonta IL. Microvascular lesions as target of anti-inflammatory and certain<br />

other drugs. Acta Physiol Pharmacol Neerl 1969; 15:188.<br />

121. Withehouse MW, Walker WR. Copper and inflammation. Agents Actions<br />

1978; 8:85.<br />

122. Sorenson JRJ. Some copper chelates and their antiinflammatory and antiulcer<br />

activities. Inflammation 1976; 1:317.<br />

123. Sokolik J, Tumova I, Blahova M, et al. Anti-inflammatory activity of copper<br />

and zinc p-cresol dihydrate. Ceska Slov Farm 2000; 49:203.<br />

124. Sokolik J, Tumova I, Blahova M, et al. Anti-inflammatory activity of (o-cresotinate)<br />

copper and zinc aquacomplexes. Ceska Slov Farm 2002; 51:205.


The Role of Copper in Inflammation 227<br />

125. West GB. The copper problem and amino acids. In: Sorenson JRJ, ed. Inflammatory<br />

Diseases and Copper. Clifton: Humana Press, 1982:319.<br />

126. Huber W, Menander-Huber KB, Saifer MPG, et al. Studies on the clinical and<br />

laboratory pharmacology of drug formulation of bovine Cu,Zn-superoxide<br />

dismutase (Orgotein). In: Willoughby DA, Giroud JP, Velo GP, eds. Perspectives<br />

in Inflammation—Future Trends and Development. Lancaster: MTP Press Limited,<br />

1977:527.<br />

127. Miesel R, Hartmann HJ, Weser U. Antiinflammatory reactivity of copper(I)thionein.<br />

Inflammation 1990; 14:471.<br />

128. Sordelli DO, Fontana PA, Amura CR. Piroxicam-copper complexes: inhibition<br />

of polymorphonuclear leukocyte migration to Pseudomonas aerugionosa<br />

chemotactins in vivo and superoxide dismutase-like activity in vitro. Agents<br />

Actions 1993; 38:196.<br />

129. Rainsford KD, Whitehouse MW. Concerning the merits of copper aspirin as<br />

potential anti-inflammatory drug. J Pharm Pharmacol 1976; 28:83.<br />

130. Bonta IL, Nooerdhoek J. Anti-inflammatory mechanism of inflamed-tissue<br />

factor. Agents Actions 1973; 3:348.<br />

131. Lewis AJ. A comparison of the anti-inflammatory effects of copper aspirinate<br />

and other copper salts in the rat and guinea pig. Agents Actions 1978; 8:244.<br />

132. Brown HD, Smith WE, Teape JW. Antiinflammatory effects of some copper<br />

complexes. J Med Chem 1980; 23:723.<br />

133. Brown HD, Dunlop J, Smith WE, et al. Total serum copper and ceruloplasmin<br />

levels following administration of copper aspirinate to rats and guinea pigs.<br />

Agents Actions 1980; 10:465.<br />

134. Arena G, Kavu G, Williams DR. Metal–ligand complexes involved in rheumatoid<br />

arthritis. V. Formation constants for calcium(II)-, magnesium(II)- and<br />

copper(II)salicylate interactions. J Inorg Nucl Chem 1978; 40:1221.<br />

135. Furnival C, May PM, Williams DR. Models of low-molecular-weight copper(II)<br />

complexing equilibria in relation to rheumatoid arthritis. In: Rainsford KD,<br />

Brune K, Whitehouse MW, eds. Trace Elements in the Pathogenesis and Treatment<br />

of Inflammation. Basel: Birkhäuser Verlag, 1981:241.<br />

136. Brumas V, Brumas B, Berthon G. Copper(II) interactions with nonsteroidal<br />

anti-inflammatory agent. I. Salicylic acid and acetylsalicylic acid. J Inorg<br />

Biochem 1995; 57:191.<br />

137. Rainsford KD. Aspirin and NSAIDs in the prevention of cancer, Alzheimer’s<br />

disease and other novel therapeutic actions. In: Raisford KD, ed. Aspirin<br />

and Related Drugs. London and New York: CRC Press, 2004:707.<br />

138. Williams DA, Waltz DT, Foye WO. Synthesis and biological evaluation of<br />

tetrakis(acetylsalicylato)-mu-dicopper(II). J Pharm Sci 1976; 65:126.<br />

139. Shuff ST, Chowdhary P, Khan MF, et al. Stable superoxide dismutase (SOD)mimetic<br />

ternary human serum albumin-Cu(II) (3,5-diisopropylsalicylate) 2/<br />

Cu(II)2(3,5-diisopropylsalicylate)4 complexes in tissue distribution of the binary<br />

complex. Biochem Pharmacol 1992; 43:1601.<br />

140. Greenway FT, Hahn JJ, Xi N, et al. Interaction of Cu(II) 3,5-diisopropylsalicylate<br />

with human serum albumin—an evaluation of spectroscopic data. Biometals<br />

1999; 11:21.


228 Milanino<br />

141. Korolkiewicz Z, Hac E, Gagalo I, et al. The pharmacological activity of<br />

complexes and mixtures with copper and salicylate or aminopyrine following<br />

oral dosing in rats. Agents Actions 1989; 26:355.<br />

142. Milanino R, Marrella M, Moretti U, et al. Anti-inflammatory activity of indomethacin<br />

in copper deficient rats. Pharmacol Res 1992; 25(suppl 2):254.<br />

143. Laroche MJ. French patent N3, 1969: French patent M 6,518. Chem Abstr<br />

1971; 74:79,713.<br />

144. Lewis AJ, Smith WE, Brown DH. A comparison of the antiinflammatory activities<br />

of copper complexes in different models of inflammation. In: Rainsford<br />

KD, Brune K, Whitehouse MW, eds. Trace Elements in the Pathogenesis<br />

and Treatment of Inflammation. Basel: Birkhäuser Verlag, 1981:327.<br />

145. Milanino R, Concari E, Conforti A, et al. Synthesis and anti-inflammatory<br />

effects of some bis(2benzimidazolyl)<br />

thioethers and their copper(II) chelates, orally administered to<br />

rats. Eur J Med Chem 1988; 23:217.<br />

146. Rainsford KD. Development and therapeutic actions of oral copper complexes<br />

of anti-inflammatory drugs. In: Sorenson JRJ, ed. Inflammatory Diseases and<br />

Copper. Clifton: Humana Press, 1982:375.<br />

147. Bressan M, Morvillo A, Vel GP, et al. Concerning the chemistry and biology of<br />

some ‘‘copper-specific’’ ligabds and their Cu complexes active as oral antiinflammatory<br />

agents in the rat. In: Milanino R, Rainsford KD, Velo GP,<br />

eds. Copper and Zinc in Inflammation. Dordrecht: Kluwer Academic Publishers,<br />

1989:93.<br />

148. Sorenson JRJ. Cu, Fe, Mn, and Zn chelates offer a medicinal chemistry<br />

approach to overcoming radiation injury. Curr Med Chem 2002; 9:639–62.<br />

149. Moretti U, Marrella M, Pasqualicchio M, et al. Copper and zinc in inflammatory<br />

processes. Pharmacol Res 1990; 22(suppl 2):345.<br />

150. Cristofori P, Terron A, Marrella M, et al. Copper supplementation in the rat:<br />

preliminary observations on the clinical, haematological and histopathological<br />

profile. Agents Actions (Special Conf Issue) 1992:C118.<br />

151. Milanino R, Moretti U, Marrella M, et al. Copper and zinc in the development<br />

and control of inflammation. In: Berthon G, ed. Handbook of Metal–Ligand<br />

Interactions in Biological Fluids. Vol. 2. New York: Marcel Dekker Inc.,<br />

1995:886.<br />

152. Medeiros DM, Wildman R, Liebes R. Metal metabolism and toxicities. In:<br />

Massaro EJ, ed. Handbook of Human Toxicology. Boca Raton: CRC Press,<br />

1997:149.<br />

153. Harris ED, DiSilvestro RA. Lysyl oxidase, a molecular target for copper. In:<br />

Sorenson JRJ, ed. Inflammatory Diseases and Copper. Clifton: Humana Press,<br />

1982:183.<br />

154. Greenaway FT, O’Gara CY. Copper and connective tissue: the mechanism of<br />

lysyl oxidase. In: Sorenson JRJ, ed. Biology of Copper Complexes. Clifton:<br />

Humana Press, 1987:67.<br />

155. Sorenson JRJ. Pharmacological activities of copper compounds. In: Berthon G,<br />

ed. Handbook of Metal–Ligand Interactions in Biological Fluids. Vol. 2. New<br />

York: Marcel Dekker Inc., 1995:1128.


The Role of Copper in Inflammation 229<br />

156. Rucker RB, Kosonen T, Clegg MS, et al. Copper, lysyl oxidase, and extracellular<br />

matrix protein cross-linking. Am J Clin Nutr 1998; 67(suppl):996S.<br />

157. Simeon A, Wegrowski Y, Bontemps Y, et al. Expression of glycosaminoglycans<br />

and small proteoglycans in wounds: modulation by the tripeptide-copper complex<br />

glycyl-L-histidyl-L-lysine-Cu(þ). J Invest Dermatol 2000; 115:962.<br />

158. Pickart L. Iamin: a human growth factor with multiple wound-healing properties.<br />

In: Sorenson JRJ, ed. Biology of Copper Complexes. Clifton: Humana<br />

Press, 1987:273.<br />

159. Canapp SO Jr., Farese JP, Schultz GS, et al. The effects of topical tripeptidecopper<br />

complex on healing of ischemic open wounds. Vet Surg 2003; 32:515.<br />

160. Pollard JD, Quan S, Kang T, et al. Effects of copper tripeptide on the<br />

growth factors by normal and irradiated fibroblasts. Arch Facial Plast Surg<br />

2005; 7:27.<br />

161. Maquart FX, Simeon A, Pasco S, et al. Regulation of cell activity by the extracellular<br />

matrix: the concept of matrikines. J Soc Biol 1999; 193:423.<br />

162. Walker WR, Keats DM. An investigation of the therapeutic value of the ‘‘copper<br />

bracelet’’: dermal assimilation of copper in arthritic/rheumatoid conditions.<br />

Agents Actions 1976; 6:454.<br />

163. Walker WR, Griffin BJ. The solubility of copper in human sweat. Search 1976;<br />

7:100.<br />

164. Beveridge SJ. Topically applied copper preparations for anti-inflammatory<br />

therapy. In: Rainsford KD, et al. eds. Copper and Zinc in Inflammatory and<br />

Degenerative Diseases. Dordrecht: Kluwer Academic Publishers, 1998:139.<br />

165. Auer DE, Ng JC, Seawright AA. Copper salicylate and copper phenylbutazone<br />

as topically applied anti-inflammatory agents in the rat and horse. J Vet Pharmacol<br />

Ther 1990; 13:67.<br />

166. Walker WR, Beveridge SJ, Whitehouse MW. Dermal copper drugs: the<br />

copper bracelet and Cu(II) salicylate complexes. In: Rainsford KD, Brune K,<br />

Whitehouse MW, eds. Trace Elements in the Pathogenesis and Treatment of<br />

Inflammation. Basel: Birkhäuser Verlag, 1981:359.<br />

167. Walker WR, Beveridge SJ, Whitehouse MW. Anti-inflammatory activity of a<br />

dermally applied copper salicylate preparation (Alcusal 1 ). Agents Actions<br />

1980; 10:38.<br />

168. Beveridge SJ, Walker WR, Whitehouse MW. Anti-inflammatory activity of<br />

copper salicylate applied to rats percutaneously in dimethyl sulphoxide with<br />

glycerol. J Pharm Pharmacol 1980; 32:425.<br />

169. Beveridge SJ, Whitehouse MW, Walker WR. Lipophilic copper(II) formulations:<br />

some correlations between their composition and anti-inflammatory/antiarthritic<br />

activity when applied to the skin of rats. Agents Actions 1982; 14:291.<br />

170. Shackel NA, Day RO, Kellett B, et al. Copper-salicylate gel for pain relief in<br />

osteoarthritis: a randomised controlled trial. Med J Aust 1997; 167(3):134.<br />

171. Hostynek JJ, Dreher F, Maibach HI. Human stratum corneum penetration by<br />

copper: in vivo study after occlusive and semi-occlusive application of the metal<br />

as powder. 5 Meeting SIDAPA (Società Italiana di Dermatologia Allergologia<br />

Professionale ed Ambientale), Tivoli, Italy, Oct 27–29, 2005, Abstract, p. 62.<br />

172. Hostynek JJ, Maibach HI. Skin irritation potential of copper compounds. Toxicol<br />

Mech Meth 2004; 14:205.


230 Milanino<br />

173. Brewer GJ. Practical recommendations and new therapies for Wilson’s disease.<br />

Drugs 1995; 50:240.<br />

174. Ogra Y, Ohmichi M, Suzuki KT. Mechanisms of selective copper removal by<br />

tetrathiomolybdate from metallothionein in LEC rats. Toxicology 1996; 106:75.<br />

175. Ceve G. Lipid vesicles and other colloids as drugs carriers on the skin. Adv<br />

Drug Deliv Rev 2004; 56:675.<br />

176. Konduri KS, Nandedkar S, Rickaby DA, et al. The use of sterically stabilized<br />

liposomes to treat asthma. Meth Enzymol 2005; 391:413.<br />

177. Justo OR, Moraes AM. Kanamycin incorporation in lipid vesicles prepared by<br />

ethanol injection designed for tuberculosis treatment. J Pharm Pharmacol 2005;<br />

57:23.<br />

178. Puglia C, Trombetta D, Venuti V, et al. Evaluation of in-vivo topical antiinflammatory<br />

activity of indomethacin from liposomal vesicles. J Pharm Pharmacol<br />

2004; 56:1225.<br />

179. Hart EB, Steenbock H, Waddel J, et al. Iron nutrition. VII. Copper is a supplement<br />

to iron for hemoglobin building in the rat. J Biol Chem 1928; 77:797.<br />

180. Harris ED. Basic and clinical aspects of copper. Crit Rev Clin Lab Sci 2003;<br />

40:547.<br />

181. Tapiero H, Towsend DM, Tew KD. Trace elements in human physiology and<br />

pathology. Copper Biomed Pharmacother 2003; 57:386.<br />

182. Uauy R, Olivares M, Gonzales M. Essentiality of copper in humans. Am J Clin<br />

Nutr 1998; 67(suppl):952S.<br />

183. Failla LM, Johnson MA, Prohaska JR. Copper. In: Present Knowledge in<br />

Nutrition. Washington, D.C.: Life Science Institute Press, 2001:373.<br />

184. Turnlund J. Copper. In: Shils ME, et al., eds. Modern Nutrition in Health and<br />

Disease. Baltimore: Williams & Wilkins, 1999:241.<br />

185. Fox PL, Mukhopadhyay C, Ehrenwald E. Structure, oxidant activity, and cardiovascular<br />

mechanisms of human ceruloplasmin. Life Sci 1995; 56:1749.<br />

186. Klotz L-O, Weser U. Biological chemistry of copper compounds. In: Rainsford<br />

KD, et al., eds. Copper and Zinc in Inflammatory and Degenerative Diseases.<br />

Dordrecht: Kluwer Academic Publishers, 1998:19.<br />

187. Patel BN, Dunn RJ, David S. Alternative RNS splicing generates a glycosylphosphatidylinositol-anchored<br />

form of ceruloplasmin in mammalian brain.<br />

J Biol Chem 2000; 275:4305.<br />

188. Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological roles<br />

inside and outside of the cell. J Cell Biochem 2003; 88:660.<br />

189. Mizuguchi H, Imamura I, Takemura M, et al. Purification and characterisation<br />

of diamine oxidase (histaminase) from rat small intestine. J Biochem 1994;<br />

116:631.<br />

190. Stewart LC, Klinman JP. Dopamine beta-hydroxylase of adrenal chromaffin<br />

granules: structure and function. Ann Rev Biochem 1986; 57:551.<br />

191. Reedy BJ, Blackburn NJ. Preparation and characterization of half-apo dopamine<br />

b-hydroxylase by selective removal of CuA. Identification of sulfur ligand<br />

at the dioxygen binding site by EXAFS and FTIR spectroscopy. J Am Chem<br />

Soc 1994; 116:1924.<br />

192. Réglier M, Amadéi E, Alilou EH, et al. Oxidation of unactivated hydrocarbons:<br />

models for tyrosinase and dopamine b-hydroxylase. In: Karlin KD, Tyeklár Z,


The Role of Copper in Inflammation 231<br />

eds. Bioinorganic Chemistry of Copper. New York: Chapman & Hall,<br />

1993:348.<br />

193. Jalkanen S, Salmi M. Cell surface monoamine oxidases: enzymes in search of<br />

function. EMBO J 2001; 20:3893.<br />

194. Murthy ASN, Mains RE, Eipper BA. Purification and characterization of<br />

peptidyl a-amidating monoxygenase from bovine neurointermediate pituitary.<br />

J Biol Chem 1986; 261:1815.<br />

195. Albergoni V, Piccinni E. Copper and zinc metallothioneins. In: Rainsford KD,<br />

ed. Copper and Zinc in Inflammatory and Degenerative Diseases. Dordrecht:<br />

Kluwer Academic Publishers, 1998:61.<br />

196. Lu ZH, Solioz M. Bacterial copper transport. Adv Protein Chem 2002; 60:93.<br />

197. Elam JS, Thomas ST, Holloway SP, et al. Copper chaperones. Adv Protein<br />

Chem 2002; 60:151.<br />

198. Voskoboinik I, Camaraki J, Mercer JFB. Understanding the mechanism and<br />

function of copper P-type ATPases. Adv Protein Chem 2002; 60:123.<br />

199. Fuentes-Prior P, Fujikawa K, Pratt KP. New insights into binding interfaces of<br />

coagulation factors V and VIII and their homologues: lessons from high resolution<br />

crystal structures. Curr Protein Pept Sci 2002; 3:313.<br />

200. Milanino R, Velo GP, Marrella M. Copper and zinc in the pathophysiology<br />

and treatment of inflammatory disorders. In: Nève J, Chappuis P, Lamand<br />

M, eds. Therapeutic Uses of Trace Elements. New York: Plenum Publishing<br />

Corporation, 1996:115.<br />

201. Spector WG, Willoughby DA. Histamine and 5-hydroxytryptamine in experimental<br />

pleurisy. J Pathol Bact 1957; 74:57.<br />

202. Majno G, Palade GE. Studies on inflammation: I. The effect of histamine and<br />

serotonine on vascular permeability. J Biophys Biochem Cytol 1961; 11:571.<br />

203. Schuschke DA. Dietary copper in the physiology of the microcirculation. J Nutr<br />

1997; 127:2274.<br />

204. Bossi F, Fischetti F, Pellis V, et al. Platelet-activating factor and kinindependent<br />

vascular leakage as a novel functional activity of the soluble terminal<br />

complement complex. J Immunol 2004; 173:6921.<br />

205. Sorenson JRJ. Down-regulation of nitric oxide synthetase may account for the<br />

antiinflammatory activities of copper chelates. In: Nève J, Chappuis P, Lamand<br />

M, eds. Therapeutic Uses of Trace Elements. New York: Plenum Publishing<br />

Corporation, 1996:133.<br />

206. Flower RJ. Drugs which inhibit prostaglandin biosynthesis. Pharmacol Rev<br />

1974; 26:33.<br />

207. Simmons DL, Botting RM, Hla T. Cyclooxygenase isozymes: the biology of<br />

prostaglandin synthesis and inhibition. Pharmacol Rev 2004; 56:387.<br />

208. Fujita T, Ohtani N, Aihara M, et al. Comparison of the effects of Fe 2þ and<br />

Cu 2þ on prostaglandin synthesis in rabbit kidney medulla slices. J Pharm Pharmacol<br />

1987; 39:230.<br />

209. Elliott GR, van Batenburg MJ, Bonta IL. Copper modulation of macrophage<br />

cyclooxygenase metabolite synthesis. Prostaglandins 1987; 34:657.<br />

210. Frenchilla D, Lasheras B, Ucelay M, et al. On the mechanisms of the antiinflammatory<br />

activity of some copper(II) complexes. Arzneimittelforschung<br />

1990; 40:1008.


232 Milanino<br />

211. Sorenson JRJ. Copper complexes in the treatment of experimental inflammatory<br />

conditions: inflammation, ulcers and pain. In: Milanino R, Rainsford<br />

KD, Velo GP, eds. Copper and Zinc in Inflammation. Dordrecht: Kluwer<br />

Academic Publishers, 1989:69.<br />

212. Hong SL, Levine L. Inhibition of arachidonic acid release from cells as the biochemical<br />

action of anti-inflammatory corticosteroids. Proc Natl Acad Sci USA<br />

1976; 73:1730.<br />

213. Pickart L. Copper peptides for tissue regeneration. Specialty Chem 2002; 5:32.<br />

214. Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology,<br />

and pharmacology. Pharmacol Rev 1991; 43:109.<br />

215. Jouzeau JY, Pacquelet S, Boileau C, et al. Nitric oxide (NO) and cartilage metabolism:<br />

NO effects are modulated by superoxide in response to IL-1. Biorheology<br />

2002; 39:201.<br />

216. Evans GH. Nitric oxide: what role does it play in inflammation and tissue<br />

destruction? Agents Actions (Suppl) 1995; 47:107.<br />

217. Baquial JG, Sorenson JRJ. Down-regulation of NADPH-diaphorase (nitric<br />

oxide synthase) may account for the pharmacological activities of Cu(II)2(3,5diisopropylsalicylate)<br />

4. J Inorg Biochem 1995; 60:133.<br />

218. Sorenson JRJ. Copper complexes for therapy of cancer and autoimmune<br />

diseases. In: Rainsford KD, ed. Copper and Zinc in Inflammatory and Degenerative<br />

Diseases. Dordrecht: Kluwer Academic Publishers, 1998:113.<br />

219. Colasanti M, Persichini T, Venturini G, et al. Modulation of the nitric<br />

oxide pathway by copper in glial cells. Biochem Biophys Res Commun 2000;<br />

257:776.<br />

220. Vega A, Chacon P, Monteseirin J, et al. A new role for monoamino oxidases in<br />

the modulation of macrophage-inducible nitric oxide synthase gene expression.<br />

J Leukoc Biol 2004; 75:1093.<br />

221. van Furth R. Phagocytic cells: development and distribution of mononuclear<br />

phagocytes in normal steady sate and inflammation. In: Gallin JI, Goldstein<br />

IM, Snyderman R, eds. Inflammation: Basic Principles and Clinical Correlates.<br />

New York: Raven Press, 1988:281.<br />

222. Rainsford KD. Side effects and toxicology of the salicylates. In: Rainsford KD,<br />

ed. Aspirin and Related Drugs. London and New York: CRC Press, 2004:215.<br />

223. McCord JM, Keele BB Jr., Fridovich I. An enzyme-based theory of obligate<br />

anaerobiosis: the physiological function of superoxide dismutase. Proc Natl<br />

Acad Sci USA 1971; 68:1024.<br />

224. Miesel R, Zuber M. Copper-dependent antioxidase defenses in inflammatory<br />

and autoimmune rheumatic diseases. Inflammation 1994; 17:283.<br />

225. Goldstein IM, Kaplan HB, Edelson HS, et al. Ceruloplasmin: an acute phase<br />

reactant that scavengers oxygen-derived free radicals. Ann N Y Acad Sci<br />

1982; 389:368.<br />

226. Ciuffi M, Cellai C, Franchi-Micheli S, et al. An in vivo, ex vivo and in vitro<br />

comparative study of the activity of copper oligopeptide complexes vs Cu(II)<br />

ions. Pharmacol Res 1998; 38:279.<br />

227. Afanas’eva IB, Ostrakhovitch EA, Mikhal’chik EV, et al. Enhancement of antioxidant<br />

and anti-inflammatory activities of bioflavonoid rutin by complexation<br />

with transition metals. Biochem Pharamcol 2001; 15:677.


The Role of Copper in Inflammation 233<br />

228. Morgant G, Dung NH, Daran JC, et al. Low-temperature crystal structures<br />

of tetrais-mu-3,5-diisopropylsalicylatobis-dimethylformamidodicopper(II) and<br />

their role in modulating polymorphonuclear leukocyte activity in overcoming<br />

seizures. J Inorg Biochem 2000; 81:11.<br />

229. Weser U, Richter C, Wendel A, et al. Reactivity of antiinflammatory and superoxide<br />

dismutase active Cu(II) salicylates. Bioinorg Chem 1978; 8:201.<br />

230. Underhill AE, Bury A, Odling RJ, et al. Metal complexes of antiinflammatory<br />

drugs. Part VII: salsalate complex of copper(II). J Inorg Biochem<br />

1989; 37:1.<br />

231. Roch-Arveiller M, Huy DP, Maman L, et al. Non-steroidal anti-inflammatory<br />

drug-copper complex modulation of polymorphonuclear leukocyte migration.<br />

Biochem Pharmacol 1990; 39:569.<br />

232. Roch-Arveiller M, Revelant V, Huy DP, et al. Effects of some non-steroidal<br />

anti-inflammatory drug copper complexes on polymorphonuclear leukocyte<br />

oxidative metabolism. Agents Actions 1990; 31:65.<br />

233. Roch-Arveiller M, Maman L, Huy DP, et al. Modulation of polymorphonuclear<br />

leukocyte responsiveness by copper(II)2niflumate)4. Inflamm Res 1995; 44:198.<br />

234. Roos D, Weening RS. Defects in the oxidative killing of microorganisms by<br />

phagocytic leukocytes. In: Oxygen Free Radicals and Tissue Damage. Ciba<br />

Foundation Symposium 65. Amsterdam: Excerpta Medica, 1979:225.<br />

235. McCord JM, Wong K. Phagocyte-produced free radicals: roles in cytotoxicity<br />

and inflammation. In: Oxygen Free Radicals and Tissue Damage. Ciba Foundation<br />

Symposium 65. Amsterdam: Excerpta Medica, 1979:343.<br />

236. Splettstoesser WD, Schuff-Werner P. Oxidative stress in phagocytes: ‘‘the<br />

enemy within.’’ Microsc Res Tech 2002; 57:441.<br />

237. Milanino R, Velo GP. Multiple action of copper in control of inflammation:<br />

studies in copper-deficient rats. In: Rainsford KD, Brune K, Whitehouse<br />

MW, eds. Trace Elements in the Pathogenesis and Treatment of Inflammation.<br />

Basel: Birkhäuser Verlag, 1981:209.<br />

238. Ōyanagui Y. Participation of superoxide anions at the prostaglandin phase of<br />

carrageenan foot-oedema. Biochem Pharmacol 1976; 25:1456.<br />

239. Kuehl FA Jr., Humes JL, Egan RW, et al. Role of prostaglandin endoperoxide<br />

PGG2 in inflammatory process. Nature 1977; 265:170.<br />

240. Pou S, Pou WS, Bredt DS, et al. Generation of superoxide by purified brain<br />

nitric oxide synthase. J Biol Chem 1992; 267:24,173.<br />

241. Kohen R, Misgav R, Ginsburg I. The SOD like activity of copper:carnosine,<br />

copper anserine and copper:homocarnosine complexes. Free Rad Res<br />

Commun 1991; 12–13(Pt 1):179.<br />

242. Liochev SI, Radonova NA, Russanova EE. A study on the ability of copper<br />

complexes to act as active oxygen species scavengers. Bulgarian Acad Sci 1987;<br />

13:40.<br />

243. Bonen M, Kampfman JK, Waterman BJ, et al. SOD-mimetic activity of simple<br />

copper (II) salts and complexes. Joint Meeting of German and Italian Pharmacologists,<br />

Venice, Oct 4-6, 1977, Abstract 214.<br />

244. Duracková Z, Labuda J. Superoxide dismutase mimetic activity of macrocyclic<br />

Cu(II)-tetraanhydroaminobenzaldehyde (TAAB) complex. J Inorg Biochem<br />

1995; 58:297.


234 Milanino<br />

245. Willingham WM, Sorenson JRJ. Copper(II)-ethylenediaminetetraacetate does<br />

disproportionate superoxide. Biochem Biophys Res Commun 1988; 150:252.<br />

246. Nilsson K. Effects of Cu(II) (diisopropylsalicylate)2 on soluble protein kinase C<br />

activity in rat liver. Cancer Lett 1989; 47:169.<br />

247. Chen X-L, Medford RM. Oxidation–reduction sensitive regulation of vascular<br />

inflammatory gene expression. In: Pearson JD, ed. Vascular Adhesion Molecules<br />

and Inflammation. Basel: Birkhäuser Verlag, 1999:161.<br />

248. Furie MB. Production and presentation of chemokines by endothelial cells. In:<br />

Pearson JD, ed. Vascular Adhesion Molecules and Inflammation in Vascular<br />

Adhesion Molecules and Inflammation. Basel: Birkhäuser Verlag, 1999:65.<br />

249. Ley K. Adhesion of leukocytes from flow: the selectins and their ligands. In:<br />

Pearson JD, ed. Vascular Adhesion Molecules and Inflammation. Basel:<br />

Birkhäuser Verlag, 1999:11.<br />

250. Smith CW, Burns AR, Scott SI. Co-operative signaling between leukocytes and<br />

endothelium mediating firm attachment. In: Pearson JD, ed. Vascular Adhesion<br />

Molecules and Inflammation. Basel: Birkhäuser Verlag, 1999:39.<br />

251. Salmi M, Tohka S, Jalkanen S. Human vascular adhesion protein-1 (VAP-1)<br />

plays a critical role in lymphocyte-endothelial cell adhesion cascade under<br />

shear. Circ Res 2000; 86:1245.<br />

252. Koskinen K, Vainio PJ, Smith DJ, et al. Granulocyte transmigration through<br />

the endothelium is regulated by the oxidase activity of vascular adhesion protein-1<br />

(VAP-1). Blood 2004; 103:3388.<br />

253. Salmi M, Jalkanen S. VAP-1: and adhesion and an enzyme. Trends Immunol<br />

2001; 22:211.<br />

254. Merinen M, Irjala H, Salmi M, et al. Vascular adhesion protein-1 is involved in<br />

both acute and chronic inflammation in the mouse. Am J Pathol 2005; 1667:93.<br />

255. Stolen CM, Marttila-Ichihara F, Koskinen K, et al. Absence of the endothelial<br />

oxidase AOC3 leads to abnormal leukocyte traffic in vivo. Immunity 2005;<br />

22:105.<br />

256. Davies ME. Pasqualicchio M. Copper and zinc compounds and cell surface<br />

interactions. In: Rainsford KD, et al., eds. Copper and Zinc in Inflammatory<br />

and Degenerative Diseases. Dordrecht: Kluwer Academic Publishers, 1998:161.<br />

257. Bellavite P, Chirumbolo S, Mansoldo C, et al. Simultaneous assay for oxidative<br />

metabolism and adhesion of human neutrophils: evidence for correlation and<br />

dissociation of the two responses. J Leukoc Biol 1992; 51:329.<br />

258. Failla ML. Trace elements and host defense: recent advances and continuing<br />

challenges. J Nutr 2003; 133:1443S.<br />

259. Boyne R, Arthur JR. Effects of selenium and copper deficiency on neutrophil<br />

functions in cattle. J Comp Pathol 1981; 91:271.<br />

260. Lukasewycz OA, Prohaska JR. Immunization against transplantable leukaemia<br />

impaired in copper-deficient mice. J Natl Cancer Inst 1982; 69:489.<br />

261. Jones DG, Suttle NF. The effect of copper deficiency on the resistance of mice<br />

to infection with Pasteurella haemolytica. J Comp Pathol 1983; 93:143.<br />

262. Korte JJ, Prohaska JR. Dietary copper deficiency alters protein and lipid<br />

composition of murine plasma membrane. J Nutr 1987; 117:1076.<br />

263. Babu U, Failla ML. Respiratory burst and candidacidal activity of peritoneal<br />

macrophages are impaired in copper-deficient rats. J Nutr 1990; 120:1692.


The Role of Copper in Inflammation 235<br />

264. Lukasewycz OA, Prohaska JR. The immune response in copper deficiency. Ann<br />

N Y Acad Sci 1990; 587:147.<br />

265. Bala S, Failla ML, Lunney JK. Alterations in splenic lymphoid cell subsets and<br />

activation antigens in copper-deficient rats. J Nutr 1991; 121:745.<br />

266. Xin Z, Waterman DF, Hemken RW, et al. Effects of copper status on neutrophils<br />

function, superoxide dismutase, and copper in steers. J Dairy Sci 1991;<br />

74:3078.<br />

267. Bala S, Failla ML. Copper deficiency reversibly impairs DNA synthesis in activated<br />

T lymphocytes by limiting interleukin 2 activity. Proc Natl Acad Sci USA<br />

1992; 89:6794.<br />

268. Kelly DS, Daudu PA, Taylor PC, et al. Effects of low-copper diets on human<br />

immune response. Am J Clin Nutr 1995; 62:412.<br />

269. Hopkins RG, Failla ML. Copper deficiency reduces interleukin-2 (IL-2) production<br />

and IL-2 mRNA in human T-lymphocytes. J Nutr 1997; 127:257.<br />

270. Hopkins RG, Failla ML. Transcriptional regulation if interleukin-2 gene<br />

expression is impaired by copper deficiency in Jurkat human T-lymphocytes.<br />

J Nutr 1999; 129:596.<br />

271. Hopkins RG, Failla ML. Chronic intake of marginally low copper diet impairs<br />

in vitro activities of lymphocytes and neutrophils from male rats despite minimal<br />

impact on conventional indicators of copper status. J Nutr 1995; 125:2658.<br />

272. Hopkins RG, Failla ML. Decreased synthesis of interleukin-2 (IL-2) in copperdeficient<br />

T-cells mediated by inhibition of the transcription factor NF-AT.<br />

FASEB J 2001; 15:A272, abstr.237.8.<br />

273. Soderberg LS, Barnett JB, Sorenson JRJ. Copper complexes stimulate hemopoiesis<br />

and lymphopoiesis. Adv Exp Med Biol 1989; 258:209.<br />

274. Pocino M, Baute l, Malave I. Influence of oral administration of excess copper<br />

on the immune response. Fund Appl Toxicol 1991; 16:249.<br />

275. Marikovsky M, Ziv V, Nevo N, et al. Cu/Zn superoxide dismutase plays<br />

important role in immune response. J Immunol 2003; 170:2993.<br />

276. Jutila MA. Selective lymphocyte migration into secondary lymphoid organs<br />

and inflamed tissues. In: Pearson JD, ed. Vascular Adhesion Molecules and<br />

Inflammation. Basel: Birkhäuser Verlag, 1999:141.<br />

277. Percival SS. Copper and immunity. Am J Clin Nutr 1998; 67:S1064.<br />

278. Aggett PJ, Fairweather-Tait S. Adaptation to high and low copper intakes: its<br />

relevance to estimate safe and adequate daily dietary intakes. Am J Clin Nutr<br />

1998; 67:S1061.<br />

279. Scheinberg HI, Sternlieb I. Copper toxicity and Wilson’s disease. In: Prasad AS,<br />

Oberleas D, eds. Trace Elements in Human Health and Diseases. Zinc and<br />

Copper. Vol. 1. New York: Academic Press, 1976:415.


10<br />

Copper Jewelry and Arthritis<br />

Brenda J. Harrison<br />

Department of Earth and Ocean Sciences, Copper Research Information Flow<br />

Project, University of British Columbia, Vancouver, British Columbia, Canada<br />

Pseudoscience differs from erroneous science. Science thrives on errors,<br />

cutting them away one by one. False conclusions are drawn all the time,<br />

but they are drawn tentatively. Hypotheses are framed so they are<br />

capable of being disproved. A succession of alternative hypotheses is<br />

confronted by experiment and observation. Science gropes and staggers<br />

toward improved understanding. Proprietary feelings are of course<br />

offended when a scientific hypothesis is disproved, but such disproofs<br />

are recognized as central to the scientific enterprise.<br />

—Carl Sagan (1)<br />

INTRODUCTION<br />

This paper provides an overview, from the point of view of a nonspecialist, of<br />

the ‘‘copper bracelet’’ issue, based on an exploration of the available scientific<br />

literature, popular press, promotional material made available on the<br />

Internet, discussions of the issue of complementary and alternative health<br />

care methods, and books for the lay reader. The focus will be on copper<br />

bracelets, since they are currently the ‘‘in vogue’’ appliances. The discussion<br />

is applicable to other means of wearing metallic copper on the human body,<br />

237


238 Harrison<br />

from copper rings, to copper disks worn under watchbands, or copper<br />

pennies worn inside socks!<br />

‘‘Arthritis’’ is the general term for a suite of over 100 distinct diseases<br />

and conditions. The most common forms are rheumatoid (involving persistent<br />

inflammation of the synovial membranes of joints) and osteoarthritis<br />

(a degenerative process involving the cartilage and bone of joints). Arthritis<br />

affects nearly 43 million Americans (one in six); although arthritis can strike<br />

at any age, aging of the ‘‘baby boom’’ generation will bring an increase in<br />

the number of sufferers in the coming decades (2). Most forms of arthritis<br />

are incurable, but effective interventions (including use of analgesics, steroidal<br />

and nonsteroidal anti-inflammatory drugs—NSAIDs), are available.<br />

They are underused (3).<br />

‘‘The wearing of copper bracelets is a folk remedy for arthritis, but<br />

there are little data to support the efficacy of this remedy’’ (4). The role<br />

of copper in the treatment of arthritis was reviewed by Milanino et al. (5)<br />

to ‘‘stimulate a thoughtful and unbiased reconsideration of the old idea of<br />

using copper as a therapeutic agent in the treatment of those chronic inflammatory<br />

diseases—in particular rheumatoid arthritis.’’<br />

Serum copper is elevated in rheumatoid arthritis although there is evidence<br />

that serum copper does not correlate with disease severity (6–8). From<br />

animal studies it has been established that the rise in serum copper is accompanied<br />

by an increase in liver copper levels (9–12). The modern use of copper<br />

complexes to treat arthritis dates from the 1940s, and the theory that<br />

copper complexes of nonsteroidal anti-inflammatory drugs are more active<br />

and less toxic than the parent compounds is supported by a substantial body<br />

of literature (13,14).<br />

The role of copper in arthritis is complex; the prevailing paradigm<br />

includes the following elements:<br />

1. Copper is an essential trace element; copper deficiency may have<br />

an adverse effect on inflammation and connective tissue disease<br />

such as arthritis.<br />

2. Serum copper is elevated in arthritis.<br />

3. The plasma protein ceruloplasmin, which binds copper, is an<br />

‘‘acute phase reactant’’; the observed elevation of serum copper<br />

is often attributed to an increase in this protein as a result of the<br />

disease.<br />

4. D-penicillamine, used to treat arthritis, binds free copper ions.<br />

5. Copper is an essential cofactor for the enzyme Cu, Zn-superoxide<br />

dismutase; this enzyme neutralizes the destructive free radicals that<br />

may play a role in tissue damage in the disease and it possesses<br />

anti-inflammatory activity.<br />

6. The enzyme lysyl oxidase requires copper as a cofactor and is<br />

required for connective tissue formation.


Copper Jewelry and Arthritis 239<br />

The established role of copper in both superoxide dismutase and ceruloplasmin<br />

offers a framework in which copper could be expected to play a<br />

role in arthritis treatment; however, ‘‘unfortunately, other than for goldsulfhydryl<br />

compounds, there exists a chronic deficiency of solid scientific<br />

information about any role of trace metallic elements, whether they are<br />

administered parenterally, orally, or topically as therapeutic agents in the<br />

management of rheumatoid arthritis (RA) (15). In an intriguing study<br />

using an animal model, a mixture of copper, gold, and silver exhibited significant<br />

antirheumatic function not shown by the individual elements (16).<br />

THE COPPER BRACELET ‘‘MYTH’’ AND HYPOTHESIS<br />

Copper bracelets, armbands, rings, etc., are a folk remedy promoted and sold<br />

for the relief of the symptoms of arthritis. They may be purchased from<br />

pharmacies, health food stores, catalogues, ‘‘new-age stores,’’ and online<br />

suppliers through the Internet. A Google search of the Internet for<br />

(þ ‘‘copper bracelets’’ þarthritis) conducted on January 17, 2005 uncovered<br />

approximately 12,300 Web pages. A Google Scholar search the same day<br />

uncovered only 32 pages using the same search terms. Google Scholar allows<br />

specific searching of scholarly literature (such as peer-reviewed papers,<br />

theses, books, and technical reports). Most of the pages turned up by the<br />

standard Google search are promotional sites, featuring enthusiastic testimonials.<br />

A variety of health-related claims are made on these sites—many claim<br />

therapeutic bracelets date from antiquity and are ‘‘powerful’’ and ‘‘natural.’’<br />

Along with the testimonials, some sites also provide descriptions of the<br />

‘‘theory’’ behind how the bracelets work.<br />

Claims for the Antiquity of the Remedy<br />

Many websites describe the use of copper bracelets for arthritis as having a<br />

very long history, although they are generally vague about how long that<br />

history might be. Many talk of ‘‘ancient writings’’ and ‘‘ancient times’’; or<br />

refer to Roman, Greek, or Egyptian times; or use by ancient Aztecs, Persians,<br />

or Hindus. Others refer to a period of thousands of years, centuries, even<br />

‘‘over 200 years.’’ Some refer to lost knowledge (especially knowledge lost<br />

or abandoned by modern medicine), while others suggest that bracelets have<br />

been in constant use for a very long time.<br />

Claims for the Power of the Remedy<br />

Many websites tell of folklore and ‘‘old wives’ tales’’ that support the idea<br />

that copper bracelets can ease the suffering of arthritis patients. They use<br />

such terms as ‘‘healing power,’’ ‘‘medicinal value,’’ ‘‘ease the pain,’’ ‘‘ease<br />

discomfort,’’ ‘‘relief of pain,’’ or ‘‘increase mobility’’—all of which suggest


240 Harrison<br />

that the bracelets can alleviate or heal arthritis. Many cite the use of copper<br />

bracelets by athletes, especially professional golfers, as evidence of their<br />

effectiveness.<br />

Claims for the ‘‘Naturalness’’ of the Remedy<br />

The ‘‘naturalness’’ of copper bracelets is a prominent feature on promotional<br />

websites. Key phrases include ‘‘natural,’’ ‘‘natural home remedy,’’ ‘‘natural<br />

remedy,’’ ‘‘natural healing,’’ and ‘‘natural relief.’’<br />

Claims About the Underlying Science Behind the Remedy and<br />

Theories About How It Works<br />

Some web pages claim that copper bracelets work by increasing blood or<br />

oxygen flow, or increasing circulation, and thus speeding healing or relieving<br />

pain. Others claim that copper interacts with ‘‘positive fields’’ or energy flow<br />

in the body, or in some way balances electrical potentials. Many claim that<br />

arthritis patients are deficient in copper and that the bracelets supply the<br />

essential element and so promote health and healing; bracelets are promoted<br />

as a worry-free, time-released source of supplemental copper that may<br />

remedy or prevent inflammation. Another group of sites claims that the<br />

copper absorbs toxins by some mechanism or that copper will combat<br />

superoxide radical damage.<br />

Consideration of the Reputed Antiquity of the Remedy<br />

There is a pervasive belief that the use of copper jewelry as a remedy for<br />

arthritis stretches across many cultures and back into history for centuries<br />

or even millennia. The basis for this belief is not clear, although it is likely<br />

that people have worn copper jewelry and amulets since the earliest working<br />

of copper. Arthritis as a condition predates civilization and even the origin of<br />

the human species. Evidence of arthritic deformation can be seen in dinosaur<br />

skeletons and cave bears of the Pleistocene epoch were also afflicted (17).<br />

Neanderthal and Neolithic skeletons, early Egyptian skeletons dating back<br />

to 4000 BC—all show evidence that arthritis was widespread.<br />

The use of copper compounds as therapeutics by early cultures is well<br />

documented and probably dates to the beginning of recorded history and<br />

earlier. Most applications relate to copper’s antibiotic properties (18). The<br />

ancient Egyptian medical text, the Smith Papyrus dating from 1600 to<br />

1300 BC, recounts the use of copper to treat infected wounds and to sterilize<br />

drinking water. Other ancient texts from Egypt, Greece, Rome, the Aztec<br />

empire, ancient India, Persia, and China make it clear that early healers used<br />

a number of copper compounds to treat a wide variety of conditions ranging<br />

from infections to ulcers, inflammation, hemorrhoids, neuralgia, leprosy,<br />

eye ailments, venereal disease, intestinal worms, lung diseases, and in the


Copper Jewelry and Arthritis 241<br />

promotion of wound healing (19). Dresher (20) gives a good, updated overview<br />

of historical and modern uses of copper in medicine.<br />

The Pharmaceutical Journal in 1974 carried a small column called<br />

‘‘Copper on the up and up,’’ which gives an entertaining (and prophetic) history<br />

of the use of copper through the ages and the prospects for its medical<br />

development (21). This brief account includes the statement, ‘‘How long ago<br />

people started to wear copper or bronze ornaments for their supposed magical<br />

or remedial effects is unknown.’’ The discovery that the skeleton of a<br />

Durotrigian defender of Maiden Castle in Dorset who was killed in 43 AD<br />

had been buried with a spiral bronze ring on one of the toes led the author<br />

to speculate that it is ‘‘ ... not too fanciful to suppose that such rings were<br />

employed to ward off the gout or the rheumatism ...’’ This story was<br />

repeated by Dr. John Sorenson (22) in a paper on the therapeutic uses of<br />

copper; however, Sorenson (23), in the next sentence classified copper bracelets<br />

per se as ‘‘modern copper-containing folklore remedies for the treatment<br />

of arthritis.’’ In an e-mail message to the author on November 1, 2004,<br />

Dr. Niall Sharples (24), Senior Lecturer in Archaeology at Cardiff University<br />

and author of an English Heritage book about the history of Maiden<br />

Castle, was able to confirm part of the skeleton story. There is ‘‘ ...indeed<br />

a copper alloy probably bronze ring around the toe of a skeleton from<br />

Maiden Castle. ... These rings are fairly common in the Iron Age of Britain<br />

and they are clearly jewellry worn on both fingers and toes. Burials however,<br />

are less common and there are very few burials with rings if any ... Whether<br />

they also functioned in a medicinal fashion I could not say.’’ The Maiden<br />

Castle book provides a drawing on page 120 of the skeleton; a tankard<br />

had also been buried with the tanker. Spiral bronze rings were common trade<br />

goods of the period and may have been worn as status symbols (24).<br />

Native North Americans have a copper bracelet myth of their own, in<br />

which copper bracelets are worn not for healing, but as a source of mystical<br />

‘‘power’’ (25).<br />

Theophrastus Phillippus Aureolus Bombastus von Hohenheim (1493–<br />

1541), better known as the celebrated Renaissance physician, alchemist, and<br />

philosopher Paracelsus, crafted ‘‘constellation rings’’ in metals that corresponded<br />

to the known planets including copper to bring the protective<br />

influence of Venus and so ‘‘was one of the first recorded copper bracelet<br />

wearers, not to ward off arthritis however, but rather to assure for himself<br />

the benevolence of Venus’’ (19).<br />

As described by Kelly Patricia O’Meara in a Washington Times article<br />

on POWs/MIAs (February 7, 2000), copper bracelets became immensely<br />

popular in 1970 when three American college students began making them<br />

to draw attention to missing-in-action servicemen. The original bracelets<br />

were modeled on a metal bracelet that Robert Dornan (later a Congressman)<br />

obtained from hill tribesmen in Vietnam. Called MIA bracelets, each<br />

had the name of a serviceman missing in Vietnam inscribed on the outside.


242 Harrison<br />

They sold at the time for $3 each, became very popular, and eventually were<br />

worn by 5 million Americans. They are still worn today.<br />

In a 1982 essay, Dr. Gerald Weissmann, NYU School of Medicine,<br />

speculated that the origin of copper adornments as a remedy for arthritis<br />

might have lain with the astrology followers of the 1960s, since the bracelet<br />

wearers Weissmann (26) interviewed talked about ‘‘ ...the healing power of<br />

the metal and the conjunction of the stars ... clear connection between the<br />

planets in their orbit and their metal talisman ... clear faith that gravitational<br />

and electrical forces generated by heavenly bodies were transmitted<br />

to their joints by way of the copper bracelet.’’<br />

It would seem then that the copper bracelet (for arthritis) myth might<br />

have an origin more recent than ‘‘ancient times,’’ and more rooted in superstition<br />

and magic than in any deep, mysterious scientific knowledge. Many<br />

of the ancient texts and new age websites allude to the mystical connection<br />

between elemental copper and the planet Venus.<br />

Consideration of the Reputed Power of the Remedy<br />

For the majority of arthritic conditions (gout is a notable exception), there is<br />

no known cause or cure. There are, however, effective drugs that can slow<br />

the progress of the disease and prevent irreversible joint damage. A number<br />

of ‘‘self-care’’ measures (including relaxation, diet, and exercise) are also<br />

helpful and are routinely recommended by doctors. Copper bracelets are<br />

not among the measures generally regarded as having any effect. Claims<br />

have also been made for a number of dietary supplements, some of which<br />

include copper; however, as of December 1996 the U.S. Food and Drug<br />

Administration (FDA) had not authorized any health claims for any food<br />

or dietary supplement and arthritis (27). This situation remains unchanged<br />

in 2005 according to information on the FDA website.<br />

Consideration of the Naturalness of the Remedy<br />

Copper is a naturally occurring element; a copper bracelet is, of course, a<br />

fabricated object. ‘‘Natural’’ by itself does not confer safety (elemental<br />

arsenic is ‘‘natural,’’ but deadly in a sufficient dose). There is a hypothetical<br />

risk of suffering an accidental injury while wearing a copper bracelet—for<br />

example, an instruction on the Q-Ray Ionized Bracelet 1 website warns<br />

against wearing the bracelets while using an electric blanket ‘‘because the<br />

blanket could heat up the metal bracelet and cause it to burn your skin.’’<br />

Other than suffering from such an accident, the only imaginable risk is developing<br />

a skin reaction to the copper. Hostynek and Maibach (28) recently<br />

conducted an extensive review of available case reports and concluded,<br />

‘‘ ... true allergic reactions to copper appear rare, particularly those induced<br />

by skin contact ...’’ The authors could find very few studies reporting


Copper Jewelry and Arthritis 243<br />

reactions (allergic contact dermatitis and eczema) among copper jewelry<br />

wearers, and it is likely that some of those were actually caused by sensitivity<br />

to nickel, not copper.<br />

Consideration of the Proposed Underlying Mechanism<br />

of the Remedy<br />

In common with materials promoting other unproven remedies (treatments<br />

that have not been shown in repeated scientific studies to work and to be<br />

safe), claims on the bracelet web pages range from the plausible to the<br />

absurd; a survey of the pseudoscience on these pages would make an interesting<br />

paper on its own. Many betray an underlying misunderstanding of basic<br />

science. ‘‘Negative and positive’’ charges and fields echo the pseudoscientific<br />

claims behind magnets and magnet therapy, also in vogue (29). There seems<br />

to be indecision on whether bracelets work by making something flow into or<br />

out of the body. Some sites do cite the scientific literature, although many do<br />

it in a highly selective manner, reprinting a sentence or two to support their<br />

claims. Among the scientific ‘‘facts’’ that are correctly claimed are the essentiality<br />

of copper, its role in superoxide dismutase and antioxidant role,<br />

formation of chelates with substances (amino acids) in sweat, and the possible<br />

role of copper deficiency in the disease. A few sites attempt a balanced<br />

treatment of the current state of scientific evidence and correctly cite supporting<br />

publications but they are the exception.<br />

THE COPPER BRACELET TRIAL<br />

Scientific Evidence for the Use of Bracelets for Arthritis<br />

Therapy Considered<br />

The International Copper Research Association in 1975 published a critical<br />

review of copper in medicine. The authors advocated the use of copper chelates<br />

as anti-inflammatory agents for arthritis treatment. Interestingly, while<br />

copper bracelets were covered briefly, their use was suggested as a means of<br />

exploiting copper’s bactericidal and fungicidal properties:<br />

Finally, it should be kept in mind that people who have a high frequency<br />

of minor skin afflictions of a fungal or a bacterial type,<br />

rather than using copper salts in liniment or salves, may wish to<br />

use the much more convenient metallic form of copper: say in the<br />

form of a copper barrette, a copper chain, or a copper bracelet worn<br />

close to the skin (21). Such a metallic copper device will slowly<br />

release copper ions, most probably in the form of amino acid complexes,<br />

which will permeate both the surrounding skin and eventually<br />

also the body. A maximum figure of the amount of copper released<br />

most likely will not exceed that released from the Copper-T device,<br />

about 45 mg per day. Such a slow, daily release of ionic copper will


244 Harrison<br />

act as a bactericide and fungicide on the skin environment of the<br />

metallic copper. At the same time, it cannot cause any harmful general<br />

effects to the individual, since the amount of copper released<br />

will only constitute a small percent (


Copper Jewelry and Arthritis 245<br />

A word of caution about efficacy—proposed medical therapies must be subject<br />

to three tests: efficacy, or the ‘‘extent to which an intervention does more<br />

good than harm under ideal circumstances’’ (can it work?); effectiveness,<br />

‘‘whether an intervention does more good than harm under usual circumstances<br />

of healthcare practice’’ (does it work in practice?); and efficiency,<br />

‘‘effect of an intervention in relation to the resources it consumes’’ (is it<br />

worth it?) (38). It must be noted that it is not necessary for any therapy<br />

to be fully understood (only that it works).<br />

In 1974, Ray Walker was a university researcher with an interest in<br />

coordination chemistry. For the initial copper bracelets trial, volunteers<br />

with self-declared ‘‘arthritis’’ were recruited by publishing letters in several<br />

Australian newspapers. A substantial number of volunteers (60 of the 300<br />

who answered a questionnaire) were rejected by a physician on the grounds<br />

that they did not fit an arthritic profile (based on an examination of their<br />

questionnaire responses); there was no direct assessment of their health<br />

and so we cannot be certain of the diagnosis of the trial participants (39).<br />

Roughly half reported having worn copper bracelets for arthritis. For the<br />

trial, the remaining 240 volunteers were randomly assigned (randomization<br />

method not specified) to one of three groups: Group I wore a copper bracelet<br />

for one month followed by an anodized aluminum bracelet (placebo) for<br />

one month; Group II did the reverse; and Group III (control) wore no bracelet<br />

for two months. Bracelets were weighed at the beginning and end of the<br />

period and the groups comprised equal numbers of experienced and inexperienced<br />

users. A subjective assessment was made by questionnaire after the<br />

end of the two-month trial—the evaluation categories were ‘‘better’’ (including<br />

both ‘‘much better’’ and ‘‘little better’’), ‘‘same,’’ and ‘‘worse’’ (including<br />

‘‘little worse’’ and ‘‘much worse’’) (35). In the evaluation, a significantly<br />

greater number of the patients who expressed a preference reported that<br />

they ‘‘were better’’ while wearing the copper bracelet than while wearing<br />

the aluminum bracelet. The effect was attributed to the supply of a steady<br />

supplemental amount of copper from the bracelets; copper bracelets were<br />

reported to have lost an average of 13 mg in a month of use and the lost<br />

copper was assumed to have dissolved into the wearers’ sweat and then to<br />

have been absorbed into their skin. Subsequent laboratory experiments<br />

showed that copper from turnings could dissolve into sweat and that copper<br />

complexed to an amino acid perfused cat skin (40,41). The trial results were<br />

reported in preliminary form in 1976, and the data were reported at least<br />

twice again (35–37).<br />

It is difficult to make a confident assessment of what was proved by<br />

the Walker and Keats bracelets trial, and since its publication the study<br />

has been criticized on several grounds:<br />

1. It was poorly controlled (15,42). Because copper bracelets discolor<br />

and turn the wearer’s skin green during use, it is doubtful that the


246 Harrison<br />

experienced users (and even a proportion of the inexperienced<br />

ones) could not distinguish the real thing from the placebo. This<br />

calls into question the ‘‘blinding’’ of the subjects in the trial since<br />

the patient must be unable to distinguish the treatment from the<br />

placebo (43). If either the researcher or patient can guess which<br />

is the real treatment and which is the sham, the study results<br />

may be biased (44). Further, such inadequate concealment in pain<br />

research and complementary and alternative medicine (CAM)<br />

trials results in the treatment effects being overestimated (45,46).<br />

There is no indication in the three papers about the Walker and<br />

Keats trial that the investigators were blinded to the treatment versus<br />

placebo bracelets when weights were taken—in fact there was<br />

no indication that the placebo bracelets were weighed at all (if they<br />

were, no data were presented for them). To establish an effect of a<br />

treatment ‘‘above and beyond natural history of the condition and<br />

non-specific effects,’’ it is essential that the trial is randomized,<br />

controlled and (preferably) double-blinded (44). The Walker and<br />

Keats trial did not meet this standard.<br />

2. The Walker and Keats trial used a vaguely defined outcome (‘‘perceived<br />

effectiveness’’) determined by a subjective evaluation by<br />

questionnaire at the end of each one-month treatment period<br />

and a comparison of the relative effectiveness of the bracelets at<br />

the end of the second period. The data presented are in categories<br />

(‘‘a little worse,’’ ‘‘much better,’’ etc.) but exact criteria for these<br />

evaluations are not specified. It would appear that ‘‘better’’ or<br />

‘‘worse’’ refers to a superficial judgment of the ‘‘effectiveness’’ of<br />

the copper bracelet versus the placebo bracelet, not to the symptoms<br />

of the wearer per se and may be based on the wearers’<br />

appraisal of the ‘‘state of their condition’’ (37). Combining ‘‘a little<br />

better’’ and ‘‘much better’’ into a single category may exaggerate<br />

the apparent effectiveness of the treatment bracelets.<br />

3. In conditions such as arthritis that are characterized by periods of<br />

remission and flare, ‘‘it is not uncommon for two out of every<br />

three patients to report subjective improvement after a treatment<br />

that, unknown to them, contains no active agent’’ (47). There is<br />

a strong suggestion of this in the Walker and Keats (35) data.<br />

A statistical test of the effect of previous experience on the experimental<br />

outcome showed that it was significantly related to the<br />

evaluation provided by the volunteers (35). The largest group of<br />

volunteers overall (30 of 77) perceived no difference in effectiveness<br />

between the copper and aluminum bracelets. The natural<br />

history of their conditions over the two-month trial period could<br />

be expected to significantly affect their perception of the effectiveness<br />

of the bracelet they were wearing during that part of the trial.


Copper Jewelry and Arthritis 247<br />

This phenomenon is common to many pain-causing conditions<br />

and greatly complicates the design of trials in pain research (44).<br />

Moreover, patients often enroll in clinical trials when their symptoms<br />

are at their worst—and ‘‘the next change is likely to be an<br />

improvement’’ (44).<br />

4. The placebo effect, ‘‘a change in a patient’s illness attributable to<br />

the symbolic import of the treatment rather than a specific pharmacologic<br />

or physiological property’’ (44), can be powerful in pain<br />

research. Paracelsus understood the importance of the placebo<br />

effect (or wishful thinking): ‘‘It is not the curse or the blessing that<br />

works, but the idea. The imagination produces the effect’’ (29).<br />

The data for the Walker and Keats trial volunteers who wore no<br />

bracelet at all for the two months (control group) were even more<br />

interesting and suggestive that a strong placebo effect might have<br />

operated in this trial. While 11 of 19 of the previous users said they<br />

were ‘‘worse’’ during the trial period (seven reported ‘‘same’’ and<br />

one ‘‘better’’), the previous nonusers were evenly split, with six<br />

reporting ‘‘worse,’’ six ‘‘better,’’ and 10 reporting no difference (35).<br />

The authors also observed that, of the previous users, 14 dropped<br />

out of the trial because they could not be without their bracelets.<br />

Assuming an initial pool of 40 volunteers in that part of the<br />

control group, seven other testers dropped out for other (unspecified)<br />

reasons. The attrition loss in the ‘‘previous nonusers group’’<br />

was 18—for reasons also not specified in the paper. Did many<br />

‘‘control’’ subjects just get better anyway and so lose interest in<br />

completing the trial? Patient expectations of success of the treatment<br />

are an important component of the placebo effect (44,48)<br />

and highly compliant patients may show better outcomes, just as<br />

they did in the Walker and Keats trial. While all participants were<br />

instructed to carry on normal routines, diet, and medications for<br />

the duration of the trial (35), no data are given on the use of medications<br />

during the trial that might have had a significant influence<br />

on the outcome.<br />

5. Bracelet weight loss was claimed by the authors to supply up to<br />

13 mg/month supplemental copper. It was argued that this copper<br />

dissolved into the sweat beneath the bracelet and perfused the skin<br />

of the wearer. There are some difficulties with the weight loss<br />

claim, however. No error estimates for the calculated mean weight<br />

loss are given in the original Walker and Keats report or subsequent<br />

republications. The bracelets were weighed before the trial<br />

and then sent in plastic bags to the subjects. They were sent back<br />

to the researchers at the end of the trial, ‘‘usually’’ in plastic bags,<br />

at which time they were again weighed. Importantly, an unspecified<br />

number of bracelets were observed to have gained weight over


248 Harrison<br />

the two-month trial period. The researchers took this as evidence<br />

that some bracelets had not been worn—those bracelet weights<br />

and the questionnaire responses of those participants were then<br />

eliminated from the analyses—a substantial source of bias for both<br />

the test and the bracelet weight loss claim. We are not told how<br />

many bracelets were in this group, but of the 160 testers, only<br />

77 wore the bracelets for the full trial, filled out all questionnaires,<br />

supplied classifiable responses, and had bracelets that did not gain<br />

weight. It would have been interesting to know what weight<br />

change was observed in the placebo bracelets and if the trial participants<br />

were able to guess which bracelet (copper or placebo) they<br />

were wearing. In a review discussing the results of the Walker<br />

and Keats trial, the authors admit that the bracelet weight loss<br />

observed might have been caused by mechanical wear, although<br />

in an earlier publication they reported that the used bracelets<br />

did not appear abraded (39,40). Walker et al. (36) also remarked<br />

that the appearance of a green stain on the skin below bracelets<br />

[from copper (II) salt build-up] could be seen as evidence that<br />

some copper must be lost from the bracelet during wear.<br />

6. In a subsequent experiment, copper was shown to dissolve from<br />

turnings that had been submerged and shaken in human sweat<br />

for 24 hours (40). Extrapolating from this result to the dissolution<br />

of copper from a solid bracelet into a thin film of sweat on a<br />

human arm would seem problematic. To begin with, the turnings<br />

present a substantially greater surface area for dissolution compared<br />

with a solid bracelet. Since study participants would have<br />

bathed regularly, it is likely that an undetermined amount of the<br />

copper (green stain) under their bracelets would simply wash off<br />

the skin’s surface and not be absorbed.<br />

7. The trial questionnaires included a semiquantitative assessment of<br />

the diet of study participants. Copper-rich foods (liver, shellfish,<br />

mushrooms, and nuts) were an insignificant part of the diet of<br />

the study participants (35). This is not surprising, since for most<br />

Western diets, these copper-rich sources do not play a large part;<br />

in fact, grains and vegetables commonly provide up to 60% of copper<br />

intake (49). The observation that most of the participants’<br />

diets were in the ‘‘low’’ category for intake of copper-rich foods<br />

is typical of populations as a whole and does not indicate anything<br />

useful about what the overall dietary copper intake of study subjects<br />

might have been—it certainly cannot be taken as robust<br />

evidence that the participants were copper deficient.<br />

Walker and Keats were initially tentative in their support for the therapeutic<br />

value of the bracelets, and suggested in their original paper that the


Copper Jewelry and Arthritis 249<br />

subjects who had not been bracelet wearers before the trial may have been<br />

‘‘less ready to commit’’ themselves. In a 1977 letter in The Medical Journal<br />

of Australia, Whitehouse and Walker (50) mention that the ‘‘Pharmacy Board<br />

in Victoria is currently reconsidering its ban on the sale of copper bracelets’’<br />

and that there had been discussion of the issue in the lay press. It is possible<br />

that people who volunteered for the trial might have felt quite passionate<br />

about the issue before the trial and that their enthusiasm could have affected<br />

the trial’s outcome, as the investigators themselves indicated. In calling for<br />

trials to fully validate the role of the bracelet, the researchers concluded, ‘‘even<br />

if it has little value, except as a placebo, it does have the merit of being of little<br />

harm (by virtue of being the most inefficient source of a potential toxin)’’ (50).<br />

Further well-designed and properly conducted clinical trials, rigorously controlled<br />

for placebo effects, and double blinded, would be necessary to confirm<br />

or refute the findings of the Walker and Keats trial and prove or disprove<br />

finally the claims of the promoters of copper bracelets.<br />

The study of ‘‘the copper bracelet myth’’ and the science behind it led<br />

to the development of a topical anti-inflammatory copper-salicylate gel<br />

(36,37,51) that is available as an over-the-counter product in several countries.<br />

An independent, double-blind, placebo-controlled, randomized trial<br />

of the gel failed to confirm its effectiveness: applied to the forearm it was<br />

no better than the placebo gel for pain relief in patients with osteoarthritis<br />

of the hip or knee (52). The treatment gel also produced significantly more<br />

skin rashes than the placebo. In 1998, The Medical Journal of Australia<br />

published a letter critical of the trial by one of the gel’s developers (and<br />

a colleague of Dr. Walker’s). Dr. Boettcher criticized the gel study on<br />

the grounds that the gel should have been applied (as directed) at or near the<br />

hip or knee and the composition of the gel was incorrectly described in<br />

the article (53). Dr. Boettcher also took exception to the statistical analysis<br />

used. Drs. Brooks and Kellett (54) countered these objections—the trial was<br />

designed to investigate systemic effects on distant joints—and they refuted<br />

Dr. Boettcher’s statistical argument.<br />

Investigation of the effectiveness of topical therapies for arthritis pain<br />

relief continues—results of several substantial studies have been published<br />

recently. A meta-analysis of randomized controlled trials of topical NSAIDs<br />

(popular over-the-counter drugs) used for the relief of osteoarthritis pain<br />

concluded that they are no more effective than a placebo beyond two weeks<br />

of treatment (55). A systematic review of double-blind, placebo-controlled<br />

trials for efficacy concluded, ‘‘topical NSAIDs were effective and safe in<br />

treating acute painful conditions for one week’’ (56).<br />

A New Bracelet Study<br />

A new study of the effects of copper/zinc bracelets on joint and muscle pain<br />

was conducted at the Mayo Clinic and published by them in 2002 (57). The


250 Harrison<br />

study was designed as a randomized, double-blind, placebo-controlled trial<br />

of ‘‘ionized’’ wrist bracelets that were advertised and marketed as effective<br />

for the treatment of pain. Because the bracelets (both ‘‘ionized’’ and placebo)<br />

used in the trial were made mostly of copper (85% copper/15% zinc)<br />

they are relevant to the discussion of the Walker and Keats bracelets trial.<br />

There are interesting similarities between the two studies:<br />

1. The subjects for both studies were volunteers with self-reported<br />

pain (in the Mayo Clinic study they were recruited by posters in<br />

the clinic).<br />

2. The patients in both studies wore bracelets for a one-month test<br />

period.<br />

3. In both studies, patients replied to questionnaires on which (among<br />

other questions) they rated their own conditions subjectively.<br />

4. The placebo effect was evident in both trials.<br />

5. In both trials, a proportion of the patients believed in the power of<br />

the remedy beforehand.<br />

In the Mayo Clinic study, participants were asked in advance if they<br />

believed the bracelets would be effective and, of the participants who<br />

answered the question, 80% replied in the affirmative; however, only 4.4%<br />

reported having ever used a bracelet before the trial. Patients wearing both<br />

placebo and ‘‘ionized’’ bracelets reported statistically significantly lower<br />

pain scores during the course of the trial, but the two groups showed the<br />

same degree of improvement, leading the authors to conclude that the treatment<br />

bracelets were no better than the placebos.<br />

The new study differed in methodology in significant ways from the<br />

Walker and Keats trial. Firstly, the Mayo Clinic study used a more clearly<br />

defined outcome (pain rated on a scale of 1–10) and they evaluated pain at<br />

several points during the month (days 1, 3, 7, 14, 21, 28). Remarkably, on<br />

day 1, 63.3% of the ‘‘ionized’’ bracelet wearers and 68.5% of the placebo bracelet<br />

wearers reported an improvement in maximum pain scores—from the<br />

study’s description, there is good reason to expect that an especially strong<br />

placebo effect may be evidenced. As part of the Mayo Clinic study design,<br />

patients were instructed in the correct placement of the bracelets according<br />

‘‘to the manufacturer’s recommendations’’ (57)—a procedure that very<br />

likely induced additional confidence in the patient that the remedy was likely<br />

to be powerful, judging by the manufacturer’s instructions available on their<br />

website—in particular, the specific instructions for changing the orientation<br />

of the terminals if the bracelet is worn on the left instead of (recommended)<br />

right wrist; the implication of the term ‘‘terminal’’ for the bobble on the<br />

open end of the bracelet; the instruction that the ‘‘terminals’’ should be 1 ⁄2<br />

to 1 1 ⁄2 inches apart during use; and the warning, ‘‘Do not let the terminals<br />

directly touch each other.’’ As Bratton et al. (57) point out, going through<br />

these instructions with patients is an excellent example of the ‘‘healing


Copper Jewelry and Arthritis 251<br />

ritual’’ described by Kaptchuk (48) as producing an ‘‘exaggerated placebo<br />

effect’’ and it could well explain the observation that 2/3 of the participants<br />

reported an improvement after just one day of wearing the bracelets. Interestingly,<br />

although 80% of the participants answering the question reported<br />

ahead of time that they believed the remedy would work, the highest proportion<br />

reporting an improvement in pain scores during the trial was 77.7%.<br />

Unfortunately, the Mayo Clinic study did not have a control group that<br />

wore no bracelet at all during the trial—this could have provided valuable<br />

information about the significance of the pain improvements (which were<br />

not large) reported by both groups of study participants.<br />

In 2003, the U.S. Federal Trade Commission (58), citing the Mayo<br />

Clinic study, charged the marketers of the Q-Ray Ionized Bracelets with<br />

making false and unsubstantiated claims, specifically ‘‘deceptively claiming<br />

that the Q-Ray Bracelet is a fast-acting effective treatment for various types<br />

of pain and that tests prove that the Q-Ray Bracelet relieves pain.’’<br />

HealthScout News reporter Ed Edelson concluded an online article about<br />

the study (HealthScout News, November 12, 2002) with this wry comment<br />

from Dr. Robert Bratton, the study’s lead author: It’s all a placebo effect and<br />

‘‘based on the study results, you may be just as well off wearing a rubber<br />

band around your wrist and saving the money spent on the bracelet.’’<br />

The Gold Ring Study<br />

Gold treatments (by injection) are used in the treatment of rheumatoid<br />

arthritis and an intriguing study published in 1997 provided some evidence<br />

that gold from rings may delay articular erosion in rheumatoid patients (59).<br />

In the study, confirmed rheumatoid arthritis patients were divided into two<br />

groups—non-ring wearers and ring wearers—and their hands and wrists<br />

examined by standard radiographic technique. While the two groups had<br />

statistically indistinguishable overall articular erosion, the fingers of the<br />

ring-bearing hands were significantly less deformed. While the study was<br />

small (55 subjects in total), and, as the authors admit, it is possible that the<br />

effect could relate to the different pattern of use between the left (ringbearing)<br />

and right hand, it does suggest that gold from the rings might<br />

provide a protective effect.<br />

THE PRESENT STATE OF THE COPPER BRACELETS ‘‘ISSUE’’<br />

More Recent Studies Have Been Less Promising or Just How<br />

Satisfied Are Bracelet Users?<br />

Two features of the most common arthritic conditions make arthritis<br />

patients vulnerable to the lure of alternative remedies: their conditions have no<br />

known cause or proven cure and they are characterized by chronic pain (33).<br />

In 1997, American consumers spent approximately $27 billion on alternative


252 Harrison<br />

therapies (60). Alternative therapies are ‘‘aggressively promoted’’ and, in<br />

the opinion of D.M. Marcus (61), MD, Baylor College of Medicine, ‘‘most<br />

AM [alternative medicine] information that is readily available to lay persons<br />

is misleading or wrong.’’ This point of view is supported by a survey<br />

made in May 1998 of Internet information for patients with rheumatoid<br />

arthritis. After evaluating the quality and source of materials offered its<br />

authors concluded, ‘‘over two-thirds of the sites with overt financial aims<br />

promoted the use of alternative therapy, often claiming that their products<br />

were effective for arthritis and other conditions ... Although some alternative<br />

therapies may be efficacious, most have not been adequately studied,<br />

and often, positive studies have major methodological flaws’’ (62).<br />

Surveys of patterns of usage of copper bracelets have been conducted<br />

in Australia/New Zealand, North America, and the United Kingdom. Let<br />

us suppose that it is true that copper bracelets provide great symptomatic<br />

relief for arthritis sufferers. Since they are widely available, inexpensive,<br />

and heavily promoted, one would expect to observe high rates of bracelet<br />

use among patients, and patients should show high rates of satisfaction<br />

(and continued use). There is evidence to suggest that patients are generally<br />

aware of the copper bracelet claims. For example, 75% of the 51 Hawaiian<br />

patients with inflammatory arthropathies in a Hawaiian survey had heard of<br />

copper bracelets as an alternative therapy (63). Findings from this and several<br />

other patient surveys are summarized in Table 1. Patients reported that<br />

they turn to CAMs to relieve pain; many discontinued conventional medications<br />

because they found medication schedules complicated (71).<br />

Patients also perceive the inability of conventional therapies to cure or<br />

adequately control their disease and the potentially serious adverse effects they<br />

may cause as reasons to turn to alternative treatments (72). Well-publicized<br />

Table 1 Use of Copper Bracelets/Bands by Arthritis Patients<br />

Location<br />

No. of<br />

patients using<br />

% of survey<br />

population<br />

Age of patients<br />

(yrs) Citation<br />

Australia 45 61 15–65þ (64)<br />

Australia/<br />

New Zealand<br />

21 70 12.1–27.2 (65)<br />

Australia 3 3 Mean ¼ 61.1 (66)<br />

Canada 4 57 9.3–19.3 (65)<br />

Canada 16 11 Mean ¼ 10.4–10.9 (67)<br />

England 75 38 N/A (68)<br />

England 45 28 N/A (69)<br />

United States 17 13 Adults (70)<br />

United States 42 29 Mean ¼ 55.5 (60)<br />

United States 11 21.6 Mean ¼ 51 (63)


Copper Jewelry and Arthritis 253<br />

side effects of conventional medicine (in contrast with the natural/safe/<br />

gentle claims for CAMs) provoke an ‘‘exaggerated fear of conventional<br />

medications’’ among arthritis patients (61). Sometimes, new studies show that<br />

patients’ concerns about the potential side effects of conventional drug therapies<br />

are justified. For example, in September 2004, it was revealed that the<br />

popular Merck arthritis and acute pain medication VIOXX 1 (rofecoxib), with<br />

worldwide sales in 2003 of $2.5 billion, increased the relative risk of heart<br />

attack and stroke when used for more than 18 months. The press coverage<br />

of this announcement and subsequent negative studies on VIOXX was extensive<br />

and featured such frightening headlines as this one that appeared in The<br />

Times (London) online edition on January 25, 2005: ‘‘VIOXX may have killed<br />

thousandsofUKpatientsstudywarns.’’<br />

CAM users tend to be female, ‘‘better educated,’’ and employed.<br />

Many arthritis patients use CAMs to supplement their conventional treatment<br />

and report that friends and relatives and the mass media are their main<br />

source of information on alternative treatments. In a recent survey of arthritis<br />

patients in New Mexico, for example, the most frequently cited sources of<br />

information on CAMs were family and friends (66.1%), medical doctors<br />

(56.1%), magazines and books (34.6%), radio/TV/newspapers (22.6%), and<br />

the Internet (11.3%) (73).<br />

As for effectiveness, in one survey, 74% of adult Americans polled<br />

expressed a belief that copper bracelets would help treat arthritis; tellingly,<br />

only 13% of arthritis patients reported having used them (70). Several small<br />

studies have reported on the success rate claimed by arthritis patients for<br />

various alternative and complementary therapies (Table 2). Overall, copper<br />

bracelets did not fare well in these surveys. Among the therapies with a<br />

Table 2 Patient Satisfaction Level with Bracelet Use: Proportion of Patients Using<br />

or Having Used Copper Bracelets Reported to Have Found Them to Be ‘‘Helpful’’<br />

Location<br />

Patients reporting a benefit from using bracelets<br />

No. (%)<br />

using<br />

bracelets<br />

No. (% of users)<br />

reporting benefit or<br />

currently using<br />

% total<br />

patient<br />

population n Citation<br />

Australia 45 (61) 6 (13) 8 74 (64)<br />

England 75 (38) 6 (8) 3 199 (68)<br />

United States 11 (21.6) 4 (36) 7.8 51 (63)<br />

United States 42 (29) a<br />

9 (21) b<br />

6 146 (60)<br />

United States N/A (3.9) N/A 612 (73)<br />

a<br />

‘‘Ever used’’ includes both presently using and used in the past.<br />

b<br />

Compare with clinical improvement observed in 30% to 35% of patients treated with a placebo<br />

in clinical drug trials (42).


254 Harrison<br />

higher reported ‘‘usefulness’’ or success rate are chiropractic, herbal therapies,<br />

nutritional supplements, medication/relaxation, electrical stimulators, special<br />

diets (especially avoidance of certain foods), acupuncture/acupressure,<br />

osteopathy, and faith healing. Most unproven remedies, including copper<br />

bracelets, are considered ‘‘harmless,’’ with the caution that even innocuous<br />

therapies could be harmful if diagnosis is delayed (especially with the advent<br />

of aggressive therapy early in the course of the disease that has been shown to<br />

prevent permanent joint damage) or proven therapy neglected (74).<br />

Rao and coworkers (75) conducted a longitudinal study on the same<br />

group of rheumatology patients they reported on in 1999, with follow-up<br />

surveys at six and 12 months. Over the one-year period, three patients<br />

(out of 174) started using a copper bracelet, three maintained their use,<br />

and five stopped using a bracelet. Of the 28 patients in their study who<br />

reported having ‘‘ever used’’ a copper bracelet but had stopped, 24 gave<br />

‘‘did not help’’ as a reason, one claimed to have been made worse by wearing<br />

a copper bracelet (two said ‘‘other’’ as a reason, and one declined to say) (75).<br />

In a recent study, a group of 40 Asian and 40 Caucasian rheumatoid arthritis<br />

patients in the United Kingdom were asked to provide a subjective<br />

ranking of effectiveness of complementary therapies on a five-point scale;<br />

‘‘The two groups rated copper bracelets and magnets very similarly, only<br />

just above useless’’ (76).<br />

Additional Research and Scientific Data Needed to<br />

Resolve the Copper Bracelets Issue<br />

The precise ‘‘ ... role of copper in inflammatory processes remains quite<br />

obscure. Both pro- and anti-inflammatory effects of copper have been documented’’<br />

(15).<br />

The studies by Ray Walker and colleagues were visionary, but inadequate<br />

to establish or refute the efficacy of copper bracelets for arthritis<br />

therapy. The comment is made repeatedly, in the scientific and other literature,<br />

that the available data are not sufficient to constitute a proof of the<br />

effectiveness of externally worn copper as therapy for arthritis. The original<br />

trials could certainly be repeated and improved on by:<br />

recruiting known patients with clearly defined and medically<br />

verified disease;<br />

better screening patients for prior bias about the therapy;<br />

improving placebo design and control;<br />

using objective measures of pain and disease activity as measurable<br />

study outcomes;<br />

rigorously measuring bracelet weight changes during wear, establishing<br />

a rate and accounting for loss of weight due to abrasion and/<br />

or increase in weight, and using a statistically reliable method of<br />

determining weight change;


Copper Jewelry and Arthritis 255<br />

modeling corrosion of copper from bracelets and complex formation<br />

in sweat during use; and<br />

using a properly double-blinded study design.<br />

Recent surveys of the use of complementary and alternative therapies<br />

by arthritis patients do not lend strong support to the belief that bracelets<br />

are found to be of much use and this must call into question any impulse<br />

to do more research on them. As discussed above, convincing evidence that<br />

bracelets exert more than a placebo effect is lacking. A survey of 2146 primary<br />

care physicians and rheumatologists ranked copper jewellery (at 25%)<br />

fifth on the list of ‘‘top 10 alternatives doctors advise against.’’ In a parallel<br />

survey of 790 people with arthritis who read Arthritis Today, ‘‘metal jewellery’’<br />

was an alternative reported as used by only 15% of the survey respondents<br />

(77). A search of the new National Institutes of Health (NIH) Clinical<br />

Trials.gov on October 20, 2004, found 136 ongoing studies on ‘‘arthritis’’<br />

but none involving copper bracelets. The nonprofit agencies and foundations<br />

dedicated to the well-being of arthritis sufferers and governmental regulatory<br />

agencies are similarly unenthusiastic about copper bracelets as a folk<br />

remedy; their position statements are compiled in Appendix A.<br />

On the other hand, there is a definite need to establish the extent of<br />

dermal absorption of copper from a metallic source. ‘‘Dermal exposure to<br />

copper can result from the use of consumer products containing copper pigments,<br />

through the use of copper as an agicide in swimming pools and the use<br />

of copper jewellery. No quantitative exposure levels could be found’’ (78).<br />

There remains a need for objective, stringent human studies involving<br />

copper and arthritis. Several lines of investigation that have been proposed<br />

over the years seemed interesting and yet, to my knowledge, remain to be<br />

fully studied:<br />

1. The role of dietary copper deficiency in the etiology of arthritis and<br />

the use of supplemental copper (or copper-rich diets) in arthritis<br />

treatment:<br />

A small double-blind, double placebo controlled factorial trial<br />

of copper (in a complexed form at 3 mg/day) and/or omega-3<br />

fish oil supplements showed no significant effect of copper on<br />

systemic lupus erythematosus disease activity—a significant<br />

decline in disease activity was demonstrated for the fish oil<br />

supplement (79).<br />

2. Quantification of the supplemental supply of copper from an<br />

intrauterine device (IUD) and an epidemiological study of arthritis<br />

among copper IUD users.<br />

3. The mechanism of formation of copper complexes with salicylates<br />

from arthritis analgesic products in vivo and how that increases<br />

their effectiveness.


256 Harrison<br />

IS THERE LIKELY TO BE A FUTURE FOR COPPER BRACELETS IN<br />

ARTHRITIS CARE?<br />

In Geneva in January 2000, the World Health Organization launched<br />

The Bone and Joint Decade: 2000–2010 (80). In recognition of the immense<br />

burden of the disease, the U.S. Congress initiated a national arthritis prevention<br />

program with a budget of $12 million for the year 2000 (2). One<br />

of its goals was to strengthen the science base for arthritis treatment options,<br />

especially the roles of good nutrition, appropriate exercise, evaluation of<br />

intervention strategies including self-help patient education programs, and<br />

promoting good communication. There is good reason to suppose that the<br />

therapeutic use of copper for arthritis will be a recurring issue. Copper bracelets<br />

are not the only copper product to be touted as therapeutic. America’s<br />

first great medical quack claimed to cure disease with copper rods passed over<br />

the body (81). The Internet is a rich source of unproven and quack remedies<br />

involving copper, and it is likely that more will spring up from time to time<br />

(Appendix B). The Internet makes it much simpler and less expensive for<br />

unproven claims to be widely promoted.<br />

In 1999, the establishment of the National Center for Complementary<br />

and Alternative Medicine (NCCAM) under the U.S. National Institutes of<br />

Health signaled a shift in the American medical establishment’s approach to<br />

alternative therapies. Scientists had been understandably reluctant to risk<br />

ridicule by attempting serious study of therapies not recognized by traditional<br />

medicine. Ray Walker experienced this first hand while embarking<br />

on the investigation of the copper bracelet by publishing the initial invitation<br />

to study participants in the popular press: ‘‘The letter caused some<br />

concern to several of my colleagues and raised the eyebrows of my medical<br />

friends. Although I had some qualms about my action, these were somewhat<br />

allayed by the work of Hagenfeldt ... and Okereke et al.’’ [(37); citing<br />

Hagenfeldt’s 1972 study on the mode of action of the copper-T IUD and<br />

Okereke, Sternlieb, Morell, and Scheinberg’s 1972 paper in Science on the<br />

systemic absorption of copper from an IUD]. NCCAM has become a formidable<br />

force in health research and for 2006 requested a budget of<br />

$122.1 million (82). Its activities include funding research projects and clinical<br />

trials (it identifies conditions affecting the elderly as a priority area for<br />

investigation), developing a comprehensive communications program, and<br />

training young scientists in CAM. In its first five years it supported more<br />

than 1200 projects on various CAMs, including clinical trials on acupuncture,<br />

mind–body medicine, and herbal products.<br />

The NCCAM is not without its critics (e.g., Ref. 83), and the conflict<br />

between conventional and alternative/complementary medicine is a hot<br />

topic in journals such as Journal of the American Medical Association,<br />

British Medical Journal, and the Medical Journal of Australia. The substantial<br />

research award amounts now available for studies on CAMs mean that


Copper Jewelry and Arthritis 257<br />

they are likely to continue to enjoy a public profile, particularly as more<br />

studies are completed, published, and covered in the popular press—as<br />

the example of the Mayo Clinic Q-Ray bracelet study demonstrated. The<br />

methods used to study CAMs are also likely to excite ongoing controversy.<br />

There is even an interesting discussion of whether CAMs should be evaluated<br />

in a fundamentally different way from conventional therapies. The<br />

placebo effect, for example, that played such a prominent role in the bracelet<br />

trials seems to play an important role in many alternative therapies. In a<br />

highly entertaining essay in the Annals of Internal Medicine, Doctor of<br />

Oriental Medicine (OMD) Ted Kaptchuk examines the possibility that there<br />

is some real value for the placebo effect in clinical practice. Kaptchuk’s (48)<br />

essay illustrates vividly a growing gulf between CAM proponents and practitioners<br />

of traditional science and medicine: ‘‘Some may dismiss these types of<br />

investigation as useless. After all, a placebo is just a placebo. Others would<br />

argue that such avoidance impoverishes and narrows the understanding of<br />

what patients receive from alternative medicine ...’’ Examination of complementary<br />

and alternative therapeutic approaches (especially emphasizing<br />

longitudinal studies) is a focus of the new ‘‘National Arthritis Action<br />

Plan’’ (74). There clearly needs to be much better patient education and public<br />

education about arthritis, and there is a definite need on the Internet for<br />

additional high-profile, reputable sites that meet the highest standards of<br />

scientific evidence. This might help to counteract the clamor of the lesscredible<br />

websites.<br />

Considering the scant evidence of the metal’s systemic absorption and<br />

anti-inflammatory effects in laboratory rats, one can safely say that<br />

nothing has been learned in the past 3,500 years that would explain<br />

the longevity of the belief in copper’s powers.<br />

—Sharon L. Kolasinski (84)<br />

APPENDIX A: POSITION STATEMENTS OF SUPPORT<br />

ORGANIZATIONS, GOVERNMENT AGENCIES, ETC.<br />

Arthritis Foundation (USA)<br />

From Arthritis Today magazine (available on the society’s website):<br />

‘‘there is no scientific research proving they provide any<br />

therapeutic benefit for arthritis. Nor is there any research proving<br />

they don’t.’’<br />

From their new guide to alternative therapies: ‘‘Expert Opinion:<br />

Wearing a copper bracelet won’t hurt, but there’s not enough scientific<br />

evidence that it helps’’ (85).


258 Harrison<br />

The Arthritis Society (Canada)<br />

‘‘On the hardware side of the store, things get even stranger:<br />

‘‘inductoscopes’’ (allegedly of magnetic induction), ‘‘solarama boards’’<br />

(to align mixed-up electrons), ‘‘earthboards’’ or ‘‘vitalators’’ (to<br />

produce rejuvenating electrons), ‘‘oxydonors’’ (to reverse the death<br />

process into the life process as a cure for arthritis) and, oh yes,<br />

copper bracelets. None of them has ever been proven effective in<br />

treating arthritis and relieving its pain’’ (86).<br />

Arthritis Foundation of Victoria (Australia)<br />

Under unproven remedies: ‘‘The use of copper has little scientific<br />

support. Copper bracelets are a popular alternative treatment for<br />

arthritis, but there have been no proper trials of copper bracelets<br />

in rheumatoid arthritis’’ (87).<br />

National Institute of Arthritis and Musculoskeletal<br />

and Skin Diseases, NIH (USA)<br />

‘‘Some of these alternative therapies have included wearing copper<br />

bracelets, drinking herbal teas, and taking mud baths. While these<br />

practices are not harmful, some can be expensive. They also cause<br />

delays in seeking medical treatment. To date, no scientific research<br />

shows these approaches to be helpful in treating osteoarthritis’’ (88).<br />

National Arthritis Action Plan (USA)<br />

‘‘Using unproven remedies for arthritis can waste time and money<br />

... may cause people to delay seeking early diagnosis and appropriate<br />

management. Because of their ongoing pain and lack of awareness<br />

of helpful intervention strategies, people with arthritis are particularly<br />

vulnerable to promoters of unproven remedies’’ (74).<br />

Federal Trade Commission—FTC (USA)<br />

‘‘Consumers spend an estimated $2 billion a year on unproven arthritis<br />

remedies—thousands of dietary and so-called natural cures, like ...<br />

magnets and copper bracelets. But these remedies are not backed by<br />

adequate science to show that they offer long-term relief’’ (89).<br />

Better Business Bureau of Eastern Massachusetts,<br />

Maine, and Vermont (USA)<br />

‘‘Quack arthritis cure ... Doctors say uranium is useless against<br />

arthritis, and a copper bracelet does nothing but turn your wrist<br />

green’’ (90).


Copper Jewelry and Arthritis 259<br />

Food and Drug Administration—FDA (USA)<br />

Under unproven remedies: ‘‘Some remedies, such as vinegar and<br />

honey or copper bracelets, seem harmless. But they can become<br />

harmful if they cause people to abandon conventional therapy’’ (27).<br />

APPENDIX B: MISCELLANY<br />

Another Copper Bracelet Myth<br />

Native North Americans recount a charming myth about a girl and a bear.<br />

It exists in several different versions and is a long and richly detailed tale.<br />

Briefly, a girl out gathering food slips on bear excrement (violating the<br />

bear’s mythical power, which is believed to be associated with its excrement).<br />

She utters an oath, further insulting the bear. The bear was considered the<br />

most powerful of animals—and thought to have the mystical powers of a<br />

shaman. The bear kidnaps the girl and leads her to a mystical world where<br />

it keeps the girl as its wife. The version collected at Bella Bella, British<br />

Columbia, includes this lovely detail: ‘‘He asks her what sort of excrement<br />

she has that would give her the right to scold him, and she says her excrements<br />

are abalone shells and copper. He tells her to sit down and show<br />

him, which she does, slipping off one of her copper bracelets’’ (25).<br />

Other Copper-Crafted ‘‘Healing Products’’ Found<br />

On the Internet:<br />

Copper cup (add warm water, let stand for 8–10 hours and drink<br />

the healing liquid).<br />

A healing wand of copper, brass, silver, quartz, and polished gemstones,<br />

with a hollow handle for addition of herbs, etc.<br />

A small copper disk to wear on the bottom of a wristwatch as an<br />

alternative to a bracelet.<br />

Copper patches as an alternative to copper bracelets.<br />

Mineral water enriched with copper (100 ppm) for joint problems.<br />

Copper coil placed in a warm water foot bath to draw toxins from<br />

the body through the feet and so treat a variety of ailments and<br />

restore energy.<br />

In the daily newspaper:<br />

Copper wires to insert in the nose to cure the common cold,<br />

described in a Reuters story carried by The Vancouver Sun on<br />

February 16, 2000.<br />

In the scientific literature:<br />

A copper bed sheet for treatment of the pain of fibromyalgia (91).


260 Harrison<br />

Early Copper Quackery from a Classic Book on Pseudoscience<br />

America’s first great quack was Dr. Elisha Perkins (1740–1799). The<br />

doctor had a theory that metals draw diseases out of the body, and in<br />

1796 patented a device consisting of two rods, each three inches long.<br />

One rod was supposed to be an alloy of copper, zinc, and gold; the<br />

other—iron, silver, and platinum. By drawing ‘‘Perkins’ Patented<br />

Metallic Tractor’’ downward over the ailing part, the disease was<br />

yanked out. Perkins sold his tractors for five guineas each to such<br />

notables as George Washington, whose entire family used it, and<br />

Chief Justice Oliver Ellsworth. His son, Benjamin D. Perkins (Yale,<br />

class of ’94) made a fortune selling the tractors in England. In<br />

Copenhagen, twelve doctors published a learned volume defending<br />

‘‘Perkinism.’’ Benjamin himself wrote a book about it in 1796, containing<br />

hundreds of stirring testimonials by well-educated people.<br />

They included doctors, ministers, university professors, and members<br />

of Congress. Most historians of the subject think the old man actually<br />

believed in his tractors, but that his son—who retired in New<br />

York City as a wealthy man—was simply a crook promoter. It is<br />

worth noting that orthodox medical opinion, by and large, ignored<br />

Perkinism, regarding it as not worthy of serious refutation. One doctor,<br />

however, did trouble to make some tests with phony tractors.<br />

They looked like the genuine article, but actually were non-metallic.<br />

His results, of course, were excellent. Oliver Wendell Holmes, in an<br />

amusing discussion of Perkinism, relates that one woman was quickly<br />

cured of pains in her arm and shoulder by using a fake tractor made<br />

of wood. ‘‘Bless me!’’ the woman exclaimed. ‘‘Why, who could have<br />

thought it, that them little things could pull the pain from one!’’ (81).<br />

Web Resources: Sites with Reliable Information on Arthritis<br />

American College of Rheumatology: http://www.rheumatology.org<br />

Arthritis Care (United Kingdom): http://www.arthritiscare.org.uk/<br />

Arthritis Foundation (United States): http://www.arthritis.org/<br />

Arthritis Foundation of Victoria (Australia): http://www.arthritisvic.<br />

org.au/<br />

Mayo Clinic: http://www.mayoclinic.com/<br />

National Center for Complementary and Alternative Medicine<br />

(NCCAM)(NIH): http://nccam.nih.gov/<br />

National Institute of Arthritis and Musculoskeletal and Skin Diseases<br />

(NIH): http://www.nih.gov/niams/<br />

NLM—National Library of Medicine: http://www.nlm.nih.gov/<br />

medlineplus/arthritis.html<br />

The Arthritis Society (Canada): http://www.arthritis.ca/home.html<br />

U.S. Food and Drug Administration: http://www.fda.gov/


Copper Jewelry and Arthritis 261<br />

A Final Word on Questionable Health Claims<br />

An intensive sweep of the Internet in 1998 targeted over 1200 sites making<br />

potentially false or deceptive advertising claims regarding the prevention,<br />

treatment, or cure of arthritis, cancer, diabetes, heart disease, AIDS, and<br />

multiple sclerosis. The ‘‘Health Claim Surf Day’’ spotlighted the tremendous<br />

power of the Internet as an information resource for consumers. It also<br />

uncovered the Web’s dark side: ‘‘it also provides promoters of fraudulent<br />

health products and treatments easy access to consumers from all over the<br />

world’’ (92).<br />

REFERENCES<br />

1. Sagan C. The demon-haunted world: Science as a Candle in the Dark. New York:<br />

Ballantine, 1996:20.<br />

2. CDC Coordinating Center for Health Promotion. Targeting arthritis: reducing disability<br />

for 43 million Americans. Atlanta: Centers for Disease Control and<br />

Prevention, March 2005. (http://www.cdc.gov/nccdphp/aag/aag_arthritis.htm).<br />

3. Lewis C. Arthritis: timely treatments for an ageless disease. FDA Consumer,<br />

May–June 2000. (http://www.fda.gov/fdac/features/2000/300_arth.html).<br />

4. Barceloux DG. Copper. Clin Toxicol 1999; 37:217.<br />

5. Milanino R, Marrella M, Velo G. Copper in the treatment of arthritic conditions:<br />

myth or reality? Chim Indust (Milan, Italy) 1994; 76:138.<br />

6. Sorenson JRJ. Copper complexes—a unique class of anti-arthritic drugs. Prog<br />

Med Chem 1978; 15:211.<br />

7. Mussalo-Rauhamma H, Konttinen YT, Lehto J, et al. Predictive clinical and<br />

laboratory parameters for serum zinc and copper in rheumatoid arthritis. Ann<br />

Rheum Dis 1988;47:816.<br />

8. Milanino R, Frigo A, Bambara LM, et al. Copper and zinc status in rheumatoid<br />

arthritis: studies of plasma, erythrocytes, and urine, and their relationship to disease<br />

activity markers and pharmacological treatment. Clin Exp Rheumatol 1993;<br />

11:271.<br />

9. Milanino R, Marrella U, Moretti U, et al. Copper and zinc status in rats with<br />

acute inflammation: focus on the inflamed area. Agents Actions 1988; 24:356.<br />

10. Milanino R, Moretti U, Concari E, et al. Copper and zinc status in adjuvantarthritic<br />

rat: studies on blood, liver, kidneys, spleen and inflamed paws. Agents<br />

Actions 1988; 24:365.<br />

11. Neve J, Fontaine J, Peretz A, et al. Changes in zinc, copper and selenium status<br />

during adjuvant-induced arthritis in rats. Agents Actions 1988; 25:146.<br />

12. Kishore V. Effects of copper aspirinate and aspirin on tissue copper, zinc, and<br />

iron concentrations following chronic oral treatment in the adjuvant arthritic<br />

rat. Biol Trace Elem Res 1990; 25:123.<br />

13. Aaseth J, Haugen M, Førre Ø. Rheumatoid arthritis and metal compounds—<br />

perspectives on the role of oxygen radical detoxification. Analyst 1998; 123:3.<br />

14. Sorenson JRJ. Copper-potentiation of non-steroidal antiinflammatory drugs. In:<br />

Berthon G, ed. Handbook of Metal–Ligand Interactions in Biological Fluids.<br />

Bioinorganic Medicine. New York: Marcel Dekker, 1995; 2:1318–1321.


262 Harrison<br />

15. Rosenstein ED, Caldwell JR. Trace elements in the treatment of rheumatic<br />

conditions. Rheum Dis Clin North Am 1999; 25:929.<br />

16. Thunus L, Dauphin JF, Moiny G, et al. Anti-inflammatory properties of copper,<br />

gold and silver, individually and as mixtures. Analyst 1995; 120:967.<br />

17. Ackerknecht EH. A Short History of Medicine. Rev. ed. Chap. 1. Baltimore:<br />

Johns Hopkins University Press, 1982.<br />

18. Majno G. The Healing Hand: Man and Wound in the Ancient World.<br />

Cambridge, MA.: Harvard University Press, 1975.<br />

19. Dollwet HHA, Sorenson JRJ. Historic uses of copper compounds in medicine.<br />

Trace Elem Med 1985; 2:80.<br />

20. Dresher WH. Copper in my medicine chest. Innovations: the online magazine of<br />

the Copper Development Association, June 2000. (http://www.copper.org/<br />

innovations/2000/06/medicine-chest.html).<br />

21. An onlooker’s notebook. Copper on the up and up? Pharm J 1974; Oct 5:318.<br />

22. Sorenson JRJ. Therapeutic uses of copper. In: Nriagu JO, ed. Copper in the<br />

Environment, Part II: Health Effects. New York: John Wiley & Sons, 1979:83–162.<br />

23. Whitehouse MW. Ambivalent role of copper in inflammatory disorders. Agents<br />

Actions 1976; 6:201.<br />

24. Sharples NM. English Heritage Book of Maiden Castle. London: B.T. Batsford,<br />

1991.<br />

25. Loucks G. The girl and the bear facts: a cross-cultural comparison. Can J Native<br />

Stud 1985; 5:218.<br />

26. Weissmann G. A fashion in metals. Hosp Pract (Hosp Ed) 1982; 17:33.<br />

27. Strange CJ. Coping with arthritis in its many forms. FDA Consumer, March 1996.<br />

(http://www.fda.gov/fdac/features/296_art.html).<br />

28. Hostynek JJ, Maibach HI. Copper hypersensitivity: dermatologic aspects—an<br />

overview. Rev Environ Health 2003; 18:153.<br />

29. Livingston JD. Magnetic therapy: plausible attraction. Skeptical Inquirer Magazine<br />

(online), July 1998. (http://www.csicop.org/si/9807/magnet.html).<br />

30. Österberg R, Sjöberg B, Branega˚rd B. A critical review of copper in medicine.<br />

Final report. INCRA Project No. 234. New York: International Copper<br />

Research Association, 1975:44.<br />

31. Shorrocks VM, Alloway BJ. Copper in plant, animal and human nutrition.<br />

Potters Bar, UK: Copper Development Association, 1985:75.<br />

32. Landner L, Lindeström L. Copper in society and in the environment: an account<br />

of the facts on fluxes, amounts and effects of copper in Sweden. 2nd rev. ed.<br />

Translated by Lars Landner. Västera˚s, Sweden: Swedish Environmental<br />

Research Group (MFG), 1999:292.<br />

33. Gaby AR. Alternative treatments for rheumatoid arthritis. Altern Med Rev<br />

1999; 4:392.<br />

34. Resman-Targoff BH. Alternative therapeutic approaches to the treatment of<br />

arthritis. J Pharm Prac 1999; 12:346.<br />

35. Walker WR, Keats DM. An investigation of the therapeutic value of the ‘copper<br />

bracelet’—dermal assimilation of copper in arthritic/rheumatoid conditions.<br />

Agents Actions 1976; 6:454.<br />

36. Walker WR, Beveridge SJ, Whitehouse MW. Dermal copper drugs: the copper<br />

bracelet and Cu(II) salicylate complexes. Agents Actions Suppl 1981; 8:359.


Copper Jewelry and Arthritis 263<br />

37. Walker WR. The results of a copper bracelet clinical trial and subsequent studies.<br />

In: Sorenson JRJ, ed. Inflammatory Diseases and Copper: The Metabolic<br />

and Therapeutic Roles of Copper and Other Essential Metalloelements in<br />

Humans. Clifton, NJ: Humana, 1982:469–478.<br />

38. Haynes B. Can it work? Does it work? Is it worth it? The testing of health care<br />

interventions is evolving. Br Med J 1999; 319:652.<br />

39. Fernández-Madrid F. Treating Arthritis: Medicine, Myth, and Magic. New<br />

York: Plenum, 1989:108.<br />

40. Walker WR, Griffin BJ. The solubility of copper in human sweat. Search 1976; 7:100.<br />

41. Walker Wr, Reeves RR, Brosnan M, et al. Perfusion of intact skin by a saline<br />

solution of bis(glycinato) copper(II). Bioinorg Chem 1977; 7:271.<br />

42. Caldwell JR. Venoms, copper, and zinc in the treatment of arthritis. Rheum Dis<br />

Clin North Am 1999; 25:919.<br />

43. HealthWatch. Folklore arthritis remedies: copper bracelets. HealthWatch Newsletter<br />

(online), 4, April 1990. (http://www.healthwatch-uk.org/nlett04.html).<br />

44. Turner JA, Deyo RA, Loeser JD, et al. The importance of placebo effects in pain<br />

treatment and research. JAMA 1994; 271:1609.<br />

45. Jadad AR, Moore RA, Carroll D, et al. Assessing the quality of reports of randomized<br />

clinical trials: is blinding necessary? Controlled Clin Trials 1996; 17:1.<br />

46. Moher D, Sampson M, Campbell K, et al. Assessing the quality of reports of<br />

randomized trials in pediatric complementary and alternative medicine. BMC<br />

Pediatr 2002; 2:2.<br />

47. The HealthWatch Committee. The placebo effect: let’s unwrap it and then tap it.<br />

HealthWatch Newsletter (online), 13, Autumn 1993. (http://www.healthwatch-uk.<br />

org/nlett13.html).<br />

48. Kaptchuk TJ. The placebo effect in alternative medicine: can the performance of<br />

a healing ritual have clinical significance? Ann Intern Med 2002; 136:817.<br />

49. Ralph A, McArdle H. Copper metabolism and requirements in the pregnant<br />

mother, her fetus and children: a critical review. Chap. 2. New York: International<br />

Copper Association, 2001.<br />

50. Whitehouse MW, Walker WR. The ‘‘copper bracelet’’ for arthritis. Med J Aust<br />

1977; 1:938.<br />

51. Walker WR, Whitehouse MW. Facilitated transdermal absorption of copper (II)<br />

suppresses experimental inflammation. Aust N Z J Med 1979; 9:112.<br />

52. Shackel NA, Day RO, Kellett B, et al. Copper-salicylate gel for pain relief in<br />

osteoarthritis: a randomised controlled trial. Med J Aust 1997; 167:134.<br />

53. Boettcher B. Copper-salicylate gel for pain relief in osteoarthritis [letter]. Med J<br />

Aust 1998; 168:312.<br />

54. Brooks PM, Kellett B. Reply to letter by B. Boettcher. Med J Aust 1998; 168:312.<br />

55. Lin J, Zhang W, Jones A, et al. Efficacy of topical non-steroidal anti-inflammatory<br />

drugs in the treatment of osteoarthritis: meta-analysis of randomised controlled<br />

trials. Br Med J 2004; 329:324.<br />

56. Mason L, Moore Ra, Edwards JE, et al. Topical NSAIDs for acute pain: a metaanalysis.<br />

BMC Fam Pract 2004; 5:10.<br />

57. Bratton RL, Montero DP, Adams KS, et al. Effect of ‘‘ionized’’ wrist bracelets<br />

on musculoskeletal pain: a randomized, double-blind, placebo-controlled trial.<br />

Mayo Clin Proc 2002; 77:1164.


264 Harrison<br />

58. Federal Trade Commission (USA). Marketers of Q-Ray Ionized Bracelet<br />

charged by FTC: FTC seeks to halt deceptive pain relief claims and provide consumer<br />

refunds. Press release, June 2, 2003.<br />

59. Mulherin DM, Struthers GR, Situnayake RD. Do gold rings protect against<br />

articular erosion in rheumatoid arthritis? Ann Rheum Dis 1997; 56:497.<br />

60. Rao JK, Mihaliak K, Kroenke K, et al. Use of complementary therapies for<br />

arthritis among patients of rheumatologists. Ann Intern Med 1999; 131:409.<br />

61. Marcus DM. Alternative medicine and the Arthritis Foundation [editorial].<br />

Arthritis Rheum 2002; 47:5.<br />

62. Suarez-Almazor ME, Kendall CJ, Dorgan M. Surfing the Net—information on<br />

the World Wide Web for persons with arthritis: patient empowerment or patient<br />

deceit? J Rheumatol 2001; 28:185.<br />

63. Camara K, Danao-Camara T. Awareness of, use and perception of efficacy of<br />

alternative therapies by patients with inflammatory arthropathies. Hawaii Med<br />

J 1999; 58:329.<br />

64. Kestin M, et al. The use of unproven remedies for rheumatoid arthritis in<br />

Australia. Med J Aust 1985; 143:516.<br />

65. Southwood TR, Malleson PN, Roberts-Thomson PJ, et al. Unconventional<br />

remedies used for patients with juvenile arthritis. Pediatrics 1990; 85:150.<br />

66. Buchbinder R, Gingold M, Hall S, et al. Non-prescription complementary treatments<br />

used by rheumatoid arthritis patients attending a community-based rheumatology<br />

practice. Intern Med J 2002; 32:208.<br />

67. Hagen LEM, Schneider R, Stephens D, et al. Use of complementary and alternative<br />

medicine by pediatric rheumatology patients. Arthritis Rheum 2003; 49:3.<br />

68. Struthers GR, Scott DL, Scott DGI. The use of ‘alternative treatments’ by<br />

patients with rheumatoid arthritis. Rheumatol Int 1983; 3:151.<br />

69. Seymour A, Mayes C, Young A, et al. The use of complimentary and alternative<br />

medicine in patients with rheumatoid arthritis. Abstr. 221. British Society for<br />

Rheumatology XIX Annual General Meeting, Brighton, United Kingdom, April<br />

23–26, 2002. Rheumatology 2002; 41(suppl 2):88.<br />

70. Price JH, Hillman KS, Toral ME, et al. The public’s perceptions and misperceptions<br />

of arthritis. Arthritis Rheum 1983; 26:1023.<br />

71. Keysor JJ, Currey SS, Callahan LF. Behavioral aspects of arthritis and rheumatic<br />

disease self-management. Dis Manage Health Outcomes 2001; 9:89.<br />

72. Barnes J. Quality, efficacy and safety of complementary medicines: fashions, facts<br />

and the future. Part I. Regulation and quality. Br J Clin Pharmacol 2003; 55:226.<br />

73. Herman CJ, Allen P, Hunt WC, et al. Use of complementary therapies among<br />

primary care clinic patients with arthritis. Prev Chronic Dis 2004; 1:A12.<br />

74. Arthritis Foundation, Association of State and Territorial Health Officials and<br />

CDC. National Arthritis Action Plan: a public health strategy. Atlanta: Arthritis<br />

Foundation, 1999. (http://www.cdc.gov/nccdphp/pdf/naap.pdf).<br />

75. Rao JK, Kroenke K, Mihaliak KA, et al. Rheumatology patients’ use of complementary<br />

therapies: results from a one-year longitudinal study. Arthritis<br />

Rheum 2003; 49:619.<br />

76. Neville CE, Hassan W. Complimentary therapy use in different ethnic groups<br />

with rheumatoid arthritis. Abstr. 237. British Society for Rheumatology XX<br />

Annual General Meeting. A joint meeting with La Société Française de Rhumatologie.<br />

Manchester, United Kingdom, April 1–4, 2003. Rheumatology 2003;<br />

42(suppl 1):94.


Copper Jewelry and Arthritis 265<br />

77. Horstman J. Why doctors aren’t asking and you aren’t telling. Arthritis Today,<br />

November–December 1999. (http://www.arthritis.org/resources/arthritistoday/<br />

1999_archives/1999_11_12alternatives.asp).<br />

78. IPCS (International Programme on Chemical Safety). Environmental Health<br />

Criteria 200. Copper. Geneva: World Health Organization, 1998:71.<br />

79. Duffy EM, Meenagh GK, McMillan SA, et al. The clinical effect of dietary supplementation<br />

with omega-3 fish oils and/or copper in systemic lupus erythematosus.<br />

J Rheumatol 2004; 31:1551.<br />

80. Brooks PM, Hart JAL. The bone and joint decade: 2000–2010. Med J Aust 2000;<br />

172:307.<br />

81. Gardner M. Fads and Fallacies in the Name of Science. 2nd rev. ed. New York:<br />

Dover, 1957:204.<br />

82. Straus SE, Beldon W. Fiscal year 2006 budget request, statement to the House<br />

Subcommittee on Labor-HHS-Education Appropriations. Bethesda, MD: National<br />

Center for Complementary and Alternative Medicine, NIH, March 9, 2005.<br />

(http://www.hhs.gov/budget/06budget/documents/NCCAM.doc).<br />

83. Sampson WI. Why the National Center for Complementary and Alternative<br />

Medicine (NCCAM) should be defunded Quackwatch. (http://www.quackwatch.org/01QuackeryRelatedTopics/nccam.html).<br />

84. Kolasinski SL. Complementary and alternative therapies for rheumatic disease.<br />

Hosp Pract 2001; 36:31.<br />

85. Horstman J. The Arthritis Foundation’s Guide to Alternative Therapies. Atlanta:<br />

Longstreet Press, 1999:151.<br />

86. The Arthritis Society (Canada). Living well with arthritis: unproven remedies;<br />

potions, elixirs, gadgets and devices. Toronto: The Arthritis Society (Canada),<br />

1999.<br />

87. Arthritis Foundation of Victoria (Australia). What can be done? Elsternwick,<br />

Australia: Arthritis Foundation of Victoria (Australia), 1998.<br />

88. National Institute of Arthritis and Musculoskeletal and Skin Diseases. Handout on<br />

health: osteoarthritis. National Institute of Arthritis and Musculoskeletal and Skin<br />

Diseases, National Institute of Health (USA), 2002.<br />

89. Federal Trade Commission (USA), ‘Miracle’ health claims: add a dose of skepticism.<br />

Federal Trade Commission (USA), produced in cooperation with the Food<br />

and Drug Association (USA), 2001. (http://www.ftc.gov/bcp/conline/pubs/<br />

health/frdheal.htm).<br />

90. Better Business Bureau. Health quackery. Natick, MA: Better Business Bureau<br />

of Eastern Massachusetts, Maine and Vermont, 1996, (http://www.bosbb.org/<br />

literature/lit_0011.asp).<br />

91. Biasi G, Badii F, Magaldi M, et al. Un nuovo approccio al trattamento della<br />

syndrome fibromialgica: L’uso del Telo Cypro 1 . [A new approach in the treatment<br />

of fibromyalgia: The use of a copper wire sheet (‘‘Telo Cypro 1 ’’)]. Minerva<br />

Med 1999; 90:39.<br />

92. Federal Trade Commission (USA). Remedies targeted in International Health<br />

Claim Surf Day; consumer protection, public health, privated agencies from<br />

24 countries assess over 1,200 Internet sites. Washington, DC: Federal Trade<br />

Commission press release. November 10, 1998. (http://www.ftc.gov/opa/<br />

1998/11/intlhlth.htm).


11<br />

Role of Copper in Anti-inflammatory<br />

Therapy and the Potential for Its<br />

Transdermal Application<br />

Jurij J. Hosty´nek<br />

Department of Dermatology, University of California at San Francisco<br />

School of Medicine, San Francisco, California, U.S.A.<br />

Roberto Milanino<br />

Facoltà di Medicina e Chirurgia, Sezione di Farmacologia,<br />

Dipartimento di Medicina e Salute Pubblica, Università di Verona,<br />

Verona, Italy<br />

INTRODUCTION<br />

Copper is an essential trace element (ETE), critical for a variety of biological<br />

processes, for example hemoglobin synthesis, enzyme activation, particularly<br />

that of superoxide dismutase, and, more generally, as a key component of<br />

mitochondrial, cytoplasmic, and nuclear enzyme systems. Absorbed mainly<br />

from dietary sources (organ meats, shellfish, nuts, whole-grain cereals),<br />

copper absorption and utilization is interdependent with that of other metals,<br />

including zinc, iron, cadmium, and molybdenum. Significant stores of copper<br />

are found in liver, muscle, and bone. It is required for bone formation,<br />

cardiac function, keratinization, and tissue pigmentation, where it binds to<br />

proteins, preferentially sulfur- and nitrogen-containing ligands. In mammals,<br />

ceruloplasmin (CP), a multifunctional endogenous plasma glycoprotein, is<br />

responsible for copper transport in the blood where it may bind some 60%<br />

to 70% of the metal present. Physiological factors that require involvement<br />

267


268 Hosty´nek and Milanino<br />

of copper as a remedial factor induce CP production in the liver, thus<br />

assuring an adequate transport medium in response to increased translocation<br />

requirements (1). Total plasma copper is a reliable indicator of<br />

circulating CP, as a highly significant correlation exists between the two<br />

values (r ¼ 0.8), in rats as well as humans (2,3).<br />

Copper plays an active role in inflammation, and animal models show<br />

that the anti-inflammatory activity of the metal is amplified when complexed<br />

with nonsteroidal anti-inflammatory drugs (NSAIDs). As a rule, such complexes<br />

also become more effective and less irritant than the parent ligands (4–9).<br />

It has been hypothesized that supplementation with exogenous copper<br />

may bring remedial action in inflammatory disease such as arthritis (A), and<br />

it was widely demonstrated that intravenous (IV) and subcutaneous (SC)<br />

administration of copper solutions is effective in controlling edema induced<br />

in a variety of animal screens (6,10–12).<br />

The inflammatory condition comprises a wide spectrum of diseases that<br />

include rheumatoid arthritis (RA), childhood arthritis, carpal tunnel syndrome,<br />

osteoarthritis, lupus, and gout—conditions associated with pain,<br />

stiffness, and swelling in and around the joints. It affects one out of every<br />

three adults and is the most common cause of disability in the United States.<br />

It results in 75,000 hospitalizations and 39 million outpatient visits annually<br />

(13). The majority of those affected by inflammatory diseases, of all<br />

racial and ethnic groups, are less than 65 years of age, as onset is most often<br />

seen between the ages of 25 and 50 years. RA and osteoarthritis, the most<br />

common forms of A, are characterized by chronic joint inflammation eroding<br />

cartilage and bone. Some forms of A, such as RA, psoriatic arthritis, and<br />

lupus, can affect multiple organs and cause widespread symptoms. RA is<br />

characterized by nonspecific, usually symmetric inflammation of the peripheral<br />

joints, and may lead to destruction of articular and periarticular<br />

structures. It is a chronic syndrome generally classified as an autoimmune<br />

disease, and a genetic predisposition has been identified. Control of the condition<br />

in humans is achieved through use of immunosuppressive drugs, and<br />

symptoms can be reduced by treatment with anti-inflammatory compounds.<br />

The majority of those afflicted improve symptomatically with conservative<br />

treatment in the early stages of the disease, but many are severely disabled<br />

over the course of their lives, as a cure is not known (14,15).<br />

TRADITIONAL AND MODERN THERAPIES FOR RA AND<br />

RELATED DISORDERS<br />

Copper Metal—Bracelets and Rings<br />

Historically, use of copper in the form of the metal, as verdigris (basic cupric<br />

acetate) or vitriol (cupric sulfate), has been a time-honored practice since<br />

antiquity, recognized for its anti-inflammatory activity (16). The potential


Copper in Anti-inflammatory Therapy 269<br />

for its activity as an anti-inflammatory agent by transdermal delivery,<br />

however, remains subject to controversy because scientific studies designed<br />

to demonstrate therapeutic benefits for arthritic conditions through dermal<br />

contact with metallic copper or its compounds have been inadequate. Quantitative<br />

data for percutaneous penetration of the metal’s putative oxidation<br />

products, which may be generated in contact with skin in humans, are not<br />

available. Because no beneficial effect could be statistically documented<br />

for skin contact with copper devices, their use was and still is relegated to<br />

‘‘quack’’ medicine. It is unfortunate that this term has been used when<br />

the term ‘‘not understood’’ would have been more appropriate.<br />

Persisting lay use of copper devices to the present day and assertions of<br />

beneficial effects obtained seem to warrant an independent study of the<br />

metal to objectively evaluate its effectiveness, separating activity from placebo<br />

effect, even if the current state of anti-inflammatory medicine casts a<br />

doubt over the economic value of such studies. A first step in reinstating<br />

the metal in the armamentarium of anti-inflammatory remedies would be<br />

a quantitative, controlled study of copper release from the metal brought<br />

in intimate contact with human skin in vivo.<br />

Release of Copper in Contact with the Skin<br />

In order to confirm the long-standing assertions that skin contact with copper<br />

bangles has an AI effect due to skin penetration of solubilized metal,<br />

Walker and Keats performed experiments to verify that copper metal was<br />

oxidized and dissolves in artificial sweat. In samples where initial copper<br />

concentration was of the order of 2 10 5 M, after 24 hours the samples<br />

turned blue and the concentration had increased to 2 10 3 M Cu. The<br />

same authors also measured the weight loss from copper bracelets due to<br />

corrosion worn by volunteers. Bracelets measuring 22 1.3 0.1 cm lost<br />

an average of 13 mg per month (17).<br />

Weight losses of 0.1% to 0.8% were observed again in copper bracelets<br />

weighing 14 g used in a trial involving volunteers and lasting one month (18).<br />

Such variation can be explained by differential mechanical wear. An alternative<br />

explanation is the variability in subjective sweat composition and sweating rate.<br />

Skin Penetration by Copper Compounds<br />

The only experiment that allows calculation of a diffusion constant Kp for<br />

a copper compound was performed by Walker. Bis(Glycinato) copper (II)<br />

complex, a copper compound likely to be formed with skin exudates, was<br />

applied to cat skin in vitro (19). A radioactive ( 64 Cu) 0.05 M solution in<br />

physiological saline applied to excised cat skin resulted in a steady-state<br />

transport rate Kp ¼ 24 10 4 cm/hr. All other diffusivity data for copper<br />

compounds had to be deduced from clinical observations or physical analytical<br />

methods on animals and humans following exposure.


270 Hosty´nek and Milanino<br />

In vivo application of copper oleate in a lanolin-petroleum base over<br />

24 hours to human back skin resulted in a significant increase in urinary copper<br />

levels over several days (20).<br />

Following topical application of lipophilic Cu(II) salicylate and -phenylbutazone<br />

complexes, designed to study their penetration and biodistribution<br />

in rats, 64 Cu was eliminated almost quantitatively, primarily in feces, an indication<br />

that Cu(II) rapidly permeates the dermal barrier (21).<br />

Electron microscopy of skin treated topically with copper acetate led<br />

to the observation that copper initially localizes in the intercellular spaces;<br />

subsequently, the cell membranes of viable cells are penetrated as well, and<br />

the metal accumulates in and around the cell nucleus (22).<br />

By electron probe analysis and analytical electron microscopy, occurrence<br />

and levels of copper were traced across human cadaver skin following<br />

application of CuSO4 using Franz cells. Copper was observed to enter the<br />

outer sc cells at high concentrations; it then encounters an apparent barrier<br />

approximately halfway within the epidermis, falling below the detection limit<br />

(0.1% by weight), to reappear at the granular interface. There, it was primarily<br />

seen traversing an intercellular route through the granular layer, to reach<br />

the spinosum layer (Warner, personal communication).<br />

Scope and Limitations of the Quantitative Structure Activity<br />

Relationships Approach to Diffusivity of Metal Compounds<br />

Principles that govern diffusion of exogenous compounds through biological<br />

membranes such as the skin are prejudicial towards penetration by metal<br />

complexes in general, governed as they are by numerous factors difficult to<br />

evaluate a priori.<br />

Mathematically derived (mechanistic) models predictive of percutaneous<br />

penetration have been developed based on in vitro and in vivo data<br />

through regression analysis of experimental results, and serve to predict diffusivity<br />

of untested structures. Within limits of molecular parameters such<br />

as size, lipid solubility, or hydrogen bonding, these models apply to organic<br />

compounds such as drugs, pesticides, cosmetic ingredients, etc., and can predict<br />

their dermal absorption with increasing accuracy (23–26). The elements<br />

required for construction of such predictive models can be reduced to the<br />

criteria of structure and physical properties (27). No such reduction is feasible<br />

when modeling diffusion of electrolytes, however. Attempts to model<br />

the dermal absorption process of metal compounds, and those of transition<br />

metals in particular, have been thwarted owing to a number of confounding<br />

factors, as became evident from in vivo and in vitro experiments. Because<br />

movement through biological membranes is highly element and chemical<br />

species specific, molecular physicochemical parameters alone do not suffice<br />

to model their migration into and through the strata of the skin. A number<br />

of factors are closely inter-related and their combined effects are neither<br />

entirely understood nor predictable. Unless the dynamics of in situ changes


Copper in Anti-inflammatory Therapy 271<br />

in valence (oxidation state), or electrophilic reactivity, among others, can be<br />

factored in, the diffusivity of metallic elements eludes modeling. Furthermore,<br />

the limited experimental data available so far from in vitro and in vivo<br />

experimentation for metal compounds have been acquired under disparate<br />

conditions and are too scarce considering the number of metals and<br />

metalloids of variable valence existing as free ions or forming chelates, coordination<br />

compounds, or complexes with electron donors such as oxygen,<br />

sulfur, or phosphorus, abundant in biological systems. The resulting database<br />

is too limited for the development of predictive algorithms. Computer<br />

calculations have been used to predict skin diffusivity for chemical structures,<br />

and of conventional AI agents in particular (28–30), but not for cupriphores.<br />

DRUG THERAPY<br />

Aspirin, Salicylates, and Other Anti-inflammatory Drugs<br />

Aspirin, first used as an antipyretic, has also been recognized for its analgesic<br />

activity, and it was elaborated in a plethora of derivatives (salicylates). Relatively<br />

safe, these derivatives, as well as aspirin itself, continue in use as the<br />

cornerstone of AI drug therapy to the present day. Introduced to replace cortisone,<br />

an AI hormonal agent first isolated in the 1930s, they act on the swelling,<br />

heat, and pain of inflammation that is suffered by A victims. They are relatively<br />

safe and inexpensive. Given PO they can induce GI symptoms affecting the<br />

gastric mucosa, however, and erosion and bleeding gastric ulcers may result.<br />

Besides derivatives of salicylic acid, NSAIDs from other chemical classes<br />

have been evaluated for their anti-inflammatory activity: N-arylanthranilic<br />

acids, e.g., mefenamic acid, derivatives of arylacetic acid, e.g., ibuprofen,<br />

diclofenac, piroxicam, aniline and p-aminophenol derivatives, acetaminophen,<br />

etc. (31). Given PO, their dosage is still limited, however, as they also carry<br />

the risk of GI symptoms.<br />

Recent anti-inflammatories are selective NSAIDs, such as the family of<br />

COX-2 cyclo-oxygenase inhibitors Vioxx (rofecoxib), Bextra (valdecoxib),<br />

or Celebrex (celecoxib). In contrast to aspirin, however, they do not impair<br />

platelet adhesiveness, which may lead to cardiovascular disorders, and in<br />

addition are suspected to induce psoriasis (32).<br />

Gold<br />

Systemic treatment with soluble gold (I) salts (chrysotherapy) represents a<br />

continuum in the armamentarium of rheumatologists since the beginning<br />

of drug therapy to combat arthritis, as it affords rapid symptomatic relief.<br />

The different aspects of gold therapy, positive and negative, are therefore<br />

reviewed here in greater depth. Initially used for the treatment of tuberculosis<br />

until the 1930s, promoted by Forestier chrysotherapy found its principal therapeutic<br />

application in the treatment of RA (33). Use continues in addition to


272 Hosty´nek and Milanino<br />

salicylates or other NSAIDs if the latter do not bring sufficient relief or<br />

suppress active joint inflammation. A serious limiting factor in systemic application<br />

of soluble gold salts has been the high incidence of adverse reactions,<br />

as some degree of toxicity appears inevitable at effective dose levels. The most<br />

common toxic effects involve the skin and mucous membranes, including<br />

pruritus, allergic contact dermatitis (ACD), and stomatitis, but gold compounds<br />

can also be toxic to the hematopoietic organs and the kidney. Gold<br />

therapy must be discontinued when any of those manifestations appear,<br />

which occurs in up to one-third of patients (34,35).<br />

Parenteral preparations include gold sodium thiomalate, gold thioglucose,<br />

or gold thiosulfate, and are administered IM at weekly intervals: 10 mg<br />

the first week, 25 mg the second, and 50 mg/wk thereafter until a total of 1 g<br />

has been given or significant improvement is apparent. When maximum<br />

improvement is achieved, dosage is decreased to 50 mg every 2–4 weeks (14).<br />

In active RA, chrysotherapy provides rapid pain relief, and can induce partial<br />

or complete remission in as high as 80% of cases, as long as therapeutic regime<br />

is maintained. Even with sustained therapy, however, there is roentgenological<br />

evidence of progression of the disease. Eventually, all patients relapse in 3–6<br />

months when therapy is discontinued; the condition then returns to the pretherapeutic<br />

status and the disease continues on its natural course (36).<br />

Despite the high incidence of serious adverse effects, chrysotherapy is<br />

still widely accepted, since for many forms of arthritis, given parenterally,<br />

orally, or more recently, intra-articularly, gold (I) salts still represent the<br />

most effective therapy (37).<br />

Thanks to its anti-inflammatory properties, over the past half century<br />

gold has found a number of applications, such as treatment of juvenile and<br />

adult rheumatoid arthritis via intramuscular injection of the water-soluble<br />

thiomalate, thioglucose, or thiosulfate (38). Other new uses include the treatment<br />

of such skin diseases as pemphigus and psoriatic arthritis, which<br />

involve an immunological component (39). Such use is no longer practiced<br />

in the United States. Following parenteral therapy, which involves relatively<br />

high doses, gold was seen to persist in the organism for several months.<br />

Carried by plasma, bound to alpha-globulin, it is carried to practically every<br />

tissue, including the skin where it becomes evident in characteristic blue-gray<br />

discoloration, or chrysiasis, most often appearing on the face and hands,<br />

areas exposed to sunlight, hair, and nails (36,38,40).<br />

While for nearly three-quarters of a century gold salts have been used to<br />

relieve the symptoms of arthritis, just how the metal exerts its antiarthritic<br />

effect is not clear, as it does not have a known biological function itself.<br />

Stoyanov and Brown (41) propose an indirect role by gold since they find that<br />

it specifically binds to a copper-activating protein, thus turning on production<br />

of a protein pump involved in copper transport, mobilizing endogenous<br />

copper reserves. The biological effect of gold antiarthritic drugs may thereby<br />

consist in their effects on copper management in eukaryotic systems.


Copper in Anti-inflammatory Therapy 273<br />

Immune Reactions in Chrysotherapy<br />

Due to increasing systemic exposure of patients for therapeutic purposes<br />

over the past 50 years, gold is now recognized as a significant factor in the<br />

etiology of cellular and humoral immunity (39,42). It is also an inducer of<br />

circulating immune complexes (43,44).<br />

Systemically, gold compounds have modulatory effects on immediate<br />

and delayed immune responses, both in vitro and in vivo (42), causing type I<br />

hypersensitivity (contact urticaria syndrome), cell-mediated (type IV) sensitization<br />

or ACD, as well as type III or Arthus reactions involving antigen–<br />

antibody complexes. Hypersensitivity reactions to both the monovalent<br />

and trivalent forms of the metal have been described. While ACD is not<br />

uncommon due to the widely practiced systemic therapy using gold<br />

compounds, or the intimate skin contact with metallic gold, urticarial reactions<br />

are extremely rare. The relatively high incidence of delayed skin<br />

reactions has been noted since the introduction of gold compounds for<br />

the treatment of rheumatoid arthritis, and is seen in more than 50% of<br />

patients so treated (45,46). Although not clinically relevant to the most part,<br />

the rate of sensitization to gold seems to be on a steady increase, with comparable<br />

prevalence in the United States, Europe, and Japan since routine<br />

screening for gold allergy was started in those countries (47–51). Injection<br />

of water-soluble gold complexes is associated with a high risk of sensitization<br />

in the form of a type I hypersensitivity response (52). Repeated<br />

exposure to gold salts in sensitized persons does not necessarily lead to<br />

recurrence of dermatitis, however. This indicates a gradual buildup of tolerance<br />

(53). Abnormally high serum IgE levels together with eosinophilia have<br />

been associated with gold therapy, but remit on cessation of such therapy<br />

(54,55). Nephrotic syndrome, and glomerulonephritis in particular, is noted<br />

as a consequence of parenteral or oral administration of gold in its organic<br />

form. Certain gold compounds, such as Ridaura (auranofin), given orally in<br />

which gold is complexed to trialkylphosphines, are suspected of giving rise<br />

to circulating immune complexes (type III hypersensitivity) (42). As an electrophile<br />

reacting with native protein, the metal forms a complete antigen<br />

that induces specific antibody formation, which, in turn, gives rise to the glomerular<br />

injury observed (43,44,56,57). In patients treated parenterally with a<br />

variety of gold compounds, the metal is seen to be retained in significant<br />

amounts (over 80% of the amount injected over 39 days of administration)<br />

bound to the plasma component alpha-globulin as a gold protein complex.<br />

The amounts excreted (primarily in urine) were not related quantitatively to<br />

the amounts injected (38), and retention may increase the risk of immune<br />

complex glomerulonephritis in patients receiving gold preparations.<br />

In a large Swedish study involving 823 <strong>dermatology</strong> patients, the incidence<br />

of 8.6% positive reactions to gold was recorded, without any signs of<br />

an irritant etiology (58). Recent confirmation of this result now makes gold


274 Hosty´nek and Milanino<br />

the second most common contact allergen (in the Swedish population) after<br />

nickel. Thus, routine patch testing of dermatitis patients in Europe has<br />

revealed a rate of positive readings approximating 10% and 14.3% in North<br />

America (59,60). Based on test results by the North American Contact<br />

Dermatitis Group, with 9.5% positive, gold is the sixth most common<br />

allergen among <strong>dermatology</strong> patients in the United States (61). A similar<br />

prevalence is noted in Europe and Japan (50,51).<br />

Systemically provoked flare-ups of contact allergy to gold appear as<br />

another characteristic of response to that allergen. Intramuscular injection<br />

of gold sodium thiomalate was reported to reactivate previously involved<br />

but clinically healed skin. It also led to corresponding pathobiochemical<br />

changes in the venous blood of the patients (i.e., activation of neutrophil<br />

granulocytes and release of several cytokines). Similarly, intradermal test<br />

sites that had healed 9 to 24 months previously were reactivated (62–64).<br />

The diverse skin reactions do not appear to depend on concentrations<br />

of the metal in the skin. Patients receiving cumulative doses of 1 to 50 g of<br />

gold (I) salts may not experience adverse reactions (65), and there is no consensus<br />

on optimal or limit in dosage. Thus, tolerance for gold salts is highly<br />

individual, and patients require constant monitoring for toxic manifestations.<br />

In summary, while contact of intact skin with metallic gold is not<br />

known to induce hypersensitivity, its occurrence in context with systemic<br />

exposure through injection of soluble salts for therapeutic purposes is<br />

now considered to be an unavoidable side effect in the risk–benefit balance<br />

of anti-inflammatory therapy.<br />

Corticosteroids<br />

Corticosteroids are effective oral and intra-articular anti-inflammatories, but<br />

their efficacy diminishes with time and they do not prevent joint destruction.<br />

Their use is also associated with various side effects, and steroid therapy is<br />

only recommended on failure of less hazardous drugs (14).<br />

Superoxide Dismutase<br />

Anti-inflammatory copper-dependent metalloenzymes, superoxide dismutase,<br />

have been developed for the treatment of arthritic diseases (66). Two<br />

such preparations, Ontosein and Orgotein, have been shown to be safe<br />

and effective for the treatment of established rheumatoid and osteoarthritis<br />

when given IV or IA into knee and hip joints in single or multiple doses.<br />

They cannot be dosed PO, however, due to gastric inactivation.<br />

Immunosuppressants<br />

Methotrexate, cyclosporine, and others are used for severe RA, but bring<br />

the risk of major side effects: liver disease, bone marrow suppression, and<br />

potentially, malignancy (14).


Copper in Anti-inflammatory Therapy 275<br />

The search for new anti-inflammatory agents has had limited success<br />

so far, owing to the lack of insight into the causes and mechanisms of<br />

inflammatory diseases.<br />

Physiotherapy<br />

Other than surgical intervention, relief from arthritic pain was sought in a<br />

number of noninvasive approaches: ultrasound, thermotherapy, balneotherapy,<br />

electrotherapy, laser therapy, acupuncture, and even tai chi.<br />

PRECEDENTS IN TOPICAL DELIVERY OF<br />

ANTI-INFLAMMATORY AGENTS<br />

Topical anti-inflammatory formulations have a history of successful application.<br />

An early precedent for topical treatment of rheumatic discomfort were<br />

thermogenic ointments based on methyl salicylate (wintergreen) and menthol,<br />

at present marketed as Ben-Gay by Pfizer. Also some of the drugs mentioned<br />

above, such as NSAIDs, corticosteroids, immunosuppressives, and also<br />

dimethylsulfoxide and capsaicin were reported to be clinically effective in<br />

placebo-controlled trials (67). The argument for developing copper<br />

complexes with proven analgesics can be made invoking the findings by<br />

Sorenson and others, cited elsewhere, that copper–ligand chelates show<br />

anti-inflammatory activity superior to that of the ligands themselves.<br />

ROLE OF COPPER IN AI ACTIVITY<br />

Knowledge sent down from early historic times has been combined with<br />

modern pharmacology. Recognition of the essential and therapeutic role<br />

that endogenous copper plays in the control of inflammatory processes<br />

led to the concept of introducing the ETE into state-of-the-art pharmacology,<br />

thus combining the two elements. A hypothesis first: the idea of using<br />

exogenous copper in combination with analgesics and anti-inflammatories<br />

may turn out to be a successful concept with augmenting effects in modulation<br />

and control of inflammatory processes.<br />

Inflammation is tissues’ response to injury and represents the first step<br />

of the repair process towards normalization. While many anti-inflammatory<br />

agents may reduce some aspects of inflammation, they do not necessarily<br />

involve repair processes towards correcting the cause of the disease. Copper<br />

complexes, on the other hand, were demonstrated to promote tissue repair<br />

processes, and they involve activity in a number of diseases with inflammatory<br />

components other than arthritis. Their therapeutic use includes infection,<br />

cancer, diabetes, and epilepsy (7,8). Copper as an ETE is present in the<br />

mammalian organism only in minimal concentration as free ionic copper<br />

(10 18 mol, with a range between 10 11 and 10 19 mol) and is predominantly<br />

complexed with proteins and amino acids, ligands that are subject to dynamic


276 Hosty´nek and Milanino<br />

exchange in response to physiological requirements and homeostatic equilibria<br />

(8,68). In animals and humans, copper levels change in response to<br />

inflammation.<br />

Heilmeyer and Stuwe (69) had first observed that serum copper rose<br />

with arthritic disease, and fell again on remission. Stores of copper normally<br />

immobilized are bound to metallothionein, released, and bound to CP that<br />

binds over 60% of total serum copper. CP was shown to possess remarkable<br />

anti-inflammatory characteristics (70) in vivo. Experimental induction of<br />

either acute or chronic inflammation in the rat promotes accumulation of<br />

copper in some body compartments that are known to be directly or indirectly<br />

involved in the development (and remission) of the inflammatory<br />

processes (2). Thus, inflammation appears to determine an increased<br />

demand for copper by the organism, a requirement fulfilled by adjusting<br />

the intestinal absorption without depleting the major body stores (11,71).<br />

A copper-supplemented diet exhibits an antiarthritic action, and, conversely,<br />

copper deficiency appears associated with proinflammatory effects, as<br />

observed in animals on a copper-deficient diet (3).<br />

In the 1960s and 1970s, publications reported on the anti-inflammatory<br />

activity in animal models for copper compounds in different forms: as<br />

mineral salts, as enzymes, or complexes, when given IM, intraperitoneal,<br />

SC, and, most of all IV. It was also suggested that active copper complexes<br />

were formed within the organism, as their activity in animal models of<br />

inflammation was found to be greater than either treatment with inorganic<br />

copper salts or the complexing agents alone, while the parent compounds or<br />

ligands were inactive in test models of inflammation even at larger screening<br />

doses, in coordination with copper they were found to be active (4,5). Thus,<br />

the therapeutic activity of a ligand appears to depend on its association with<br />

(endogenous or exogenous) copper. This may be attributed to the ligand acting<br />

as the carrier for the metal, or copper in the complex acting as a catalyst,<br />

interfering with the inflammatory process.<br />

As reviewed by Bonta, occasional publications reported that<br />

sodium-3(N-allylcuprothiouredo)-l-benzoate and cuprous iodide had antiinflammatory<br />

activity and that Cu(II)(salicylate)2 had fever-lowering effects<br />

in various animal models of inflammation or fever. In addition, copper<br />

complexes of antiarthritic drugs, including salicylic acid, acetylsalicylic acid,<br />

penicillamine, and several corticoids, were found to be more active than the<br />

parent drugs. A comparison of copper, gold, and silver thiomalate and<br />

thiosulfate complexes in models of inflammation revealed that the copper<br />

complexes were effective, while the gold and silver complexes were virtually<br />

inactive. As a general rule, copper complexes are less toxic and suppress the<br />

ulcerogenic effect of many of the ligand drugs themselves. Those early<br />

reports of anti-inflammatory activity of copper complexed with various<br />

ligands were based on IV or parenteral administration (72).


Copper in Anti-inflammatory Therapy 277<br />

Based on this hypothesis, the compounds investigated subsequently<br />

included coppercomplexes of well-known antiarthritic drugs, including steroidal<br />

and nonsteroidal anti-inflammatory drugs: niflumic acid, D-penicillamine,<br />

hydrocortisone, dexamethasone, dimethylsulfoxide, clopirac, ketoprofen,<br />

(þ)-naproxen, indomethacin, mefenamic acid, diclofenac, ibuprofen among<br />

a number of others. All the copper complexes of those drugs have been<br />

found to be active or more effective anti-inflammatory agents than the parent<br />

drug (8,73).<br />

Oral and local administration of Cu(I) and Cu(II) complexes in animals<br />

mostly led to equivalent responses in the edema model. Since the two<br />

forms of the metal undergo different reactions in vitro, a catalytic function<br />

is attributed to exogenous copper ion, also suggesting that both<br />

Cu(I) and Cu(II) act by the same anti-inflammatory mechanism involving<br />

a common metabolite (74). The ETE, normally absorbed as Cu(II) ion,<br />

is carried in the plasma by CP, albumin, transcuprein or low molecular<br />

weight complexes such as those with different amino acids in that oxidation<br />

state, since, as is the case for Fe(II), Cu(I) can potentially prime the<br />

harmful Fenton reaction. As in the cell interior, the metal seems to exist<br />

almost exclusively in the Cu(I) form, the necessary reduction step is performed<br />

by plasma membrane reductase, an enzyme likely to be expressed<br />

by all body cell types. The cells themselves are well equipped with copper<br />

chaperones for the control of intrinsic Cu(I) toxicity. These metallochaperone<br />

proteins escort copper ions directly to enzymes that require<br />

the metal to function, such as the antioxidant copper–zinc superoxide<br />

dismutase.<br />

In a review of anti-inflammatory activity of exogenous copper,<br />

Milanino et al. (11) also concluded that copper indeed is active as an acute<br />

anti-inflammatory agent irrespective of chemical form given, including inorganic<br />

copper salts.<br />

Administration of exogenous copper is effective in controlling inflammation<br />

associated with arthritis, and may lead to complete (albeit temporary)<br />

remission. Sorenson (8) reviewed the AI activity of several dozen copper complexes,<br />

mostly exploratory and several as commercial drugs, the categories of<br />

ligands including non-anti-inflammatory compounds such as amino acids,<br />

heterocyclic carboxylic acids and amines, among others. The AI activities<br />

reported were based on oral or parenteral administration. However, copper<br />

salts compounded with salicylic acid or ethyl salicylate have also been shown<br />

to have AI activity in animal models of inflammation following topical<br />

application (75).<br />

Another indication of the anti-inflammatory action of copper is the rise<br />

of serum copper in pregnancy, in tandem with remission of RA, which is<br />

attributed to the protective AI action due to an increase in CP levels. This<br />

is followed by a postpartum relapse to pre-existing RA (76).


278 Hosty´nek and Milanino<br />

PAST USE OF COPPER CHELATES IN THE TREATMENT<br />

OF RHEUMATOID ARTHRITIS<br />

Mixtures of copper salts and salicylic acid or ethyl salicylate in aqueous<br />

or glycerol-dimethyl sulfoxide solutions were tested successfully for antiinflammatory<br />

activity following topical application in a variety of animal<br />

models of inflammation. During the period 1940–1950, the copper-containing<br />

compounds sodium 3-(N-allylcuprothiouredo)benzoate (Cupralene) and<br />

Alcuprin, given IV, sodium meta-(N-allylcuprothiocarbamide), given IM,<br />

and Cu(II) (4,6diethylammonium-8-hydroxyquinoline)2 sulfonate (Dicuprene<br />

or Cuprimyl), given IM, were reported by Forestier to be effective in<br />

treating a variety of arthritic diseases, including acute and chronic RA,<br />

chronic polyarticular gout, ankylosing spondylitis, and disseminated spondylitis.<br />

Forestier et al. (77) provided evidence in support of the suggestion that<br />

these copper complexes were superior to gold therapy.<br />

Work linking copper and inflammation has been done by Sorenson<br />

(4,5), who demonstrated that copper (II) acetate alone has an AI activity<br />

in experimental models of inflammation, with the suggestion that the active<br />

metabolites for AI activity are copper chelates. Sorenson and Hangarter (6)<br />

reported on clinical data obtained for some 1500 patients suffering from diffuse<br />

connective tissue disease over 30 years, noting that toxic side effects<br />

such as those occurring with chrysotherapy were not observed with copper<br />

therapy. Although these results were confirmed by others, the advent of<br />

hydrocortisone and the belief that it was going to ‘‘cure’’ arthritis led to the<br />

disuse of these preparations.<br />

Hangarter’s preparation of aq. copper salicylate, an aqueous mixture of<br />

12.5 mM of sodium salicylate and 0.04 mM Cu(II) chloride (Permalon), given<br />

IV, was more effective than the other clinically used complexes in the treatment<br />

of A and other degenerative diseases. That complex was effective in treating<br />

acute rheumatic fever, acute and chronic RA, erythema nodosum, and sciatica<br />

either with or without lower back spinal degenerative disease. From a total of<br />

620 patients treated over a 20-year period, 65% benefited by remission through<br />

this therapy, with a mean remission time of three years, 23% experienced<br />

improvements in their condition, while 12% remained unchanged (7). Hangarter<br />

mentions a lack of gastrointestinal complaints in the cohort of patients,<br />

which were common when only sodium salicylate was used in therapy. In<br />

1970, the manufacture of Permalon was discontinued for economic reasons,<br />

however, and Hangarter’s approach to therapy came to an end.<br />

TRANSDERMAL DELIVERY OF ANTI-INFLAMMATORY COPPER<br />

CHELATES VS. CONVENTIONAL (SYSTEMIC)<br />

ANTI-INFLAMMATORY THERAPY<br />

Since it appears important to maintain a needed minimal level of copper in<br />

tissues for the control of inflammation (78), the skin may be an adequate site


Copper in Anti-inflammatory Therapy 279<br />

for the delivery of cupriphores at maintenance levels. The case can therefore<br />

be made in favor of dermatologicals for copper-based therapy in the treatment<br />

of inflammation, since it may avoid patient discomfort and potential<br />

disadvantages such as the first-pass metabolism or idiosyncratic drug reactions<br />

and GI side effects inherent in conventional routes of administration<br />

(PO, ID, IP, IV, IA) (79–81). Due to lessened systemic clearance, local tissue<br />

accumulation of the AI agent would be possible in the treatment of conditions<br />

such as muscle pain and inflammation by low-level, controlled, and<br />

sustained release. As computer simulation of metal–ligand equilibria reveals<br />

that salicylates and similar complexes are thermodynamically unstable in<br />

plasma (82,83), this implies that the exogenous form of copper administered<br />

is different from the complex formed with endogenous ligands, and has no<br />

direct bearing on the anti-inflammatory activity. It thus appears fortuitous<br />

that most AI activity of copper has been studied using salicylate derivatives,<br />

in animals and humans, as AI activity of copper was exhibited by a wide variety<br />

of other ligands also. It is the increased availability of endogenous copper<br />

per se, which affords protection against inflammation. This is particularly<br />

encouraging in the search for skin-diffusible copper compounds, since the<br />

mechanism by which copper acts as the actual anti-inflammatory and antiarthritic<br />

agent, and what its bioactive moieties are, is still under discussion.<br />

In topical therapy, substances can be applied in concentrations that<br />

would be too toxic for systemic exposure. Given orally, compounds may<br />

contribute to gastric irritation (e.g., the salicylates), and the pharmacologically<br />

active form of copper complexes is also likely to be compromised (84).<br />

Copper complexes dissociate at the varying pH prevailing in the GI tract<br />

and become subject to sequestration and biliary excretion. Diffusion<br />

through the skin on the other hand avoids irritation in the GI tract and<br />

first-pass inactivation by the liver, and avoids discontinuity in therapy due<br />

to lack of patient compliance.<br />

Several theoretical and practical obstacles need to be overcome. With<br />

data presently available, progress in the development of copper-based<br />

dermatologicals for inflammatory disease therapy is severely limited. One<br />

key element awaiting development is data on the penetration of the ETE<br />

through human skin in any of its forms: as a polar mineral salt, or as a<br />

low molecular weight hydrophilic or larger lipophilic complex.<br />

Although dermatological copper preparations are, or were available,<br />

e.g., the topical anti-inflammatory copper-salicylate gels Alcusal 1 or Dermcusal<br />

1 , found to be effective in animals, they are not commonly employed<br />

for the treatment of RA in humans. This can be attributed to skepticism on<br />

the part of the medical profession due to the folkloristic component attached<br />

to such therapy, and also due to a lack of adequate controlled human studies.<br />

New treatments must be shown to be effective through unbiased, objective<br />

testing before the medical community acknowledges its potential benefits.<br />

Thus, an independent randomized placebo-controlled trial of the copper


280 Hosty´nek and Milanino<br />

salicylate–ethanol complex failed to show the effectiveness of the product:<br />

applied to the forearm it was no better than the placebo gel for (subjectively<br />

evaluated) pain relief in patients with osteoarthritis of the hip or knee (85).<br />

Skepticism prevails vis-à-vis copper-based therapy because its mode of<br />

anti-inflammatory action in the organism is insufficiently understood, and<br />

stability and fate of complexes delivered is unknown. For the electrophilic<br />

copper ion in particular, primarily binding sites in proteins may interfere<br />

with the dermal diffusion process.<br />

A major effort should be dedicated to the cutaneous delivery of cupriphores<br />

in quantities significant for local therapeutical activity. A first step is<br />

a de minimis approach: certain types of compounds could be defined as<br />

priority candidates for topical application of cupriphores for which Kps<br />

can be determined using standard in vitro permeation protocols, in order<br />

to put them on a relative scale of diffusivity. Such priority would go to small<br />

molecular weight complexes with ready solubility in polar media, which do<br />

not exhibit skin toxicity, bound to ligands of established safety. A prime<br />

such category that comes to mind are:<br />

Reaction products with low molecular weight essential amino acids<br />

such as glycine or histidine. Molecular size is a key determinant for<br />

optimal flux across the skin, and low molecular weight complexes will<br />

also determine the ability of plasma copper to diffuse into tissues.<br />

Unstable copper complexes with toxicologically safe profiles,<br />

which may release the metal to endogenous protein-binding sites<br />

in significant amounts, such as citrate, gluconate, histidinate, or<br />

sebacate. The latter category may also enhance the amount of copper<br />

absorbed, overall thanks to significant SC diffusion.<br />

The case can be made in favor of dermatologicals for copper-based<br />

therapy in the treatment of localized musculoskeletal disorders, as it allows<br />

targeted application of higher local drug concentrations too toxic systemically,<br />

avoiding or minimizing patient discomfort and untoward effects<br />

potentially incurred using conventional forms (IM, PO, IP, IV, IA) of treatment.<br />

Targeted exposure focusing on specific areas of inflammation becomes<br />

possible, providing higher local tissue concentration, rather than the blunderbuss<br />

approach through systemic therapy, which renders the agent subject<br />

to the vagaries of enterohepatic circulation and biliary excretion (81).<br />

As an instance, given orally compounds may contribute to gastric irritation<br />

or worse (e.g., the salicylates), and the pharmacological activity of<br />

copper complexes is also likely to be compromised (84). The compounds dissociate<br />

at the varying pH prevailing in the GI tract, and become subject to<br />

sequestration and excretion.<br />

Also, when a localized condition such as arthritis is treated systemically,<br />

the drug reaches the intended target only after resorption, passage<br />

through the liver, and distribution throughout the entire system. Apart from


Copper in Anti-inflammatory Therapy 281<br />

the potential for selective accumulation in the target area upon systemic dosing,<br />

the quantity of drug, which eventually becomes available on the intended<br />

target, is thus limited. Following systemic application, the pharmacokinetics<br />

are entirely different from those resulting from topical exposure. In the latter<br />

case, the drug can be applied directly targeting the afflicted part of the<br />

anatomy, i.e., the sites of clinical significance, with the potential for local<br />

skin reaction as the worst case. Since dissipation of the therapeutic agent<br />

due to transport throughout the system does not occur, topical, local therapy<br />

affords higher tissue concentrations, especially in the outer skin layers, which<br />

would be too toxic systemically.<br />

The potential for copper activity as an anti-inflammatory agent by<br />

transdermal delivery, however, is subject to controversy. The studies designed<br />

to demonstrate therapeutic benefits of topical therapy for arthritic conditions<br />

with either metallic copper or dermaceuticals such as Alcusal or Dermcusal 1<br />

by Walker et al. were not conducted lege artis. They were based on subjective<br />

patient assessment, obscured by placebo effect. Also, quantitative data for<br />

percutaneous penetration of copper’s putative oxidation products generated<br />

in contact of the metal with skin in humans, a critical factor in accrediting<br />

beneficial effects of copper bangles, have not been determined. Walker measured<br />

such absorption, albeit by one degree of separation, by gravimetrically<br />

determining the amount of copper lost in function of contact time only, and<br />

evaluating the resulting beneficial effect in afflicted patients by using questionnaires<br />

and psychologic parameters (18).<br />

Shackel et al. assessed the efficacy and safety of a copper-salicylate gel<br />

in osteoarthritis patients in a randomized, double-blind, placebo-controlled<br />

study, to be sure. But they were again based on self-assessment of pain,<br />

scores decreasing in both the treated and placebo groups, with no significant<br />

difference between the two groups for decrease in pain (85).<br />

Aside from the copper bangles issue, one missing, important factor for<br />

a convincing case of copper’s potential benefits is thus the deficiency in<br />

adequate, systematic research into the benefits of topical application of cupriphores.<br />

A measure of copper penetration through human skin in any of its<br />

forms, as polar mineral salts or as the more lipophilic chelates, is missing.<br />

Subcutaneous Administration of Cupriphores<br />

Favorable for transdermal therapy is evidence of anti-inflammatory activity<br />

obtained when bypassing the epidermal barrier through sc injection of<br />

cupriphores. Transdermal therapy as an effective route was documented<br />

repeatedly in animal models by sc administration of anti-inflammatory copper<br />

compounds (74,86,87). Notably, one of those investigations concluded<br />

that sc administration of some copper complexes was more effective than if<br />

they were given orally, while noting also that no difference was observed<br />

between the activities of copper (I) and copper (II) compounds in those


282 Hosty´nek and Milanino<br />

experiments (74). The latter serves to counter the argument that, in applying<br />

copper compounds, speciation of that ETE may be of importance.<br />

That levels of copper introduced into plasma strongly correlate with<br />

the anti-inflammatory effect was demonstrated when a wide range of<br />

copper–ligand complexes, administered sc, i.e., reaching the plasma without<br />

having to traverse the skin, were effective in controlling edema induced in<br />

animal screens (4,5,88).<br />

Positive such results were also obtained through sc administration of<br />

copper complexes by Lewis et al. An AI effect by sc dosing of copper aspirinate<br />

and other copper compounds in different animal species has been<br />

described (86).<br />

Precedents in Effective Transdermal Cupriphore Delivery<br />

in Animals and Humans<br />

High levels of 64 Cu radioactivity localized in loci of inflammation following<br />

dermal application of copper chelates by Beveridge indicated that exogenous<br />

copper was sequestered at inflammatory sites, and provides evidence<br />

of percutaneously absorbed copper complexes (75).<br />

Two lipophilic formulations of Cu(II) salicylate, Alcusal 1 and<br />

Dermcusal 1 , were developed by Walker et al. for dermal treatment of<br />

inflammatory conditions. Alcusal 1 is a copper salicylate–ethanol complex<br />

prepared by refluxing Cu(II) hydroxide with salicylic acid in anhydrous<br />

ethanol, Dermcusal 1 from copper salicylate prepared in a dimethyl sulfoxide–<br />

glycerol solution. Applied on the shaved dorsal skin of rats, they significantly<br />

reduced artificially induced paw edema and suppressed experimental arthritis,<br />

thus demonstrating facile cutaneous penetration (21,89,90).<br />

When the copper salicylate–ethanol complex was applied topically on<br />

the skin of human volunteers, analysis of elevated plasma salicylate levels<br />

did indicate that penetration had occurred. However, upon application on<br />

self-diagnosed arthritis and rheumatism patients, upon subjective evaluation<br />

no statistically significant results in pain relief could be documented (18).<br />

Overall, organic copper compounds to treat RA, albeit promising in<br />

the hands of many clinicians over time, led to inconsistent, and sometimes<br />

contradictory results. Pharmaceutical copper preparations are available<br />

but are not widely employed for the treatment of RA due to the effective<br />

and continuing use of chrysotherapy and the application of steroidal preparations,<br />

and also due to skepticism on the part of the medical profession<br />

in consideration of the folkloristic component attached to therapy based on<br />

copper. Further research proved inconclusive because of deficient experimental<br />

design in selection and evaluation/classification of patient cohorts,<br />

in the diagnostic criteria applied, the lack of controls, the confounding placebo<br />

effect, and to subjective evaluation by patients themselves, which was<br />

based on perception. Adding to such procedural deficiencies came the lack


Copper in Anti-inflammatory Therapy 283<br />

of marketing incentives at the pharmaceutical industry level. Thus, use of<br />

copper derivatives to treat RA and related inflammatory diseases gave<br />

way to other emerging anti-inflammatories such as Cox-2 inhibitors, which<br />

inhibit inducible cyclo-oxygenase enzymes and thereby inhibit prostaglandins,<br />

important mediators of inflammation. Health complications have<br />

become associated with that class of drugs now: aggravation of pre-existing<br />

ulcers, increase in the incidence of thrombosis, rise in blood pressure among<br />

others (32,91,92). They thus give rise to criticism and prompt inquiries on<br />

the part of health officials, favoring efforts to find alternatives for the treatment<br />

of inflammation.<br />

Should the epidermal barrier prove too formidable for diffusion of<br />

copper chelates to reach concentrations required for clinically meaningful<br />

analgesic and anti-inflammatory effect, however, several alternative techniques<br />

are now are available to carry drugs into or through the skin.<br />

Percutaneous Drug Delivery Systems and Methods<br />

Chemical Adjuvants<br />

There are limitations in the selection of drugs that may qualify for passive<br />

transdermal delivery (93). Passive percutaneous penetration, whether transcellular,<br />

via intercellular lipids or shunts, depends on several limiting factors,<br />

including molecular weight, optimally below 500 Da, or volume, lipophilicity,<br />

charge, and drug potency. To overcome some of the obstacles to passage<br />

of drugs through the SC, the first and most restricting barrier of the skin,<br />

research has endeavored to find chemical adjuvants, which would overcome<br />

the relatively low SC permeability to enhance penetration. Enhancement to<br />

deliver therapeutic levels has been achieved with formulations in which the<br />

active ingredients were compounded with terpene derivatives, e.g., linalool,<br />

limonene, menthone, or eugenol (94,95). Enhancers leading to higher drug<br />

levels in the dermis have chemical characteristics, which have the potential<br />

of disrupting the arrangement of the intercellular lipid bilayers and liquefing<br />

lipid sheets, structural changes that paradoxically are the very prerequisites<br />

for enhanced diffusion. Because exogenous penetration enhancers can<br />

exhibit overt toxic or irritant effects, their continued use for improved drug<br />

delivery is put in doubt.<br />

Dermal absorption of copper complexes by simple passive diffusion, particularly<br />

of those with higher molecular weight, e.g., Cu(II)-diclofenac or<br />

Cu(II)-ibuprofenato complex with AI effect, may be problematic (96,97).<br />

Methods of entrapment (liposomes) or active delivery by physical methods<br />

have been developed, however, and could also be successfully applied.<br />

Liposomes<br />

Liposomes (LSs) have been shown to efficiently deliver the large molecular<br />

weight class of proteins, such as antigens for immunization studies (98).


284 Hosty´nek and Milanino<br />

Depending on the molecular moiety to be delivered, LSs can be formulated<br />

to incorporate a wide array of substances as a payload, either in the water or<br />

in the lipid compartments, to contain antibodies, large molecular weight<br />

ligands, or polysaccharides.<br />

LSs are spherical uni- or multilamellar lipid/water vesicles with diameters<br />

in the micron-to-nanometer range. They are formed when thin lipid<br />

films or lipid cakes of natural, nontoxic phospholipids and cholesterol are<br />

hydrated and stacks of liquid crystalline bilayers become fluid and swell.<br />

The hydrated lipid sheets detach during agitation and self-assembly to form<br />

large, multilamellar vesicles that prevents interaction of water with the hydrocarbon<br />

core of the bilayer at the edges. The size of the particles can be reduced<br />

by sonication or extrusion. Properties of lipid formulations can vary depending<br />

on the composition (cationic, anionic, or neutral species) and degree of<br />

fatty acid saturation; the same preparation method can be used for all lipid<br />

vesicles regardless of composition (99).<br />

Biological particulate materials can be incorporated in LSs without<br />

compromising their structure or activity by exposing them to organic<br />

solvents, or light-sensitive compounds can be protected from radiation by<br />

incorporating a light-absorbing compound in the liposomal structure. Also,<br />

sensitive pharmaceuticals or labile bioactive materials can be encapsulated<br />

in an LS designed for controlled or sustained release, to degrade only at a<br />

target-specific application site. The LS structure is very similar, physiologically,<br />

to the material of cell membranes. When a formulation containing<br />

liposomes is applied to the skin, for example, the LSs are deposited on<br />

the skin and begin to merge with the cellular membranes. In the process, the<br />

LSs release their payload of active materials into the cells. As a consequence,<br />

not only is delivery of the actives very specific—directly into the intended<br />

cells—but the delivery takes place over a longer period of time. Thus, LSs<br />

as a delivery system can be designed for slow or fast release of both a hydrophilic<br />

or hydrophobic payload (100).<br />

The SC is recognized as the main barrier to skin penetration, as well as<br />

the major pathway for intracellular and intercellular drug delivery. Going a<br />

step further, Scheuplein (101–104) suggested that hair follicles also act as<br />

shunts, permitting rapid transport of charged and large polar molecules.<br />

Feldmann and Maibach (105) observed increased percutaneous transport<br />

through skin areas with high follicular density, also suggestive of follicular<br />

delivery. Refinements in methods of observation later confirmed this route<br />

for a wide range of molecules, particularly identifying particulate moieties<br />

such as LSs, localizing them in follicular and sebaceous areas to act as drug<br />

reservoirs (106,107). The potential for targeted percutaneous delivery of<br />

copper complexes of differing polarity and large molecular weight in LSs<br />

as carriers to the hair follicle may also offer an alternative in achieving therapeutically<br />

effective levels of that anti-inflammatory agent. Particularly<br />

attractive appears liposomal delivery of copper complexed with conventional


Copper in Anti-inflammatory Therapy 285<br />

anti-inflammatories that exhibit an augmenting activity, such as ibuprofen or<br />

diclofenac (96,97). These complexes appear unsuitable for conventional<br />

passive diffusion due to molecular size and limited solubility. Potentially<br />

nontoxic, degradable, and nonimmunogenic (108,109) LS acting as slow<br />

release vehicles could thus create a copper reservoir in the SC.<br />

Physical Methods<br />

Enhancement in skin absorption can be achieved by occlusion or different<br />

forms of energy input, characteristic of active transfer of drugs through the<br />

skin. These approaches utilize patch techniques, iontophoresis, sonophoresis,<br />

or electroporation to overcome the severe restrictions imposed on passage<br />

through the skin.<br />

Patches<br />

Transdermal absorption is a slow process of passive diffusion, driven by the<br />

gradient between the given concentration in the external delivery system and<br />

the zero concentration present in the skin. The delivery system using patches<br />

keeps the penetrant in continuous intimate contact with the skin with occlusion<br />

over days. Given adequate diffusion rates, steady blood levels can<br />

thereby be achieved through such continuous and even delivery over<br />

extended periods, which is preferable to the spikes and troughs occurring<br />

when administration occurs by other routes such as PO or IV. As an<br />

instance in delivery of analgesics, the fentanyl patch acts for 72 hours,<br />

providing long-lasting pain relief. Some other major drugs currently qualified<br />

for marketing as transdermal patches, intended for daily or weekly<br />

application, are clonidine (hypertension), estradiol (estrogen deficiency),<br />

estrogen-progestin (contraception), fentanyl (analgesic), lidocaine (anesthetic),<br />

nicotine (smoking cessation), nitroglycerine (angina pectoris), scopolamine<br />

(motion sickness), and testosterone.<br />

Iontophoresis<br />

Iontophoresis facilitates diffusion of ionic drugs and higher molecular<br />

weight compounds such as peptides. By anodal or cathodal electrophoresis<br />

negatively or positively charged drugs can be delivered respectively through<br />

the patient’s skin; under application of electric current one electrode serves<br />

to deliver the drug, the other to close the circuit. By this method luteinizing<br />

hormone-releasing hormone, a decapeptide of 1200 Da molecular weight<br />

was successfully dosed (110).<br />

Electroporation<br />

Electroporation is a technique that delivers high voltage pulses to the<br />

skin, causing transient changes in cell membranes or lipid bilayers (111).


286 Hosty´nek and Milanino<br />

Such pretreatment of the skin can facilitate delivery of large, polar molecules<br />

such as peptides.<br />

Sonophoresis<br />

Low-frequency ultrasound treatment of the skin, or sonophoresis, has been<br />

developed as another physical method to promote diffusion; this is for<br />

highly hydrophilic drugs like mannitol (112).<br />

CONCLUSIONS<br />

Copper as an essential trace element and its role in current anti-inflammatory<br />

therapies are reviewed, from the controversial, age-old practice of wearing<br />

copper bangles to the copper complexes as anti-inflammatory drugs. Oxidation<br />

products formed by copper in contact with the skin and their potential<br />

for their skin diffusion are discussed. Considering the potentially negative<br />

side effects as well as the discomfort associated with anti-inflammatory<br />

therapies using current methods of application, transdermal delivery of<br />

cupriphores appears to be an attractive alternative worth exploring, although<br />

the present lack of data on the skin diffusivity for all categories of copper<br />

compounds is a shortcoming, which must first be overcome. Should transdermal<br />

delivery of copper chelates lessen or suppress inflammation, untoward<br />

systemic effects associated with conventional modes of anti-inflammatory<br />

therapy could be lessened or avoided.<br />

Aside from general principles of percutaneous absorption and prerequisites<br />

for skin penetration by chemicals, the potential for topical delivery<br />

of copper complexes is reviewed with the intent to impart renewed impetus<br />

to the evaluation of transdermal anti-inflammatory therapy with cupriphores.<br />

Strategies are identified for their passive or active diffusion in order<br />

to penetrate the stratum corneum barrier, with the purpose to compensate<br />

with exogenous sources delivered through the skin potential, local shortfalls<br />

in endogenous reserves of copper.<br />

Evidence gathered in animals and humans affected by inflammatory<br />

processes suggests that percutaneous administration of copper may represent<br />

an appealing therapeutic approach, particularly in consideration of<br />

the unfavorable aspects of adverse side effects and discomfort associated<br />

with more conventional therapies. Also, applied dermally, the drug in the<br />

target tissues can be expected to exceed the concentrations achievable<br />

through more conventional, systemic avenues.<br />

At this time, several theoretical and practical obstacles prejudice<br />

endeavors in the development of copper-based dermatologicals<br />

for the therapy of inflammatory diseases, regardless of the form<br />

in which that metal may be applied.


Copper in Anti-inflammatory Therapy 287<br />

Although dermatological copper preparations are available, they<br />

are not commonly employed for the treatment of RA in humans.<br />

Skepticism on the part of the medical profession persists due to a<br />

lack of controlled efficacy studies.<br />

A missing building block towards making a convincing case for the<br />

potential benefits in transdermal therapy is the lack of quantitative<br />

data on the penetration rate of copper through human skin, in any<br />

of its forms: derivatives of skin-identical amino acids, prepared in<br />

analogy with peptides potentially formed in contact of the metal<br />

with the skin (glycinate, histidinate, etc.), as polar mineral salts,<br />

or as the more lipophilic chelates (e.g., as ibuprofen or diclofenac<br />

complexes) applied as liposomes.<br />

Principles governing the transfer of metal compounds through biological<br />

membranes, including the skin, are prejudiced by numerous<br />

factors that are difficult to anticipate a priori. For copper in particular<br />

as an ETE, binding sites in proteins may interfere with the<br />

dermal diffusion process, resulting in degradation or blockage<br />

(homeostatic controls).<br />

Studies designed to demonstrate significant therapeutic benefits for<br />

arthritic conditions through skin contact with metallic copper or its<br />

complexes in humans, e.g., with oxidation products that may be<br />

generated in contact of the metal with the skin have not been conducted<br />

in a convincing manner.<br />

That transdermal AI therapy can be effective has already been demonstrated<br />

in animal models by administration of a number of copper<br />

compounds (74,86,87). The investigation by Brown et al. (74) concluded that<br />

administration of some of the complexes was more effective than when given<br />

orally, and no obvious differences were observed between the activities of<br />

copper (I) and copper (II) compounds.<br />

Endeavors should focus on achieving transport of copper compounds<br />

through the skin in pharmacologically significant quantity. To overcome<br />

some of the obstacles, as a first step in a de minimis approach, small<br />

molecular weight amino acids could be investigated for skin diffusivity.<br />

Such copper chelates may release the metal to protein binding sites in significant<br />

amounts.<br />

Technical arguments elaborated at this time in favor of transdermal<br />

cupriphores indicate that:<br />

1. Regardless of the ligand with which copper reaches the organism,<br />

the metal in the initial complex recombines with endogenous proteins<br />

and is homeostatically directed to the target locus in the<br />

organism where the metal exercises its function.<br />

2. Both Cu(I) and Cu(II) in the exogenous complex are equally<br />

effective in reducing inflammation, speaking for a common


288 Hosty´nek and Milanino<br />

(recombinant) transition state of that highly reactive metal within<br />

the organism, a characteristic shared by other transition metals.<br />

Copper exerts its role as ETE, thanks to that ability of the outer<br />

electrons in the 3d and 4s orbital shells, which allow it to fluctuate<br />

between the monovalent and the divalent oxidation state. Such<br />

facile changes in valence play a decisive role for their transfer<br />

across cell membranes and epithelia, including the skin.<br />

OUTLOOK<br />

Summing up the factors enumerated, the essential groundwork for the transdermal<br />

delivery of cupriphores appears to have been laid. An array of<br />

technologies for their delivery are available; preliminary results obtained<br />

so far in animals and humans warrant pursuit of that form of therapy<br />

and make a resurgence in the use of copper as anti-inflammatory possible.<br />

Albeit dermal diffusion of copper complexes formed by the metal in contact<br />

with the skin have been demonstrated (not published), transdermal application<br />

in form of patches would appear to be a more efficient method of<br />

dosing exogenous copper.<br />

Closer definition of copper complex stability is not necessary, since<br />

homeostasis takes control of the ETE in whatever form exogenous copper<br />

reaches the organism and converts it to endogenous complexes appropriate<br />

for anti-inflammatory function. Should cupriphores be adapted as<br />

patches for needle-free transdermal delivery, by use of chemical adjuvants,<br />

physical preconditioning of the skin, or imbedded in LS, some of the traditional<br />

remedies for RA (e.g., gold therapy or copper bracelets) may be<br />

replaced by effective and less controversial approaches. Transdermal therapy<br />

would bring with it the advantages of lesser immunotoxicity, of selfadministration,<br />

avoidance of discomfort, and economy.<br />

ABBREVIATIONS<br />

A arthritis<br />

AI anti-inflammatory<br />

CP ceruloplasmin<br />

ETE essential trace element<br />

GI gastrointestinal<br />

IA intra-articular<br />

IM intramuscular<br />

IV intravenous<br />

L ligand<br />

LS liposomes<br />

MT metallothionein<br />

NSAIDs nonsteroidal anti-inflammatory drugs


Copper in Anti-inflammatory Therapy 289<br />

PO per os<br />

QSAR quantitative structure activity relationships<br />

RA rheumatoid arthritis<br />

sc subcutaneous<br />

SC stratum corneum<br />

REFERENCES<br />

1. Linder MC. Copper uptake by nongastrointestinal vertebrate cells. In: Biochemistry<br />

of copper. New York, NY: Plenum Press, 1991:58–71.<br />

2. Conforti A, Franco L, Milanino R, et al. Copper and ceruloplasmin concentrations<br />

during the acute inflammatory process in the rat. Agents Actions 1982;<br />

12:303.<br />

3. Milanino R, Marrella M, Crivellente F, et al. Nutritional supplementation with<br />

copper in the rat. Vol. I. Effects on adjuvant arthritis development and on some<br />

in vivo and ex vivo-markers of blood neutrophils. Inflam Res 2000; 49:214.<br />

4. Sorenson JRJ. Copper chelates as possible active forms of the antiarthritic<br />

agents. J Med Chem 1976; 19:135.<br />

5. Sorenson JRJ. Some copper coordination compounds and their antiinflammatory<br />

and antiulcer activities. Inflammation 1976; 1:317.<br />

6. Sorenson JRJ, Hangarter W. Treatment of rheumatoid arthritis and degenerative<br />

diseases with copper complexes. Inflammation 1977; 2:217.<br />

7. Hangarter W. Copper salicylate in rheumatoid arthritis and similar degenerative<br />

diseases. In: Sorenson JRJ, ed. Inflammatory diseases and copper. Clifton,<br />

NJ: Humana Press, 1982:439.<br />

8. Sorenson JRJ. Copper complexes as the active metabolites of antiinflammatory<br />

agents. In: Sorenson JRJ, ed. Inflammatory diseases and copper. Clifton, NJ:<br />

Humana Press, 1982.<br />

9. Sorenson JRJ, Berthon G. Copper potentiation of non-steroidal antiinflammatory<br />

drugs. In: Handbook of metal–ligand interactions in biological fluids.<br />

New York: Marcel Dekker, Inc., 1995:1318.<br />

10. Milanino R, Frigo A, Bambara LM, et al. Copper and zinc status in rheumatoid<br />

arthritis: studies of plasma, erythrocytes, and urine, and their relationships to<br />

disease activity markers and pharmacological treatment. Clin Exp Rheumatol<br />

1993; 11:271.<br />

11. Milanino R, Conforti A, Franco L, et al. Copper and inflammation—a possible<br />

rationale for the pharmacological manipulation of inflammatory disorders.<br />

Agents Actions 1985; 16:6.<br />

12. Rainsford KD, Whitehouse MW. Concerning the merits of copper aspirin as<br />

potential anti-inflammatory drug. J Pharm Pharmacol 1976; 28:83.<br />

13. CDC. Targeting arthritis. Atlanta, Ga.: DHHS, 2003.<br />

14. Merck. Manual of diagnosis and treatment. 17th ed. Rahway: Merck & Co.,<br />

Inc., 1999.<br />

15. Pincus T, Sokka T. Rheumatoid arthritis. In: Rakel RE, Bope ET, eds. Conn’s<br />

current therapy. Philadelphia: Saunders Elsevier Inc., 2004:1021.<br />

16. Mez-Mangold L. A history of drugs. Basel: F. Hoffman La Roche & Co., 1971.


290 Hosty´nek and Milanino<br />

17. Walker WR, Griffin BJ. The solubility of copper in human sweat. Search 1976;<br />

7:100.<br />

18. Walker WR, Beveridge SJ, Whitehouse MW. Dermal copper drugs: the<br />

copper bracelet and Cu(II) salicylate complexes. Agents Actions Suppl 1981;<br />

8:359.<br />

19. Walker WR, et al. Perfusion of intact skin by a saline solution of bis(glycinato)<br />

copper(II). Bioinorg Chem 1977; 7:271.<br />

20. Schmid R, Winkler J. Über die Kutane Kupferresorption aus einer Kupfer<br />

enthaltenden Salbe. Klin Wochenschr 1938; 17:559.<br />

21. Beveridge SJ, Boettcher B, Walker WR, et al. Biodistribution of 64Cu in rats<br />

after topical application of two lipophilic anti-inflammatory Cu(II) formulations.<br />

Agents Actions 1984; 14:291.<br />

22. Odintsova NA. Permeability of human skin to potassium and copper ions and<br />

their ultrastructural localization. Chem Abstr 1978; 89:360.<br />

23. Abraham MH, Chadha HS, Mitchell RC. The factors that influence skin penetration<br />

of solutes. J Pharm Pharmacol 1995; 47:8.<br />

24. Flynn GL. Physicochemical determinants of skin absorption. In: Gerrity TR,<br />

Henry CJ, eds. Principles of route-to-route extrapolation for risk assessment.<br />

New York: Elsevier Science Publishing Co., 1990:93.<br />

25. Lim CW, Fujiwra S, Yamashita F, et al. Prediction of human skin permeability<br />

using a combination of molecular orbital calculations and artificial neural network.<br />

Biol Pharm Bull 2002; 25:361.<br />

26. McCarley KD, Bunge AL. Physiologically relevant two-compartment pharmacokinetic<br />

models for skin. J Pharm Sci 2000; 89:1212.<br />

27. Potts RO, Guy RH. A predictive algorithm for skin permeability: the effects of<br />

molecular size and hydrogen bond activity. Pharm Res 1995; 12:1628.<br />

28. Singh P, Roberts MS. Skin permeability and local tissue concentrations of nonsteroidal<br />

anti-inflammatory drugs after topical application. J Pharmacol Exp<br />

Therap 1994; 268:144.<br />

29. Hadgraft J, du Plessis J, Goosen C. The selection of non-steroidal anti-inflammatory<br />

agents for dermal delivery. Int J Pharm 2000; 207:31.<br />

30. Jackson GE, Mkhonta-Gama L, Voye A, et al. Design of copper-based antiinflammatory<br />

drugs. J Inorg Biochem 2000; 79:147.<br />

31. Dillon CT, Hambley TW, Kennedy BJ, et al. Copper and zinc complexes as<br />

antiinflammatory drugs. Metal Ions Biol Sys 2004; 41:253.<br />

32. Muscara MN, Vergnolle N, Lovren F, et al. Selective cyclo-oxygenase-2 inhibition<br />

with celecoxib elevates blood pressure and promotes leukocyte adherence.<br />

Br J Pharmacol 2000; 129:1423.<br />

33. Forestier JL. Aurotherapie dans les rheumatismes chroniques. Bulletin et Membres<br />

Societe Medicale d’Hopital de Paris 1929; 53:323.<br />

34. Davis P. Undesirable effects of gold. J Rheumatol (Suppl) 1979; 6:18.<br />

35. Husby G, Gran JT. Risk factors in the treatment of rheumatoid arthritis with<br />

parenteral gold. Scand J Rheumatol (Suppl) 1983; 51:112.<br />

36. Zwaifler J. Gold and automalarial therapy. In: McCarty DJ, ed. Arthritis and<br />

applied conditions. 9th ed. Philadelphia: Lea & Febiger, 1979:355.<br />

37. Anonymous. Diffuse connective tissue disease: gold compounds. In: Beers MH,<br />

Berkow R, eds. The Merck manual. 17th ed. Whitehouse Station, NJ: Merck<br />

Research Laboratories, 2004:420.


Copper in Anti-inflammatory Therapy 291<br />

38. Freyberg RH. Gold therapy for rheumatoid arthritis. In: Hollander JL, ed.<br />

Arthritis and allied conditions. Philadelphia: Lea and Febiger, 1972:455.<br />

39. Penneys NS. Gold therapy: dermatologic uses and toxicities. J Am Acad<br />

Dermatol 1979; 1:315.<br />

40. Comroe BI. The use and abuse of gold therapy in rheumatoid arthritis. JAMA<br />

1945; 128:848.<br />

41. Stoyanov JV, Brown NL. The Escherichia coli copper-responsive copA promoter<br />

is activated by gold. J Biol Chem 2005; 278:1407.<br />

42. Harth M. Modulation of immune response by gold salts. Agents Actions 1981;<br />

8:465.<br />

43. Kazantzis G. The role of hypersensitivity and the immune response in influencing<br />

susceptibility to metal toxicity. Environ Health Persp 1978; 25:111.<br />

44. Palosuo T, Provost TT, Milgrom F. Gold nephropathy: serologic data suggesting<br />

an immune complex disease. Clin Exp Immunol 1976; 25:311.<br />

45. Browning JS, et al. Gold therapy in rheumatoid arthritis. N Engl J Med 1947;<br />

237:428.<br />

46. Fisher AA. Possible combined nickel and gold allergy. Am J Contact Dermatitis<br />

1992; 3:52.<br />

47. Silva R, Pereira F, Silva E, et al. Contact allergy to gold sodium thiosulfate. In:<br />

Jadassohn centenary congress abstracts. London: E. S. o. C. D. a. A. C. D.<br />

Society, 1996:13.<br />

48. Bruze M, Björkner B, Möller H. Skin testing with gold sodium thiomalate and<br />

gold sodium thiosulfate. Contact Dermatitis 1995; 32:5.<br />

49. Bruze M, Hedman H, Björkner B, et al. The development and course of test<br />

reactions to gold sodium thiosulfate. Contact Dermatitis 1995; 33:386.<br />

50. McKenna KE, Dolan O, Walsh MY, et al. Contact allergy to gold sodium thiosulfate.<br />

Contact Dermatitis 1995; 32:143.<br />

51. Nakada T, Iijima M, Fujisawa R. The dissociation of results of patch test to<br />

ionized/non-ionized mercury between skin contact dermatitis due to mercury<br />

and gold. Jap J Dermatoallergol 1994; 1:237.<br />

52. Denman EJ, Denman AM. The lymphocyte transformation test and gold<br />

hypersensitivity. Ann Rheum Dis 1968; 27:582.<br />

53. Cortell R, Richards RK. Development of tolerance to gold salts in rats. Proc<br />

Soc Exp Biol Med 1942; 49:121.<br />

54. Davis P, Ezeoke A, Munro J, et al. Immunological studies on the mechanism of<br />

gold hypersensitivity reactions. BMJ 1973; 3:676.<br />

55. Davis P, Hughes GRV. A serial study of eosinophilia and raised IgE antibodies<br />

during gold therapy. Ann Rheum Dis 1975; 34:203.<br />

56. Törnroth T, Skrifvars B. Gold nephropathy prototype of membranous glomerulonephritis.<br />

Am J Pathol 1974; 75:573.<br />

57. Watanabe I, et al. Gold nephropathy. Arch Pathol Lab Med 1976; 100:632.<br />

58. Björkner B, Bruze M, Möller H. High frequency of contact allergy to gold<br />

sodium thiosulfate. Contact Dermatitis 1994; 30:144.<br />

59. Bruze M, Edman B, Björkner B, et al. Clinical relevance of contact allergy to<br />

gold sodium thiosulfate. J Am Acad Dermatol 1994; 31:579.<br />

60. Marks JG, Belsito DV, DeLeo VA, et al. North American Contact Dermatitis<br />

Group patch test results for the detection of delayed-type hypersensitivity to<br />

topical allergens. Am Acad Dermatol 1998; 38:911.


292 Hosty´nek and Milanino<br />

61. Fowler JF. Allergic contact dermatitis to gold in North America [abstract #46]<br />

Jadassohn Centenary Congress Abstracts. London, 1996:13.<br />

62. Möller H, Björkner B, Bruze M. Clinical reactions to systemic provocation with<br />

gold sodium thiomalate in patients with contact allergy to gold (abstract #50).<br />

In: Jadassohn centenary congress abstracts. London: E. S. o. C. D. a. A. C. D.<br />

Society, 1996:14.<br />

63. Möller H, Ohlsson K, Björkner B, et al. Morphology of flare up reactions in<br />

contact allergy to gold (abstract #245). In: Jadassohn centenary congress<br />

abstracts. London: E. S. o. C. D. a. A. C. D. Society, 1996:63.<br />

64. Möller H, Björkner B, Bruze M, et al. Cytokines and C-reactive protein in the<br />

systemically provoked flare up of contact allergy to gold (abstract #246). In:<br />

Jadassohn centenary congress abstracts. London: E. S. o. D. a. A. C. D.<br />

Society, 1996:63.<br />

65. Beliles RP. Gold, Au. In: Clayton GD, Clayton FE, eds. Patty’s industrial<br />

hygiene and toxicology. 4th ed. New York: John Wiley & Sons, Inc., 1994:2021.<br />

66. Rister M. Superoxide anion and superoxide dismutase activity in arthritic<br />

condition, Agents Actions Suppl 1981; 8:137.<br />

67. Rosenstein ED. Topical agents in the treatment of rheumatic disorders. Rheum<br />

Dis Clin North Am 1999; 25:899.<br />

68. Furnival C, May PM, Williams DR. Models of low-molecular-weight copper(II)<br />

complexing equilibria in relation to rheumatoid arthritis. In: Rainsford KD,<br />

Brune K, Whitehouse MW, eds. Trace elements in the pathogenesis and treatment<br />

of inflammation. Basel: Birkhaeuser Verlag, 1981:241.<br />

69. Heilmeyer I, Stuwe G. Der Eisen-kupferantagonismus im Blut-pasma beim<br />

Infektionsgeschehen. Klin Wochenschr 1938; 17:925.<br />

70. Denko CW. Protective role of ceruloplasmin in inflammation. Agents Actions<br />

1979; 9:333.<br />

71. Milanino R, Cassini A, Conforti A, et al. Copper and zinc status during acute<br />

inflammation: studies on blood, liver and kidneys metal levels in normal and<br />

inflamed rats. Agents Actions 1986; 19:755.<br />

72. Bonta IL, Parnham MJ, Vincent JE, et al. Anti-rheumatic drugs: present deadlock<br />

and new vistas. Progr Med Chem 1980; 17:186.<br />

73. Sorenson JRJ. The anti-inflammatory activities of copper complexes. In:<br />

Siegel A, ed. Metal ions in biological systems. New York: Marcel Dekker,<br />

1982:77.<br />

74. Brown DH, Smith WE, Teape JW, et al. Antiinflammatory effects of some copper<br />

complexes. J Med Chem 1980; 23:729.<br />

75. Beveridge SJ, Walker WR, Whitehouse MW. Anti-inflammatory activity of<br />

copper salicylates applied to rats percutaneously in dimethyl sulphoxide with<br />

glycerol. J Pharm Pharmacol 1980; 32:425.<br />

76. Powanda MC. Systemic alterations in metal metabolism during inflammation<br />

as part of an integrated response to inflammation. Agents Actions Suppl<br />

1981; 8:121.<br />

77. Forestier J, Certonciny A, Jacqueline F. Therapeutic value of copper salts inn<br />

rheumatoid arthritis. Stanford Med Bull 1950; 8:12.<br />

78. Milanino R, Marrella M, Gasperini R, et al. Carrageenan oedema in copperdeficient<br />

rats. Agents Actions 1978; 8:618.


Copper in Anti-inflammatory Therapy 293<br />

79. Szabo S, Spill WF, Rainsford KD. Non-steroidal anti-inflammatory druginduced<br />

gastropathy. Mechanism and management. Med Toxicol Adv Drug<br />

Experience 1989; 4:77.<br />

80. Hawkey CJ. Non-steroidal anti-inflammatory drugs and peptic ulcers. BMJ<br />

1990; 300:278.<br />

81. Crispens GG, Sorenson JRJ. Evaluation of the anticancer activity of CuDIPS<br />

in SJL/J mice. Anticancer Res 1988; 8:77.<br />

82. May PM, Linder PW, Williams DR. Computer simulation of metal-ion equilibria<br />

in biofluids: models for the low-molecular-weight complex distribution of<br />

calcium(II), magnesium(II), manganese(II), iron(III), copper(II), zinc(II), and<br />

lead(II) ions in human blood plasma. J Chem Soc Dalton Trans 1977; 6:588.<br />

83. Jackson GE, May PM, Williams DR. Metal–ligand complexes involved in<br />

rheumatoid arthritis—I. J Inorg Nucl Chem 1978; 40:1189.<br />

84. Milanino R, Concari E, Conforti A, et al. Synthesis and anti-inflammatory<br />

effects of some bis-(2-benzimidazolyl) thioethers and their copper(II) chelates,<br />

orally administered to rats. Eur J Med Chem 1988; 23:217.<br />

85. Shackel NA, Day RO, Kellett B, et al. Copper-salicylate gel for pain relief in<br />

osteoarthritis: a randomised controlled trial. Med J Aust 1997; 167:134.<br />

86. Lewis AJ. A comparison of the anti-inflammatory effects of copper aspirinate<br />

and other copper salts in the rat and guinea pig. Agents Actions 1978; 8:244.<br />

87. Mann JR, Camakaris J, Danks DM, et al. Copper metabolism in mottled<br />

mouse mutants. Biochem J 1979; 180:605.<br />

88. Jackson GE, May PM, Williams DR. Metal–ligand complexes involved in<br />

rheumatoid arthritis—VI. J Inorg Nucl Chem 1978; 40:1227.<br />

89. Walker WR, Beveridge SJ, Whitehouse MW. Antiinflammatory activity of a dermally<br />

applied copper salicylate preparation (Alcusal). Agents Actions 1980; 10:1.<br />

90. Beveridge SJ, Whitehouse MW, Walker WR. Lipophilic copper(II) formulations:<br />

some correlations between their composition and anti-inflammatory/antiarthritic<br />

activity when applied to the skin of rats. Agents Actions 1982; 12:225.<br />

91. Laudanno OM, Cesolari JA, Esnarriaga J, et al. Gastrointestinal damage induced<br />

by celecoxib and rofecoxib in rats. Acta Gastroenterol Latinoam 2000; 30:27.<br />

92. Wallace JL, Muscara MN. Selective cyclo-oxygenase-2 inhibitors: cardiovascular<br />

and gastrointestinal toxicity. Digest Liver Dis 2001; 33(suppl 2):S21.<br />

93. Smith EW, Maibach HI. Percutaneous penetration enhancers: the fundamentals.<br />

In: Smith EW, Maibach HI, eds. Percutaneous penetration enhancers. Boca<br />

Raton: CRC Press, 1995.<br />

94. Vaddi HK, Ho PC, Chan SY. Terpenes in propylene glycol as skin-penetration<br />

enhancers. J Pharm Sci 2002; 91:1639.<br />

95. Zhao K, Singh J. Mechanism of in vitro percutaneous penetration enhancers of<br />

tamoxifen. J Pharm Sci 2000; 89:771.<br />

96. Kovala-Demertzi D. Transition metal complexes of diclofenac with potentially<br />

interesting anti-inflammatory activity. J Inorg Biochem 2000; 79:153.<br />

97. Andrade A, Namora SF, Woisky RG, et al. Synthesis and characterization of a<br />

diruthenium-ibuprofenato complex. Comparing its anti-inflammatory activity<br />

with that of a copper(II)-ibuprofenato complex. J Inorg Biochem 2000; 81:23.<br />

98. Choy I, Maibach HI. Topical vaccination of DNA antigens. Skin Pharmacol<br />

Appl Physiol 2003; 16:271.


294 Hosty´nek and Milanino<br />

99. Cevc G. Lipid vesicles and other colloids as drug carriers on the skin. Adv Drug<br />

Del Rev 2004; 56:675.<br />

100. Schwendener RA, Trub T, Schott H, et al. Comparative studies of the preparation<br />

of immunoliposomes with the use of two bifunctional coupling agents and<br />

investigation of in vitro immunoliposome-target cell binding by cytofluorometry<br />

and electron microscopy. Biochim Biophys Acta 1990; 1026:69.<br />

101. Scheuplein RJ. Mechanism of percutaneous absorption. I. Routes of penetration<br />

and the influence of solubility. J Invest Dermatol 1965; 45:334.<br />

102. Scheuplein RJ. Mechanism of percutaneous absorption II. Transient diffusion<br />

and the relative importance of various routes of skin penetration. J Invest<br />

Dermatol 1967; 48:79.<br />

103. Scheuplein RJ, Blank IH. Molecular structure and diffusional processes across<br />

intact skin. Report to the US Army Chemical R&D Laboratories, Edgewood<br />

Arsenal, Md., 1967.<br />

104. Scheuplein RJ, Blank IH. Mechanism of percutaneous absorption. Vol. IV.<br />

Penetration of nonelectrolytes (alcohols) from aqueous solutions and from pure<br />

liquids. J Invest Dermatol 1973; 60:286.<br />

105. Feldmann RJ, Maibach HI. Regional variation in percutaneous penetration of<br />

C-14 cortisol in man. J Invest Dermatol 1967; 48:181.<br />

106. Lauer AC, Elder JT, Weiner ND. Transfollicular drug delivery. Pharm Res<br />

1995; 12:179.<br />

107. Lieb LM, Ramachandran C, Egbaria K, et al. Topical delivery enhancement<br />

with multilamellar liposomes into pilosebaceous units: I. In vitro evaluation<br />

using fluorescent techniques with the hamster ear model. J Invest Dermatol<br />

1992; 99:108.<br />

108. Egbaria K, Ramachandran C, Kittayanond D, et al. Topical delivery of liposomically<br />

encapsulated interferon evaluated by in vitro diffusion studies. Antimicrob<br />

Agents Chemother 1990; 34:107.<br />

109. Egbaria K, Ramachandran C, Weiner ND. Topical application of liposomally<br />

entrapped cyclosporin evaluated in vitro diffusion studies with human skin.<br />

Skin Pharmacol 1991; 4:21.<br />

110. Swarbrick J, Boylan JC, eds. Encyclopedia of pharmaceutical technology.<br />

2nd ed. New York: Marcel Dekker, Inc., 2002:953.<br />

111. Sharna A, Kara M, Smith FR, et al. Transdermal drug delivery using electroporation<br />

I. Factors influencing in vitro delivery of terazolin HCl in hairless rats.<br />

J Pharm Sci 2000; 89:528.<br />

112. Tang H, Blankschtein D, Langer R. Effects of low-frequency ultrasound on the<br />

transdermal penetration of mannitol. Comparative studies with in vivo and in<br />

vitro skin. J Pharm Sci 2002; 91:1776.


Absorption, 22<br />

degree of, 76<br />

dermal, 22<br />

measuring percutaneous, 45<br />

limitations and problems<br />

in, 46<br />

radioactivity method for, 48<br />

in vitro method for, 45–47, 52<br />

in vivo methods for, 47–52<br />

of metal compounds, 22, 46<br />

skin, 22<br />

Acute inflammation, 162, 163, 166<br />

Acute periodontal disease, 161<br />

Adjuvants, chemical, 283<br />

Alcusal 1 , 200<br />

Allergic contact dermatitis (ACD),<br />

116, 272<br />

copper, 128<br />

test concentrations for, 123<br />

diagnostic tool for, 120<br />

predictive test for, 126<br />

See also Allergy, delayed-type<br />

Allergy<br />

delayed-type, 116, 120, 139<br />

immediate-type, 119<br />

Alloys<br />

brass, 5<br />

bronze, 5–6<br />

cast, 4<br />

Index<br />

295<br />

[Alloys]<br />

chromium coppers, 5<br />

compositions of, 104<br />

copper, wrought, 4<br />

copper–nickel, 2, 6<br />

nickel–silver, 6<br />

solubility of particulate, 107–108<br />

Alpha-ceruloplasmin, 74<br />

Aluminum bronzes, 5<br />

applications of, 6<br />

characteristics of, 6<br />

American Conference of Governmental<br />

and Industrial Hygienists<br />

(ACGIH), 98<br />

Amino acids, 11<br />

Analgesic–narcotic principles, 152<br />

Analytical electron microscopy, 69<br />

Angry back syndrome, 131<br />

Anti-inflammatory (AI)<br />

activity, 68, 167<br />

copper chelates as, 243<br />

topical delivery of, 275<br />

Anti-inflammatory drugs,<br />

nonsteroidal, 238<br />

Anti-inflammatory potentials, 150<br />

Anti-inflammatory properties, 150<br />

Antiarthritic<br />

agents, 162<br />

application, 244


296 Index<br />

[Antiarthritic]<br />

copper potential, 159<br />

drugs, 162<br />

Antirheumatic agents, 187<br />

Antiseptic properties, 151<br />

Area under the curve (AUC), of<br />

triplicate stripping<br />

experiments, 88<br />

Arthritic deformation, 240<br />

Arthritis<br />

analgesic products, 255<br />

pain relief, 249<br />

patients with copper jewelry, 82<br />

remedy for, 238<br />

rheumatoid, 238, 250<br />

treatment, 243<br />

Artificial sweat<br />

copper ion in, 13<br />

skin penetration data for, 13<br />

Aspirin, 190, 271<br />

for analgesic activity, 271<br />

an antipyretic, 271<br />

Atomic absorption analysis, 14<br />

Biliary excretion, 279<br />

Blue copper oxidases, 186<br />

Bracelets, 240<br />

copper, 239<br />

therapeutic, 240<br />

weight loss, 247–248<br />

Brass, (red or yellow), 5<br />

Breathable tape, 82<br />

Bronze<br />

alloy, 5–6<br />

aluminum, 5<br />

silicon, 5<br />

tin, properties of, 5<br />

Bronze Age, 5<br />

Carrageenan-induced paw edema<br />

(CPE), 163, 165<br />

Carrageenan-induced pleurisy<br />

(CP), 165<br />

Cast alloys, 4<br />

Ceruloplasmin, 164, 167<br />

concentrations, 172<br />

[Ceruloplasmin]<br />

in serum, 181<br />

as markers, 172<br />

transcription of, 171<br />

Chemical adjuvants, 283<br />

Chemical irritants, 98<br />

Chlorophyllin copper complex, 102<br />

Cholinergic urticaria reaction, 139<br />

Chondrocytes, sensitivity of, 207<br />

Chromatography, ion exchange, 11<br />

Chromium copper alloys, applications<br />

for, 5<br />

Chronic inflammation, 162,<br />

168–170, 171<br />

Chronic inflammatory agents, 152, 162<br />

Chronic inflammatory disease, 238<br />

Chronic inflammatory processes, 163<br />

Chronic systemic pathologies, 199<br />

Chrysotherapy, 272<br />

applications of, 282<br />

immune reactions in, 273<br />

Complementary and alternative<br />

medicine (CAM), 246<br />

Connective tissue metabolism, 206<br />

Contact urticaria syndrome, 116<br />

Copper<br />

absorption, 67, 74, 72, 83, 244, 267<br />

cutaneous, 67, 74<br />

dermal, 255, 270, 283<br />

administration<br />

exogenous effects, 184–203<br />

percutaneous, 199–203<br />

routes of, 184–186<br />

subcutaneous and intraperitoneal,<br />

188–191<br />

allergy, 121, 123, 128<br />

alloy<br />

applications of, 2<br />

biofouling resistance of, 3<br />

castability of, 3<br />

characteristics of, 2–3<br />

corrosion resistance of, 2<br />

fabricability of, 3<br />

families, 3<br />

friction rates of, 3<br />

wear rates of, 3<br />

ANA 68 alloy, 104<br />

animal models for, 276


Index 297<br />

[Copper]<br />

anti-inflammatory activity of, 67, 188,<br />

203–216, 268, 275<br />

anti-inflammatory effects, 91–92<br />

antimicrobials, 102<br />

as applications, 84–85<br />

articles, corrosion of, 68<br />

baseline values of, 82–84, 88<br />

blood clotting factors, 8<br />

bracelet, 239, 255, 269<br />

myth, study of, 249<br />

weight loss, 13, 269<br />

chelates in rheumatoid arthritis<br />

(RA), 278<br />

chemicals, 3<br />

chlorophyllin, 102<br />

comparison of nickel and, 87, 89<br />

complexes, 7–8, 158<br />

compounds, 67<br />

concentration, 172, 181, 182<br />

content of hair and skin, 89<br />

contraceptive effects, 14<br />

corrosion<br />

in environment, 8<br />

in physiologic media, 9<br />

cross-reactivity between palladium<br />

and, 133<br />

decontamination, 84–85<br />

deficiency, 101, 163, 255<br />

alimentary regimen, 181<br />

experimental animals study, 163–170<br />

feeding, 163<br />

organism, 75, 76<br />

dental alloys, released from, 100<br />

in dental materials, 124<br />

depletion, proinflammatory effect<br />

of, 163<br />

deprivation, 163<br />

derivatives, used as pesticides, 118<br />

dermal irritation by, 111<br />

in <strong>dermatology</strong>, 118<br />

dermatopharmacokinetics, 75<br />

detection of limits for, 81–82<br />

diffusible compounds, 118<br />

diffusion, 67, 69, 81, 83, 269, 288<br />

diffusivity, 269, 270<br />

electron configuration and reactivity<br />

of, 8<br />

[Copper]<br />

electron microscopy for, 270<br />

electron probe analysis for, 270<br />

electrophilic reactivity of, 271<br />

endogenous, 102<br />

epicutaneous tests for, 135<br />

exogenous, 103, 277<br />

effects of, 91<br />

exposure, 97<br />

types, 125<br />

formation of, 99, 255<br />

and hemolytic anemia, 101<br />

in human organism, 14<br />

human stratum corneum penetration<br />

by, 81<br />

hypersensitivity, 115, 132<br />

immune reactions<br />

case reports, 138<br />

individual reports, 138<br />

reports of, 115–116<br />

immune response, 125<br />

as immunogen, 117<br />

immunogenic potential of, 123<br />

immunologic contact urticaria<br />

due to, 127<br />

in intrauterine devices (IUDs), 100,<br />

115, 125<br />

intravenous administration<br />

of, 186–188<br />

ionic, 243<br />

irritation<br />

epidemiology of, 100<br />

incidence of, 100<br />

in skin and mucosa, 103<br />

in jewelry, 82, 98<br />

machinability of, 3<br />

mechanical properties of, 3<br />

properties of, 2–4<br />

thermal and electrical conductivity<br />

of, 3<br />

types of, 6<br />

wrought, 3<br />

leaching of, 10<br />

LG-1/ANA 68, 105<br />

LM-hard gold alloy, 104<br />

in mammalian organism, 275<br />

in medicine, 243<br />

application, 244


298 Index<br />

[Copper]<br />

development of, 240<br />

metabolism disorder, 101<br />

metal<br />

dissolution, 82<br />

solubilization, 98<br />

in metallic bonding, 8<br />

metallurgy of, 117<br />

micro/ANA 68, 104<br />

midi/ANA 68, 105<br />

noxious effects on, 91<br />

occlusion, 84–85<br />

oleate, in vivo application of, 118<br />

oral administration of, 191–198<br />

oxidases, blue, 186<br />

oxidation<br />

in body fluids, 117<br />

states for, 8<br />

oxidation potential of nickel and, 90<br />

parameters for, 270<br />

patch test material, 133<br />

penetration<br />

under occlusion, 87–88<br />

under semiocclusion, 88–89<br />

PGE2 released, 109–110<br />

pharmaceutical, 282<br />

pharmacological activity of, 280<br />

pharmacology of, 155<br />

physiological factors for, 267<br />

plasma, 268<br />

polystyrene, 104<br />

population-based studies, 134–140<br />

powder, 84<br />

predictive immunology test<br />

results for, 119<br />

properties of, 1–2, 83, 240<br />

protein, multifunctional, 158<br />

quantitative data for, 102<br />

radioactivity of, 282<br />

removal by sequential tape stripping,<br />

86–87<br />

salicylate gel, 281<br />

salicylate–ethanol complex, 282<br />

scope and limitations of, 270<br />

sensitivity, 126<br />

sensitization potential of, 115<br />

as sensitizer, 117<br />

skin contact with, 83, 269<br />

[Copper]<br />

skin penetration by, 84, 86, 269<br />

skin reactions to, 82<br />

skin-diffusible, 279<br />

sources of exposure to, 9<br />

status<br />

in blood and urine, 170–173<br />

in solid tissues and inflamed areas,<br />

173–179<br />

stems, 97<br />

stripping, 84–85<br />

in synthetic sweat, 99<br />

systemic allergic contact dermatitis<br />

due to, 129–130<br />

systemic lupus erythematosus disease<br />

activity in, 254<br />

T-cell-mediated reactions to, 123<br />

as therapeutic agent, 238<br />

therapeutic uses of, 240<br />

in therapy, 150<br />

threshold limit values applicable to, 98<br />

toxicity, 72<br />

and therapeutic indices of, 91–92<br />

toxicological considerations of, 91<br />

transdermal anti-inflammatory action<br />

of, 102–103<br />

transdermal therapy by, 278, 279<br />

urticaria, screening procedure, 131<br />

utilization, 267<br />

in vitro assays, 109<br />

in vivo assays<br />

agarose overlay test in, 106<br />

comparison of the different<br />

tests in, 108<br />

erythrocyte lysis assay of, 107<br />

implantation test of, 103–105<br />

toxicity test using murine<br />

macrophages in, 107<br />

Copper metabolism, studies on<br />

endogenous, in acute chronic<br />

inflammation, 170–179, 179–184<br />

Copper sulfate<br />

diffusion of, 72<br />

pharmacological use of, 153<br />

Copper-dependent enzymes, 186, 211<br />

lysyl oxidase, 199<br />

role of, 206<br />

Copper–nickel alloys, 6


Index 299<br />

Copper/zinc bracelets, study of, 249<br />

Corneocytes, 23<br />

Corpus hippocraticum, 154<br />

Corrosion-resistant materials, 3<br />

Corticosteroids, 274<br />

CP. See Carrageenan-induced pleurisy<br />

CPE. See Carrageenan-induced paw<br />

edema<br />

Cranial trepanation procedure, 153<br />

Cupric ion, testing of, 119<br />

Cupriphores<br />

effective transdermal, 282<br />

subcutaneous administration of, 281<br />

Cutaneous treatment, efficacy of, 199<br />

Cyclo-oxygenase (COX)<br />

activities, 205<br />

pathways, 205–206<br />

Cytochrome-c oxidase activity, 183<br />

Cytolysis, degree of, 106<br />

Delayed contact stomatitis, 124<br />

Dental alloy contact dermatitis, 124<br />

Dental materials<br />

copper alloys used in, 124<br />

copper sensitivity induced by, 126<br />

Dermal absorption experiments,<br />

22–27, 75<br />

approaches for, 25<br />

descriptors of, 25<br />

percent of, 26<br />

permeability coefficient of, 25–26<br />

Dermatitis<br />

dental alloy contact, 124<br />

systemic contact, 117<br />

Dermatome, use of, 44<br />

Dermatopharmacokinetics, 26<br />

Dermatosis, 116<br />

Dermatotoxicology, 26<br />

Dermcusal 1 , 200<br />

Dermis, 22<br />

Diagnostic equivocation, 133<br />

Diet, semiquantitative assessment of, 248<br />

Dietary copper deficiency, effects of, 168<br />

Diffusion<br />

characteristics of, 76<br />

in copper, 269, 270<br />

experiments, in vitro, 76<br />

[Diffusion]<br />

Fick’s law of, 25<br />

Kalia’s equation, 23<br />

membrane analysis, 76<br />

passive, 75<br />

rates of, 68<br />

Dimethyl acetamide, 37<br />

Dimethyl formamide, 37<br />

Dimethyl mercury, 22<br />

Dimethylsulfoxide (DMSO), 37<br />

applications of, 122<br />

skin penetration by, 122<br />

as vehicle, 122<br />

Divalent oxidation state, 68<br />

D-penicillamine, 162<br />

Drug<br />

delivery systems, 76<br />

intercellular, 284<br />

percutaneous, 283<br />

ionic, 285<br />

therapy, 271<br />

Elastic connective tissue, 24<br />

Electric resistance, 29<br />

Electrolyte<br />

composition, 10<br />

concentration, 11<br />

Electromotive forces and potential, 10<br />

Electron probe analysis, 69<br />

Electron-dense granules, 24<br />

Electrophilic chromic ion [Cr(III)], 39<br />

Electrophilic metals, 31<br />

Electroporation, 286<br />

Electropositivity, 8<br />

Endogenous origin of skin exudates, 33<br />

Energy dispersion X-ray (EDAX)<br />

analysis, disadvantage<br />

of, 105–106<br />

Enterohepatic circulation, 280<br />

Enzyme, 209<br />

lipolytic, 12<br />

as marker, 183<br />

Epidermis, constitution of, 22, 23<br />

Epithelial cells, 23, 69<br />

Erythrocytes, copper concentration, 181<br />

Erythropoiesis, 203<br />

Escherichia coli, 3


300 Index<br />

Essential trace element (ETE), 67<br />

for biological processes, 294<br />

elimination of, 22<br />

Excited skin syndrome, 133<br />

Exogenous copper administration,<br />

effects of, 184–203<br />

Extracellular-matrix<br />

macromolecules, 199<br />

Femur fracture, 174<br />

Fertility-related phenomenon, 15<br />

Fibroblast–keratinocyte cocultures, 109<br />

Fick’s law of diffusion, 25<br />

Fluid fraction, 166<br />

Food and Drug Administration<br />

(FDA), 242<br />

Gastric irritation, 279<br />

Generic therapeutic remedies, 152<br />

GHK-Cu. See Glycyl-L-histidyl-Llysine-Cu(II)<br />

Glomerulonephritis, immune complex<br />

of, 273<br />

Glycyl-L-histidyl-L-lysine-Cu(II)<br />

(GHK-Cu), 198, 199, 206<br />

Gold, 271<br />

antiarthritic drugs, biological effect<br />

of, 272<br />

ring study, 250<br />

therapy<br />

anti-inflammatory properties<br />

for, 272<br />

soluble gold salts for, 272<br />

therapeutic application, 271<br />

Gold sodium thiomalate, 272<br />

intramuscular injection of, 274<br />

pathobiochemical effects of, 274<br />

Gold sulfhydryl compounds, 239<br />

Gold thioglucose, 272<br />

Gold thiosulfate, 272<br />

contact allergy to, 274<br />

modulatory effects on, 273<br />

tolerance for, 274<br />

GPMT<br />

on 20 guinea pigs, 119<br />

as predictor of skin<br />

sensitization, 119<br />

Gyrotory TM water bath model, 85<br />

HaCaT cells, cytotoxic potential<br />

in, 110–111<br />

Hair follicles, 33<br />

Hapten–protein conjugate, 120<br />

Haruspecism, definition of, 152<br />

Hematochemical serum markers, 163<br />

Hemoglobin, synthesis of, 157<br />

Histamine<br />

activity, 203–205<br />

release, 203–205<br />

Histopathology, 105<br />

Homeostasis, 42, 74<br />

Homeostatic mechanisms, 74, 76<br />

Human<br />

abdominal epidermis, 72<br />

inflammatory process, 171<br />

plasma or serum, 118<br />

rheumatoid arthritis, 158, 184<br />

serum albumin (HAS), 120<br />

skeleton, 156<br />

skin<br />

dermatoglyphics, 83<br />

skin features, 12<br />

stratum corneum penetration by<br />

copper, 81<br />

metal analysis of, 85<br />

statistical analysis of, 85<br />

Hypercupremia, 172<br />

Hypersensitivity<br />

asymptomatic contact, 124<br />

copper, 115<br />

copper allergic, 121<br />

population studies of, 132<br />

diagnostic tests for, 119<br />

immediate allergic, 120<br />

open test for, 119<br />

radioallergosorbent test (RAST)<br />

for, 120<br />

ICAMs. See Intercellular adhesion<br />

molecules<br />

ICU. See Immunologic contact<br />

urticaria<br />

IgE antibodies, 120<br />

production of, 124<br />

Immune system development and<br />

reactivity, 212–216


Index 301<br />

Immunologic contact stomatitis, 124<br />

Immunologic contact urticaria (ICU),<br />

115–116, 119<br />

due to copper, 127<br />

See also Allergy, immediate-type<br />

Immunosuppressants, 274<br />

In vivo methods, 47–51<br />

advantages of, 49<br />

approaches for, 48<br />

conclusions of, 50<br />

semiquantitative, 50<br />

skin stripping in, 50<br />

Inductively coupled plasma-mass<br />

spectrometry, 69<br />

Inductively coupled plasma–mass<br />

spectroscopy (ICP-MS)<br />

analysis, 85<br />

Inflammatory arthropathies, 252<br />

Inflammatory reaction, 165, 207<br />

Intercellular adhesion molecules<br />

(ICAMs), 211, 212<br />

Intrathoracic injection, 165<br />

Intrauterine devices (IUDs), 98<br />

Ion, cupric, testing of, 119<br />

Ion exchange chromatography, 11<br />

Ionic copper, release of, 243<br />

Ionic drugs, 285<br />

Iontophoresis, 285<br />

Irritant contact dermatitis (ICD),<br />

incidence of, 101<br />

Irritant chemicals, 98<br />

Kalia’s diffusion equation, 23<br />

Keratinocytes, 23<br />

Lactic dehydrogenase (LDH), 107<br />

Leukocyte<br />

activity, 207–212<br />

migration, 166, 207–212<br />

Lipid bilayers, intercellular, 283<br />

Lipolytic enzymes, 12<br />

Lipophilic compounds, 28<br />

Lipophilic formulations, 282<br />

properties of, 284<br />

Liposomes (LS), 284<br />

biological particulate materials<br />

in, 284<br />

oxidation, 76<br />

particle size of, 284<br />

Local lymph node assay (LLNA),<br />

cuprous chloride in, 119<br />

Mammalian sweat, 11<br />

Membrane transport, 8<br />

Membrane-bounded organelles, 24<br />

Menkes’ syndrome, 101–102<br />

Metabolism, of connective tissue, 206<br />

Metal(s)<br />

corrosion rates between two, 90<br />

oxidation of, 89<br />

Metal detection, 53–56<br />

analytical methods for, 53<br />

atomic absorption spectrophotometry<br />

(AAS) for, 54–55<br />

electron spin resonance (ESR)<br />

for, 55<br />

inductively coupled plasma–atomic<br />

emission spectroscopy<br />

(ICP-AES) technique for, 53<br />

inductively coupled plasma–mass<br />

spectroscopy (ICP-MS)<br />

technique for, 50, 53–54<br />

particle-induced X-ray emission<br />

for, 56<br />

stable-isotope inductively coupled<br />

plasma–mass spectroscopy<br />

technique for, 54<br />

Metal surfaces, sebum and sweat in, 36<br />

Metal–ligand equilibria, computer<br />

simulation of, 279<br />

Metallo-element–deficient animals, 163<br />

Metallothionein, 32<br />

Micronutrients, physiological dynamics<br />

of, 75<br />

Microparticle-induced X-ray emission<br />

(PIXE), 29<br />

Mononuclear leukocytes, activity<br />

of, 207<br />

Musculoskeletal disorders, localized, 280<br />

Mycobacterium butyricum, 168


302 Index<br />

Nephrotic syndrome, 273<br />

Neurotoxic antimicrobial<br />

hexachlorophene (1–3), 22<br />

Nickel<br />

cross-reactivity between copper<br />

and, 133<br />

cross-reactivity between palladium<br />

and, 133<br />

dermatopharmacokinetics of, 50<br />

epicutaneous tests for, 135<br />

ion-specific T-cell clones, 133<br />

reactivity of, 133<br />

Nickel ion–specific T-cell clones, types of<br />

reactivity of, 133<br />

Nickel–silver, 2<br />

alloys, 6<br />

Nitric oxide synthase activity, regulation<br />

of, 206<br />

Nonarthritic copper deficiency, 170<br />

Nonsteroidal anti-inflammatory drugs<br />

(NSAIDs), 187, 188, 191, 200,<br />

238, 268<br />

piroxicam, 210<br />

NSAIDs. See Nonsteroidal antiinflammatory<br />

drugs<br />

Optimal flux, 280<br />

Orbital shells, 68<br />

Osteoarthritis, 249<br />

Patch testing<br />

copper sulfate under, 121<br />

for delayed-type allergy, 120–121<br />

detection of allergens by, 121<br />

diagnostic, 37<br />

optimal solvent in suspected<br />

delayed-type copper allergy,<br />

123, 126<br />

positive reactions, 90<br />

role of vehicle in, 121–122<br />

Periodontal disease, 158<br />

acute, 161<br />

Perkin-Elmer 4300 dual view ICP optical<br />

emission spectrometer, 85<br />

Permeability coefficients, measurement<br />

of, 72<br />

Permeation protocols, in vitro, 280<br />

Petrolatum, 122–123<br />

Pharmacokinetics, 281<br />

principles, 47<br />

Pharmacological therapy, 159<br />

Phorbol miristate acetate<br />

(PMA), 209<br />

Physiotherapy, 275<br />

Pilosebaceous glands, 12<br />

Placebo effect, 247<br />

Plasma copper concentration, 172<br />

Plasma glycoprotein, multifunctional<br />

endogenous, 267<br />

Plasma-membrane barrier, 68<br />

PMNLs. See Polymorphonuclear<br />

(neutrophils) and mononuclear<br />

leukocytes<br />

Polymorphonuclear (neutrophils),<br />

activity of, 207<br />

Polymorphonuclear (neutrophils) and<br />

mononuclear leukocytes<br />

(PMNLs), 207, 209<br />

migration, 210–211<br />

phagocytosis, 207<br />

phagosomes, 208<br />

respiratory burst, 209<br />

Polypropylene tape, 85<br />

Proinflammatory effect, 163<br />

Prostaglandin E2 (PGE2), 109<br />

Prostaglandin-synthetase activity, 169<br />

Provocative use test (PUT), 126<br />

Pseudoscience, survey of, 243<br />

Quantitative cutaneous diffusion<br />

rates, 67<br />

Quantitative structure–activity<br />

relationships, 28<br />

RA. See Rheumatoid arthritis<br />

Radioactivity, 26<br />

Radioallergosorbent test (RAST)<br />

inhibition test, 120


Index 303<br />

Reactive oxygen species (ROS)–induced<br />

reactions, 209<br />

Red blood cell, copper level, 181<br />

Repeat open application test<br />

(ROAT), 126<br />

Rheumatic patients, 243<br />

articular erosion in, 250<br />

Rheumatoid arthritis (RA), 76, 171,<br />

179–184, 268<br />

autoimmune disease, 268<br />

chronic syndrome, 268<br />

copper chelates in, 278<br />

inflammatory disease, 268<br />

patients, 161<br />

spontaneous remission of, 162<br />

symmetric inflammation of, 268<br />

therapy of, 72<br />

traditional and modern therapies<br />

for, 268<br />

treatment of, 238, 250<br />

ROS. See Reactive oxygen species–<br />

induced reactions<br />

‘‘Rusters’’, 11, 34<br />

Sebaceous glands, 12<br />

Sebum, classes of, 12, 36<br />

Serum copper levels, determination<br />

of, 167<br />

Silicon bronzes, 5<br />

Skin<br />

absorption, 22<br />

by different sites of the<br />

body, 42<br />

methods for measuring<br />

percutaneous, 45<br />

afflictions, 243<br />

age of, 42<br />

allergic reactions in, 39<br />

arsenic in, 45<br />

aspects of metals, 23<br />

chemical microenvironment on, 9<br />

chromium applied on, 44<br />

compounds formed by metals in<br />

contact with, 31–35<br />

diffusion of<br />

metal compounds, 35<br />

[Skin]<br />

nonelectrolytes, 28<br />

paths of, 28<br />

discoloration of, 44<br />

electrolytes across, 29–31<br />

principal conditions of, 29<br />

electron micrographs of, 14<br />

function as diffusion barrier, 23<br />

gold in contact with, 90<br />

immune reactions to chemicals<br />

in, 116<br />

as membrane, 22<br />

metabolism (red/ox), 44<br />

nickel<br />

as powder on, 50<br />

in contact with, 90<br />

organic compounds, 28–29<br />

penetration, 21<br />

by DMSO, 122<br />

by nickel chloride, 43<br />

by xenobiotics, 22, 33<br />

permeant categories of, 28<br />

pH effect on, 39<br />

reactions to copper, 82–83<br />

sensitization, 119<br />

structure of, 23<br />

Surface, formation of free acids<br />

on, 99<br />

sweat and sebum, 9, 90<br />

in vitro, in function of polarity, 38<br />

Skin-diffusible derivatives, 90<br />

Skin-diffusible oxidation products, 32<br />

Skin diffusion<br />

endogenous factors for, 41<br />

age of skin as, 42<br />

homeostatic control mechanisms,<br />

43–43<br />

regional variation, 41<br />

exogenous factors for, 35<br />

counterion and molecular<br />

volume, 37<br />

dose, 35–36<br />

nature of chemical bond, 38<br />

solubility, 39<br />

time postapplication, 41<br />

valence, 39<br />

vehicle as, 36–37


304 Index<br />

[Skin diffusion]<br />

polarity determinates for, 38<br />

variables determining, 35<br />

Skin diffusivity rates, 67<br />

Skin prick device, sterile, 120<br />

Skin prick test (SPT), 119<br />

for immediate-type allergy, 119–120<br />

positives, 120<br />

Solubility, in polar media, 280<br />

Sonophoresis, 286<br />

Sorenson’s hypothesis, soundness<br />

of, 191<br />

Staphylococcus aureus, methicillinresistant,<br />

3<br />

Steady-state transport rate, 14<br />

Steroid therapy, 274<br />

Stomatitis<br />

delayed contact, 124<br />

immunologic contact, 124<br />

systemic anaphylactic, 124<br />

Stratum corneum (SC), 23, 98<br />

absorption of copper into, 91<br />

barrier, 286<br />

components of, 24<br />

copper diffusion through, 81,<br />

83, 89<br />

depots in, 90, 40–41<br />

intercellular lipid domains in, 30<br />

occlusion of, 84<br />

protein reactivity, 40–41<br />

stripping, 83<br />

swelling of basal, 116, 122<br />

Student t-test, 85<br />

Superoxide dismutase, 243<br />

anti-inflammatory copper-dependent<br />

metalloenzymes, 274<br />

Superoxide metabolism, regulation<br />

of, 168<br />

Sweat<br />

components, 10, 33<br />

duct, 29<br />

glands, 33<br />

mammalian, 11<br />

mean body, 11<br />

mean levels of, 33<br />

proteins and amino acids in, 34<br />

Sweating rate, 12<br />

Syndrome<br />

contact urticaria, 116<br />

nephrotic, 273<br />

Systemic anaphylactic stomatitis, 124<br />

Systemic contact dermatitis, 117,<br />

129–130<br />

Talcum powder, formulation of, 22<br />

Therapeutic agent, dissipation of, 281<br />

Therapeutic properties of copper, 150<br />

Therapy<br />

drug, 271<br />

effectiveness of topical, 249<br />

gold, 271<br />

pharmacological, 159<br />

steroid, 274<br />

Thermal stimulation, 12<br />

Thermogenic ointments, 275<br />

Threshold limit values (TLV), 98<br />

Throat inflammation, 155<br />

Tin bronzes, 5<br />

Topical therapy, 279<br />

Total serum copper, increase of, 171<br />

Transdermal<br />

absorption, 285<br />

applications, 288<br />

Transepidermal water loss<br />

(TEWL), 23<br />

Transition metals<br />

in biological systems, 8<br />

characteristic for, 8<br />

Transpore TM tape, 85<br />

Tuberculosis and syphilis, infection<br />

of, 157<br />

Ulcerogenic effect, 276<br />

VAP-1. See Vascular adhesion protein-1<br />

Vascular adhesion molecule-1<br />

(VCAM-1), 211<br />

Vascular adhesion protein-1 (VAP-1),<br />

211, 212<br />

VCAM-1. See Vascular adhesion<br />

molecule-1


Index 305<br />

Wilson’s disease (WD), 101, 162<br />

Wilson’s disease protein (WND), 185<br />

Wounds, treatment of<br />

postoperative, 151<br />

Wrought alloys, 4<br />

Wrought copper alloys, 3<br />

Xenobiotics, Fick’s law applied<br />

to, 22, 25<br />

Zinc absorption, measurement<br />

of, 75

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!