31.05.2015 Views

NcXHF

NcXHF

NcXHF

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.

ANTIBODY-DRUG CONJUGATES AND T-DM1<br />

What Can We Learn about Antibody-Drug Conjugates from the<br />

T-DM1 Experience?<br />

Francisco J. Esteva, MD, PhD, Kathy D. Miller, MD, and Beverly A. Teicher, PhD<br />

OVERVIEW<br />

Antibody conjugates are a diverse class of therapeutics that consist of a cytotoxic agent linked covalently to an antibody or<br />

antibody fragment directed toward a specific cell surface target expressed by tumor cells. The notion that antibodies directed<br />

toward targets on the surface of malignant cells could be used for drug delivery is not new. The history of antibody conjugates<br />

has been marked by hurdles identified and overcome. Early conjugates used mouse antibodies, drugs that either were not<br />

sufficiently potent, were immunogenic (proteins), or were too toxic, and linkers that were not sufficiently stable in circulation.<br />

Four main avenues have been explored using antibodies to target cytotoxic agents to malignant cells: antibody-protein toxin (or<br />

antibody fragment–protein toxin fusion) conjugates, antibody-chelated radionuclide conjugates, antibody-small molecule conjugates,<br />

and antibody-enzyme conjugates administered along with small molecule prodrugs that require metabolism by the<br />

conjugated enzyme to release the activated species. Technology is continuing to evolve regarding the protein and small molecule<br />

components, and it is likely that single chemical entities soon will be the norm for antibody-drug conjugates. Only antibodyradionuclide<br />

conjugates and antibody-drug conjugates have reached the regulatory approval stage, and there are more than 40<br />

antibody conjugates in clinical trials. The time may have come for this technology to become a major contributor to improving<br />

treatment for patients with cancer.<br />

The challenges posed by the development of therapeutic<br />

antibody-drug conjugates (ADCs) are formidable. Over<br />

the past 30 years, many cell surface proteins that have selective<br />

aberrant expression on malignant cells or are aberrantly<br />

highly expressed on the surface of malignant cells have been<br />

identifıed. In many cases, specifıc antibodies that bind tightly<br />

to malignant cell surface proteins were developed. However,<br />

these antibodies often were not active antitumor agents.<br />

ADCs provide an opportunity to make use of antibodies that<br />

are specifıc to cell surface proteins. 1 Successful ADCs have<br />

improved tumor specifıcity and potency compared with traditional<br />

drugs. 2,3 Heterogeneity of antibody target expression<br />

on the tumor surface, and expression of the antigen by normal<br />

tissues, can limit the effectiveness of ADCs. For some<br />

hematologic malignancies, antigen expression is specifıc and<br />

homogeneous.<br />

Early ADCs were composed of tumor-specifıc murine<br />

monoclonal antibodies covalently linked to anticancer drugs,<br />

such as doxorubicin, vinblastine, and methotrexate. These<br />

early conjugates were evaluated in human clinical trials but<br />

had limited success because of immunogenicity, lack of potency,<br />

and insuffıcient selectivity for tumor versus normal<br />

tissue. The lessons learned from these early explorations led<br />

to improvements in all aspects of antibody conjugate therapeutics<br />

and, hence, to renewed interest in ADC technology. 4<br />

Immunogenicity was overcome by replacing murine antibodies<br />

with humanized or fully human antibodies. Potency<br />

was improved by using drugs that were 100- to 1,000-fold<br />

more potent than previously used drugs. Selectivity was addressed<br />

by performing more careful target and antibody selection.<br />

As a result of such improvements, gemtuzumab<br />

ozogamicin (Mylotarg; Pfızer, New York, NY) was granted<br />

accelerated U.S. Food and Drug Administration (FDA) approval<br />

for the treatment of acute myelogenous leukemia in<br />

2000, becoming the fırst commercially available ADC. However,<br />

gemtuzumab ozogamicin was withdrawn from the market<br />

in 2010 because, in postmarketing follow-up clinical<br />

trials, it failed to meet the prospective effıcacy targets. Two<br />

ADCs, trastuzumab emtansine (T-DM1, Kadcyla; Genentech/Roche,<br />

South San Francisco, CA) and brentuximab vedotin<br />

(SGN-35; Adcetris, Seattle Genetics, Seattle, WA;<br />

Millennium/Takeda, Boston, MA), reached FDA approval in<br />

2014 and 2011, respectively, for treatment of metastatic<br />

breast cancer and refractory Hodgkin lymphoma and systemic<br />

anaplastic large cell lymphoma, respectively. More<br />

than 40 ADCs are in clinical trials (Table 1).<br />

From the Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; National Cancer<br />

Institute at the National Institutes of Health, Bethesda, MD.<br />

Disclosures of potential conflicts of interest are found at the end of this article.<br />

Corresponding author: Beverly A. Teicher, PhD, Molecular Pharmacology Branch, National Cancer Institute, 9609 Medical Center Dr., Room 4-W602, MSC 9735, Bethesda, MD 20892;<br />

email: beverly.teicher@nih.gov.<br />

© 2015 by American Society of Clinical Oncology.<br />

asco.org/edbook | 2015 ASCO EDUCATIONAL BOOK<br />

e117

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!