01.01.2015 Views

Annals of Diagnostic Paediatric Pathology

Annals of Diagnostic Paediatric Pathology

Annals of Diagnostic Paediatric Pathology

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.

<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong><br />

Official Journal <strong>of</strong> the Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

and Section <strong>of</strong> Oncological Surgery <strong>of</strong> Polish Association <strong>of</strong> <strong>Paediatric</strong> Surgeons<br />

Editor-in-Chief<br />

Co-Editors<br />

Associate Editors<br />

ProductIon Editor<br />

B. M. WoŸniewicz, Warsaw<br />

B. M. Cukrowska, Warsaw<br />

A. I. Prokurat, Bydgoszcz<br />

J. Cielecka-Kuszyk, Warsaw<br />

E. Czarnowska, Warsaw<br />

W. T. Dura, Warsaw<br />

W. Grajkowska, Warsaw<br />

M. Liebhardt, Warsaw<br />

A. Wasiutyñski, Warsaw<br />

G. Napiórkowski, Warsaw<br />

A. Bysiek, Cracow<br />

P. Czauderna, Gdansk<br />

J. Godziñski, Wroc³aw<br />

J. Niedzielski, Lodz<br />

W. WoŸniak, Warsaw<br />

M. Wysoki, Bydgoszcz<br />

Editorial Office<br />

<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong><br />

Department <strong>of</strong> <strong>Pathology</strong><br />

The Children´s Memorial Health Institute<br />

Aleja Dzieci Polskich 20<br />

04 736 Warszawa, Poland<br />

Tel.: +48-22-815-19-72<br />

Fax: +48-22-815-19-73<br />

E-mail: ptpd@czd.waw.pl<br />

Editorial Board<br />

J. P. Barbet, Paris<br />

L. A. Boccon-Gibod, Paris<br />

P. E. Campbell, Melbourne<br />

A. Chilarski, Lodz<br />

J. Czernik, Wroclaw<br />

E. Gilbert-Baarness, Tampa<br />

A. A. Greco, New York<br />

M. D. Haust, Londyn<br />

A. Hinek, Toronto<br />

J. Huber, Utrecht<br />

C. G. Gopalakrishnam, Trivandrum<br />

S. Gogus, Ankara<br />

A. Jankowski, Poznan<br />

P. Januszewicz, Warsaw<br />

B. Jarz¹b, Gliwice<br />

B. A. Kakulas, Perth<br />

R. O. C. Kaschula, Rondebosch<br />

W. Kawalec, Warsaw<br />

J. Kobos, Lodz<br />

G. Karpati, Montreal<br />

J. Las Heras, Santiago de Chile<br />

K. Madaliñski, Warsaw<br />

D. M. F. Menezes, Rio de Janeiro<br />

W. A. Newton, Jr., Johnstown<br />

B. Otte, Brussels<br />

S. A. Pileri, Bologna<br />

J. Plaschkes, Berne<br />

F. Raafat, Birmingham<br />

S. W. Sadowinski, Mexico City<br />

K. Sawicz-Birkowska, Wroclaw<br />

J. Stejskal, Prague<br />

Cz. Stoba, Gdansk<br />

G. Thiene, Padova<br />

S. Variend, Shieffield<br />

T. H. Wyszyñska, Warsaw<br />

A. Zimmermann, Berne<br />

The journal is supported by the State Committee for Research.<br />

Aims and scope<br />

The <strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> is an international peer-reviewed journal. The focus <strong>of</strong> the journal is current<br />

progress in clinical paediatric pathology and surgical oncology in both basic and clinical applications. Experimental studies and<br />

clinical trials are accepted for publication, as are case reports supported by literature review. The main policy <strong>of</strong> the <strong>Annals</strong> is<br />

to publish papers that present practical knowledge that can be applied by clinicians.<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society, 2001<br />

ISSN 1427-4426


Review<br />

Papers<br />

<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong><br />

Official Journal <strong>of</strong> the Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

and Section <strong>of</strong> Oncological Surgery <strong>of</strong> Polish Association <strong>of</strong> <strong>Paediatric</strong> Surgeons<br />

Volume 8 Number 3-4 Winter 2004<br />

CONTENTS<br />

Genetics and biology <strong>of</strong> neuroblastoma – an overview ....................................................................................69<br />

Peter F. Ambros, Ruth Ladenstein, Helmut Gadner, Ingeborg M. Ambros<br />

Neuroblastoma: histoprognostic classification and handling <strong>of</strong> material for biology<br />

– a challenge for the pathologist .........................................................................................................................73<br />

Klaus Beiske<br />

Original<br />

Papers<br />

Intracranial ependymomas in children: correlation <strong>of</strong> selected factors and event-free<br />

survival in the group <strong>of</strong> 142 consecutive patients treated at one institution. .................................................83<br />

S³awomir Barszcz, Wies³awa Grajkowska, Danuta Perek, Marcin Roszkowski, Monika Drogosiewicz,<br />

Marta Perek-Polnik, El¿bieta Jurkiewicz, Pawe³ Daszkiewicz<br />

<strong>Pathology</strong> <strong>of</strong> fatty liver in children with LCHAD deficiency...........................................................................89<br />

Katarzyna Iwanicka, Janusz Ksi¹¿yk, Monika Pohorecka, Maciej Pronicki<br />

Isolation <strong>of</strong> urothelial cells for tissue engineered bladder augmentation ....................................................... 95<br />

Tomasz Drewa, Przemys³aw Galazka, Zbigniew Wolski, Andrzej I. Prokurat, Jan Sir<br />

Status <strong>of</strong> oral hygiene and dentition in children exposed to anticancer treatment .......................................99<br />

Dorota Olczak-Kowalczyk, Marta Daszkiewicz, Barbara Adamowicz-Klepalska, El¿bieta Czarnowska, Agnieszka Sowiñska,<br />

Bo¿ena Dembowska-Bagiñska, Danuta Perek, Pawe³ Daszkiewicz<br />

Preliminary review <strong>of</strong> the treatment results <strong>of</strong> hepatoblastoma and hepatocarcinoma<br />

in children in Poland in the period 1998-2002 ................................................................................................107<br />

Czes³aw Stoba, Katarzyna Nierzwicka, Piotr Czauderna, Barbara Skoczylas-Stoba , Stefan Popadiuk, Jolanta Bonar,<br />

Krystyna Sawicz-Birkowska, Hanna Kaczmarek-Kanold, Przemys³aw Mañkowski, Krzyszt<strong>of</strong> K¹tski, Wojciech Madziara,<br />

Magdalena Rych³owska, Anna Sitkiewicz, Anna Szo³kiewicz, Sabina Szymik-Kantorowicz, Violetta Œwi¹tkiewicz<br />

Liver transplantation for primary malignant liver tumors in children – single center experience ...........111<br />

Piotr Kaliciñski, Hor Ismail, Andrzej Kamiñski, Danuta Perek, Joanna Paw³owska, Przemys³aw Kluge, Joanna Teisserye,<br />

Marek Szymczak, Tomasz Drewniak, Pawe³ Nachulewicz, Bo¿ena Dembowska-Bagiñska, Marek Stefanowicz,<br />

Andrzej Byszewski, Ma³gorzata Manowska<br />

Hepatocyte ultrastructure in non-hemolytic hyperbilirubinemias ...............................................................115<br />

El¿bieta Czarnowska, Bogdan WoŸniewicz<br />

Congenital Heart Diseases Database: analysis <strong>of</strong> occurrence,<br />

diagnostics and coexistence with anomalies <strong>of</strong> other systems. Single institution experience .....................119<br />

Anna Rybak, Aneta Wójcik, Bogdan WoŸniewicz<br />

Case<br />

Reports<br />

Invited<br />

Lectures<br />

Solid pseudopapillary tumor <strong>of</strong> pancreas. Case report and the review <strong>of</strong> literature ..................................121<br />

Andrzej Chilarski, Anna Piaseczna-Piotrowska, Jan Strzelczyk, Stanis³aw £ukaszek, Andrzej Kulig<br />

Liver tumors <strong>of</strong> childhood - pathology. Report <strong>of</strong> the Kiel Pediatric Tumor Registry................................125<br />

Dieter Harms


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Genetics and biology <strong>of</strong> neuroblastoma – an overview<br />

Peter F. Ambros 1 , Ruth Ladenstein 2 , Helmut Gadner 1, 2 , Ingeborg M. Ambros 1<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

1<br />

CCRI, Children’s Cancer Research Institute,<br />

St. Anna Children’s Hospital,<br />

A-1090 Vienna, Austria<br />

2<br />

St. Anna Children’s Hospital,<br />

A-1090 Vienna, Austria<br />

Abstract<br />

Neuroblastoma, the most common extra cranial solid tumor in early childhood, is a heterogeneous disease<br />

with different clinical and biological behavior. Unique features are the spontaneous regression and maturation<br />

which both require similar genetic prerequisites and, in the case <strong>of</strong> maturation, also the presence <strong>of</strong> the<br />

non-neoplastic Schwann cells. However, over the half <strong>of</strong> the tumors display an aggressive clinical course.<br />

Besides the stage and age at diagnosis, the MYCN oncogene (amplified versus non-amplified) has gained<br />

fundamental importance for therapy stratification, particularly in patients with localized disease and in infants.<br />

Further prognostic markers associated with an unfavorable outcome are DNA di-tetraploidy, deletion<br />

at chromosomal regions 1p36, 3p and 11q and gain <strong>of</strong> 17q, besides others. Most <strong>of</strong> these genetic markers<br />

have been shown to predict outcome in patients with localized disease while their impact in stage 4 patients<br />

is discussed controversially. Recent data indicate that not even MYCN amplification can separate prognostic<br />

subgroups in the latter patient group. However, a rapid BM-clearing seems to be associated with a far more<br />

favorable outcome even in the group <strong>of</strong> patients with unfavorable genetic features, thus expressing the response<br />

to chemotherapy and possibly providing an excellent prognostic marker. Furthermore, recent gene<br />

expression studies highlighting the whole transcriptome may provide also valuable information on the dignity<br />

<strong>of</strong> neuroblastic tumors. We can conclude that MYCN amplification still represents an excellent way to<br />

discriminate aggressive from non- or less aggressive tumors especially in patients with localized disease or<br />

infants. However, new biological and genetic information is necessary to be able to reach our final goal<br />

which is patient tailored therapy based on the genetic and biological pr<strong>of</strong>ile <strong>of</strong> the tumor.<br />

Key words: genetics, MYCN, oncogene<br />

Genetic hallmarks in neuroblastic tumors<br />

Over the last few years the amplification <strong>of</strong> the MYCN<br />

oncogene [11, 32] has taken shape as the most important<br />

marker for the assessment <strong>of</strong> the dignity <strong>of</strong> neuroblastic tumors.<br />

Amplifications <strong>of</strong> this oncogene can be observed in approximately<br />

20% <strong>of</strong> tumors and are <strong>of</strong>ten associated with a<br />

deletion at the short arm <strong>of</strong> chromosome 1, del1p36 [10]. Both<br />

aberrations can be found in both genetic subgroups (Fig. 1)<br />

which are based on the different DNA-content (di-/tetraploid<br />

versus near-triploid or penta-/hexaploid). What up to now is<br />

entirely unclear is why MYCN amplifications and/or aberrations<br />

<strong>of</strong> the chromosomal region 1p36.3 occur in only about<br />

15% <strong>of</strong> the near-triploid neuroblastic tumors while the same<br />

aberrations can be found in approx. 56% <strong>of</strong> the diploid group<br />

(unpublished data). The genetic aberration which is most frequently<br />

detected in neuroblastic tumors is gain <strong>of</strong> the long<br />

arm <strong>of</strong> chromosome 17 (in 66-69% <strong>of</strong> all cases) [9, 20, 39].<br />

The prognostic value <strong>of</strong> this aberration has however not yet<br />

been completely clarified and was already opposed [36]. Also<br />

other chromosome aberrations like del11q23 [13, 15, 22, 35,<br />

40] and del3p26 [13, 40] have been described frequently.<br />

Whereby, loss <strong>of</strong> sequences at 11q may indicate a rather promising<br />

genetic marker especially in MYCN non amplified tumors.<br />

However, one has to keep in mind that genetic/biological<br />

studies will only give clear results when the material to be<br />

tested is <strong>of</strong> good quality and is present in sufficient amount.<br />

Address for correspondence<br />

Assoc. Pr<strong>of</strong>. Dr. Peter F. Ambros<br />

CCRI, Children's Cancer Research Institute, St. Anna Kinderspital<br />

Kinderspitalgasse 6<br />

A-1090 Vienna, Austria<br />

Phone: +43 1 40470 411<br />

Fax: +431 408 7230<br />

E-mail: ambros@ccri.univie.ac.at


70<br />

Therefore, the sampling and storage <strong>of</strong> tumor, blood and bone<br />

marrow should be done in the appropriate way to guarantee<br />

the highest possible quality <strong>of</strong> the results [5]. Furthermore,<br />

quality assessment studies and a uniform nomenclature to<br />

describe the results <strong>of</strong> the genetic/biological studies provide<br />

additional security on the high quality <strong>of</strong> the results [1].<br />

Subgroups <strong>of</strong> neuroblastic tumors based on different<br />

DNA-content<br />

Based on a difference in DNA-content, neuroblastic tumors<br />

can be divided into two major groups (Fig. 1). Slightly more<br />

than half <strong>of</strong> the tumors (55%) have a near-triploid DNA-content<br />

(DNA-index between 1.26 and 1.74) while in approx. 45%<br />

<strong>of</strong> neuroblastic tumors a diploid (or tetraploid or even combined<br />

diploid/tetraploid) DNA content can be observed. Spontaneous<br />

regression or maturation, not triggered by therapy,<br />

and which can <strong>of</strong>ten be found in neuroblastic tumors, is very<br />

likely limited to the near-triploid subgroup [2, 7]. In approx.<br />

15% <strong>of</strong> the cases, however, aggressive tumor behavior can<br />

also be observed in this subgroup (Ambros, unpublished). In<br />

these cases structural chromosome aberrations (MYCN amplification,<br />

1p-deletion) may be found. Most <strong>of</strong> the diploid<br />

tumors, on the other hand, are aggressive even without MYCN<br />

amplification or 1p36.3 aberration, irrespective <strong>of</strong> the patient’s<br />

age at diagnosis or the stage <strong>of</strong> the tumor [19, 21]. Alterations<br />

<strong>of</strong> the genome can be observed in almost all diploid tumors.<br />

Genesis <strong>of</strong> neuroblastic tumors and genetic intratumoral<br />

heterogeneity (focal MYCN amplification)<br />

When focusing on the two different ploidy groups, diploid<br />

versus near-triploid neuroblastic tumors, we can identify different<br />

frequency <strong>of</strong> structural chromosomal aberrations (i.e.<br />

deletions, gains or even amplifications). The majority <strong>of</strong> neartriploid<br />

tumors exclusively have only numerical chromosome<br />

aberrations – which seems to be sufficient for (<strong>of</strong>ten self-limited)<br />

tumor growth. Succeeding secondary changes, as for<br />

example MYCN-amplification or 1p36.3 aberration, lead, on<br />

the other hand, to malignant transformations (Fig. 1). As regards<br />

the diploid (2n) tumor group (without gain <strong>of</strong> additional<br />

chromosomes), the initiating event has not been identified so<br />

far, even though other chromosome aberrations like del11q23,<br />

del3p26 and 17q-gain have been described frequently [10].<br />

What all these diploid tumors have in common, however, is<br />

their aggressive cell growth (Fig.1).<br />

3n<br />

trkA<br />

expression<br />

3n<br />

no NGF<br />

Schwann cell<br />

regression<br />

maturation<br />

(GNB, GN)<br />

neuroblasts<br />

del1p, MNA<br />

progression<br />

2n<br />

LOH 3p, 11q, etc.<br />

progression<br />

Fig. 1 Spontaneous regression and spontaneous maturation are restricted to those tumors with a near triploid (+ 3n) or near pentaploid/hexaploid (+5n/6n)<br />

DNA content. In tumors <strong>of</strong> patients under 1 year <strong>of</strong> age, lack <strong>of</strong> nerve growth factor (NGF) is a common finding [18, 27] and is probably involved in the<br />

phenomenon <strong>of</strong> spontaneous regression. Tumors undergoing spontaneous maturation processes, on the other hand, occur in patients over 1 year <strong>of</strong> age and are<br />

characterized by the presence <strong>of</strong> a reactive, i.e. non neoplastic cell population, namely the Schwann cells, leading to a mixed population <strong>of</strong> near triploid<br />

(penta-, hexaploid) neuroblastic/ganglionic cells and diploid Schwann cells [2]. The tumor cells in both groups are usually characterized by whole chromosome<br />

gains, structural aberrations are found only occasionally. However, aggressive neuroblastomas frequently display diploid tumor cells, which can either<br />

show amplification <strong>of</strong> the MYCN oncogene and/or deletions at the short arm <strong>of</strong> chromosome 1, or display losses at the chromosomal regions 3p, 11q, 14q or<br />

other aberrations and high telomerase activity. 17q gain was found in both groups. Interestingly, also triploid tumors can acquire MYCN amplifications or 1p<br />

deletions, or other aberrations thus transforming this benign tumor into a highly aggressive one (dotted line). (MNA = MYCN amplification, LOH = loss <strong>of</strong><br />

heterozygosity, NGF = nerve growth factor).<br />

Figure modified from the article: 'Neuroblastom' by Peter F. Ambros and Inge M. Ambros in Medizinische Genetik 2:119-124 (2002)


71<br />

Until recently, it has been assumed that the individual neuroblastic<br />

tumors are genetically homogenous and that for this<br />

reason the investigation <strong>of</strong> a very small area <strong>of</strong> the tumor (e.g.<br />

biopsy) is genetically representative <strong>of</strong> the entire tumor. However,<br />

various facts led to quite different results. New and more<br />

sensitive techniques, i.e. serial genetic investigations <strong>of</strong> individual<br />

tumors used according to therapy-protocols and neuroblastoma<br />

screening, discovered also genetically in-homogenous<br />

tumors. Thus it could be shown that in up to 15%<br />

<strong>of</strong> all MYCN-amplified tumors the MYCN-amplification is<br />

focal, i.e. not ubiquitously detectable in the tumor tissue [4].<br />

The discovery <strong>of</strong> the focal MYCN-amplification implies that<br />

this genetic change occurs only in the course <strong>of</strong> tumor progression<br />

– at least with some tumors – and causes, at least<br />

temporarily ,heterogeneous’ cell populations within a tumor.<br />

It may furthermore be assumed that the amplified cells “overgrow”<br />

the non-amplified cells because <strong>of</strong> their advanced proliferation<br />

rate. These observations regarding heterogeneous<br />

tumors may be unexpected for neuroblastic tumors, but are<br />

not at all unusual in other tumor types in which amplified and<br />

non-amplified areas can be found side by side.<br />

Since the first time that the familial neuroblastomas<br />

have been described in the middle <strong>of</strong> the last century, only a<br />

very few cases have been investigated and the involvement <strong>of</strong><br />

the chromosomal region 16p seems to be important [24].<br />

Spontaneous tumor regression<br />

Complete spontaneous regression <strong>of</strong> tumors (even <strong>of</strong> “metastasized”<br />

tumors) without cytotoxic treatment is a phenomenon<br />

which scientists have not yet been able to explain. And even<br />

though spontaneous regression most <strong>of</strong>ten occurs in 4s stage<br />

tumors [12, 14], it is by no means limited to this stage. It can<br />

be observed in localised tumors [41] in 6 month old patients<br />

who have been diagnosed through the urinary mass screening<br />

and even in “classical” stage 4 tumors in the first year <strong>of</strong> life.<br />

In tumors which have the ability to regress spontaneously,<br />

near-triploidy (or polyploidy) was found but no MYCN-amplification<br />

or 1p-deletion [7] (Fig.1) was detected. The latter<br />

two genetic aberrations represent markers <strong>of</strong> aggressive tumor<br />

behavior. Moreover, the telomerase activity in aggressive<br />

neuroblastomas is increased as is the case in many other human<br />

neoplasias [16, 29]. 4s tumors and spontaneously regressing<br />

tumors however had low or absent telomerase activity.<br />

The only exceptions are patients with a stage 4s tumor and<br />

lethal outcome who did actually show an increase in telomerase<br />

activity [16, 29].<br />

Spontaneous tumor maturation<br />

Spontaneous tumor maturation (which is also not induced by<br />

cytotoxic treatment) does not occur in patients younger than<br />

12 months but is observed in patients over one year <strong>of</strong> age.<br />

Fully matured tumors, i.e. ganglioneuromas, are usually only<br />

diagnosed in patients over 3 or 4 years <strong>of</strong> age. The genetic<br />

change consists <strong>of</strong> triploidization <strong>of</strong> the genome (or penta- or<br />

hexaploidization respectively) and a lack <strong>of</strong> chromosome 1p36<br />

and MYCN-amplification [2]. In all ganglioneuroblastomas<br />

and ganglioneuromas a diploid cell population can be found<br />

besides a triploid cell population. Via in situ-hybridization it<br />

could then be shown that this diploid cell population is made<br />

up <strong>of</strong> Schwann cells which usually occur in mature tumors<br />

[2]. Until then, these Schwann cells, the amount <strong>of</strong> which increases<br />

during maturation, were considered to be tumor cells<br />

(like the ganglionic cells in the tumor) which led to the opinion<br />

that neuroblastic tumors evolve from “pluripotent” neural<br />

crest cell [37]. It is however, quite unlikely that a triploid neuroblast<br />

differentiates into a triploid ganglionic cell and a diploid<br />

Schwann cell. According to the current model [3], it is<br />

the neuroblastoma cell which, in the course <strong>of</strong> their cellular<br />

differentiation process, start to express chemotactic, mitogenic<br />

and also differentiation-inducing factors and thus “attract”<br />

Schwann cells, prompting them to proliferate and differentiate<br />

[23, 25, 26]. In addition, Schwann cells have an<br />

antiproliferative effect on NBs while at the same time stimulating<br />

differentiation [3, 4, 30, 34]. These findings confirm<br />

the central role <strong>of</strong> Schwann cells in the maturation process.<br />

Prognostic impact <strong>of</strong> bone marrow clearing in stage 4<br />

patients<br />

Different approaches were used so far to investigate the prognostic<br />

value <strong>of</strong> tumor cell clearing in neuroblastoma patients.<br />

The techniques mainly used are based on immunological detection<br />

<strong>of</strong> GD2 stained cells using different detection systems.<br />

They range from conventional immunohistochemical detection<br />

assays [33], fluorescence microscopy [31] and a combined<br />

approach using fluorescence detection <strong>of</strong> the immunological<br />

target and subsequent FISH <strong>of</strong> unclear results [1,<br />

Modritz et al. in preparation]. Furthermore, RT-PCR was used<br />

to detect TH mRNA [17]. The different reports imply a similarity<br />

to the data in leukemia with their correlation between<br />

rapid bone marrow clearing and an increased probability <strong>of</strong><br />

survival [28, 38]. However, we have to keep in mind that the<br />

ideal time point to obtain insights into the dynamics <strong>of</strong> the<br />

disease has to be determined in large multi-centre studies.<br />

References<br />

1. Ambros IM, Benard J, Boavida M, et al<br />

(2003) Quality assessment <strong>of</strong> genetic<br />

markers used for therapy stratification.<br />

J Clin Oncol 21: 2077-2084<br />

2. Ambros IM, Zellner A, Roald B, et al<br />

(1996) Role <strong>of</strong> ploidy, chromosome 1p,<br />

and Schwann cells in the maturation <strong>of</strong><br />

neuroblastoma [see comments]. N Engl<br />

J Med 334:1505-1511<br />

3. Ambros IM, Ambros PF (2000) The role<br />

<strong>of</strong> Schwann cells in neuroblastoma. In:<br />

Brodeur GM, Sawada T, Tsuchida Y,<br />

Voute PA (eds) Neuroblastoma, Elsevier,<br />

Amsterdam, The Netherlands, pp 229-<br />

239


4. Ambros IM, Attarbaschi A, Rumpler, S,<br />

Luegmayr A, Turk<strong>of</strong> E, Gadner H, Ambros<br />

PF (2001): Neuroblastoma cells provoke<br />

Schwann cell proliferation in vitro. Med<br />

Pediatr Oncol 36:163-168<br />

5. Ambros PF, Ambros IM (2001) <strong>Pathology</strong><br />

and biology guidelines for resectable and<br />

unresectable neuroblastic tumors and bone<br />

marrow examination guidelines. Med<br />

Pediatr Oncol 37:492-504<br />

6. Ambros PF, Ambros IM, Kerbl R, et al<br />

(2001): Intratumoural heterogeneity <strong>of</strong> 1p<br />

deletions and MYCN amplification in neuroblastomas.<br />

Med Pediatr Oncol 36:1-4<br />

7. Ambros PF, Ambros IM, Strehl S, et al<br />

(1995) Regression and progression in<br />

neuroblastoma. Does genetics predict<br />

tumour behaviour Eur J Cancer<br />

31A:510-515<br />

8. Ambros P F, Mehes G, Ambros I M,<br />

Ladenstein R (2003) Disseminated tumor<br />

cells in the bone marrow - chances and<br />

consequences <strong>of</strong> microscopical detection<br />

methods. Cancer Lett 197:29-34<br />

9. Bown, N, Cotterill S, Lastowska M, et<br />

al (1999) Gain <strong>of</strong> chromosome arm 17q<br />

and adverse outcome in patients with<br />

neuroblastoma [see comments]. N Engl<br />

J Med 340:1954-1961<br />

10. Brodeur G M, Ambros PF (2000) Genetic<br />

and biological markers <strong>of</strong> prognosis in<br />

neuroblastoma. In: Brodeur GM, Sawada<br />

T, Tsuchida Y, Voute PA (eds) Neuroblastoma,<br />

Elsevier, Amsterdam, The Netherlands,<br />

pp 355-365<br />

11. Brodeur GM, Seeger RC, Schwab M,<br />

Varmus HE, Bishop JM (1984) Amplification<br />

<strong>of</strong> N-myc in untreated human neuroblastomas<br />

correlates with advanced disease<br />

stage. Science 224:1121-1124<br />

12. D’Angio GJ, Evans AE, Koop CE<br />

(1971) Special pattern <strong>of</strong> widespread<br />

neuroblastoma with a favourable prognosis.<br />

Lancet 1:1046-1049<br />

13. Ejeskar K, Aburatani H, Abrahamsson<br />

J, Kogner P, Martinsson T (1998) Loss<br />

<strong>of</strong> heterozygosity <strong>of</strong> 3p markers in neuroblastoma<br />

tumours implicate a tumoursuppressor<br />

locus distal to the FHIT gene.<br />

Br J Cancer 77:1787-1791<br />

14. Evans AE, Chatten J, D’Angio GJ,<br />

Gerson JM, Robinson J, Schnaufer L<br />

(1980) A review <strong>of</strong> 17 IV-S neuroblastoma<br />

patients at the Children’s Hospital<br />

<strong>of</strong> Philadelphia. Cancer 45: 833-839<br />

15. Guo C, White PS, Weiss MJ, Hogarty<br />

MD, et al (1999) Allelic deletion at<br />

11q23 is common in MYCN single copy<br />

neuroblastomas. Oncogene 18:4948-<br />

4957<br />

16. Hiyama E, Hiyama K, Ohtsu K,<br />

Yamaoka H, Ichikawa T, Shay JW,<br />

Yokoyama T (1997) Telomerase activity<br />

in neuroblastoma: is it a prognostic<br />

indicator <strong>of</strong> clinical behaviour Eur J<br />

Cancer 33:1932-1936<br />

17. Horibe K, Fukuda M, Miyajima Y,<br />

Matsumoto K, Kondo M, Inaba J,<br />

Miyashita Y (2001) Outcome prediction<br />

by molecular detection <strong>of</strong> minimal residual<br />

disease in bone marrow for advanced<br />

neuroblastoma. Med Pediatr<br />

Oncol 36:203-204<br />

18. Kogner P, et al (1994) Trk mRNA and<br />

low affinity nerve growth factor receptor<br />

mRNA expression and triploid DNA content<br />

in favorable neuroblastoma tumors.<br />

Prog Clin Biol Res 385:137-145<br />

19. Ladenstein R, Ambros IM, Potschger U,<br />

et al (2001) Prognostic significance <strong>of</strong><br />

DNA di-tetraploidy in neuroblastoma.<br />

Med Pediatr Oncol 36:83-92<br />

20. Lastowska M, Cotterill S, Bown N, et<br />

al (2002) Breakpoint position on 17q<br />

identifies the most aggressive neuroblastoma<br />

tumors. Genes Chromosomes Cancer<br />

34:428-36<br />

21. Look AT, Hayes FA, Shuster JJ, et al.<br />

(1991) Clinical relevance <strong>of</strong> tumor cell<br />

ploidy and N-myc gene amplification in<br />

childhood neuroblastoma: a Pediatric<br />

Oncology Group study. J Clin Oncol<br />

9:581-591<br />

22. Luttikhuis ME, Powell JE, Rees SA, et<br />

al (2001) Neuroblastomas with chromosome<br />

11q loss and single copy MYCN<br />

comprise a biologically distinct group<br />

<strong>of</strong> tumours with adverse prognosis. Br J<br />

Cancer 85:531-537<br />

23. Marchionni MA, Goodearl AD, Chen<br />

MS, et al (1993) Glial growth factors<br />

are alternatively spliced erbB2 ligands<br />

expressed in the nervous system [see<br />

comments]. Nature 362:312-318<br />

24. Maris JM, Weiss MJ, Mosse Y, et al<br />

(2002) Evidence for a hereditary neuroblastoma<br />

predisposition locus at chromosome<br />

16p12-13. Cancer Res<br />

62:6651-6658<br />

25. Minghetti L, Goodearl AD, Mistry K,<br />

Stroobant P (1996) Glial growth factors<br />

I-III are specific mitogens for glial cells.<br />

J Neurosci Res 43:684-693<br />

26. Morrissey TK, Levi AD, Nuijens A,<br />

Sliwkowski, MX, Bunge RP (1995)<br />

Axon-induced mitogenesis <strong>of</strong> human<br />

Schwann cells involves heregulin and<br />

p185erbB2. Proc Natl Acad Sci USA<br />

92:1431-1435<br />

27. Nakagawara A, et al (1993) Association<br />

between high levels <strong>of</strong> expression <strong>of</strong> the<br />

TRK gene and favorable outcome in<br />

human neuroblastoma. N Engl J Med<br />

328:847-854<br />

28. Panzer-Grumayer ER, Schneider M,<br />

Panzer S, Fasching K, Gadner H (2000)<br />

Rapid molecular response during early<br />

induction chemotherapy predicts a good<br />

outcome in childhood acute lymphoblastic<br />

leukemia. Blood 95:790-794<br />

29. Poremba C, Willenbring H, Hero B, et<br />

al (1999) Telomerase activity distinguishes<br />

between neuroblastomas with<br />

good and poor prognosis [In Process Citation].<br />

Ann Oncol 10:715-721<br />

30. Reynolds ML, Woolf CJ (1993) Reciprocal<br />

Schwann cell-axon interactions.<br />

Curr Opin Neurobiol 3:683-693<br />

31. Saarinen UM, Wikstrom S, Makipernaa<br />

A, et al (1996 In vivo purging <strong>of</strong> bone<br />

marrow in children with poor-risk neuroblastoma<br />

for marrow collection and<br />

autologous bone marrow transplantation.<br />

J Clin Oncol 14:2791-2802<br />

32. Seeger RC, Brodeur GM, Sather H,<br />

Dalton A, Siegel SE, Wong KY,<br />

Hammond D (1985) Association <strong>of</strong><br />

multiple copies <strong>of</strong> the N-myc oncogene<br />

with rapid progression <strong>of</strong> neuroblastomas.<br />

N Engl J Med 313:1111-1116<br />

33. Seeger RC, Reynolds CP, Gallego R,<br />

Stram DO, Gerbing RB, Matthay KK<br />

(2000) Quantitative tumor cell content<br />

<strong>of</strong> bone marrow and blood as a predictor<br />

<strong>of</strong> outcome in stage IV neuroblastoma:<br />

a Children’s Cancer Group Study.<br />

J Clin Oncol 18:4067-4076<br />

34. Snider WD (1994) Functions <strong>of</strong> the<br />

neurotrophins during nervous system<br />

development: what the knockouts are<br />

teaching us. Cell 77:627-638<br />

35. Spitz R, Hero B, Ernestus K, Berthold<br />

F (2003) Deletions in chromosome arms<br />

3p and 11q are new prognostic markers<br />

in localized and 4s neuroblastoma. Clin<br />

Cancer Res 9:52-58<br />

36. Spitz R, Hero B, Ernestus K, Berthold<br />

F (2003) Gain <strong>of</strong> distal chromosome arm<br />

17q is not associated with poor prognosis<br />

in neuroblastoma. Clin Cancer Res<br />

9:4835-4840<br />

37. Tsokos M, Scarpa S, Ross RA, Triche TJ<br />

(1987) Differentiation <strong>of</strong> human neuroblastoma<br />

recapitulates neural crest development.<br />

Study <strong>of</strong> morphology, neurotransmitter<br />

enzymes, and extracellular matrix proteins.<br />

Am J Pathol 128:484-96<br />

38. van Dongen JJ, Seriu T, Panzer-<br />

Grumayer ER, et al (1998) Prognostic<br />

value <strong>of</strong> minimal residual disease in<br />

acute lymphoblastic leukaemia in childhood.<br />

Lancet 352:1731-1738<br />

39. van Roy N, Laureys G, Van, GM, et al<br />

(1997) Analysis <strong>of</strong> 1;17 translocation<br />

breakpoints in neuroblastoma: implications<br />

for mapping <strong>of</strong> neuroblastoma<br />

genes. Eur J Cancer 33:1974-1978<br />

40. Vandesompele J, Van RN, Van GM, et<br />

al (1998) Genetic heterogeneity <strong>of</strong> neuroblastoma<br />

studied by comparative genomic<br />

hybridization. Genes Chromosomes<br />

Cancer 23:141-152<br />

41. Yamamoto K, Hanada R, Kikuchi A, et al<br />

(1998) Spontaneous regression <strong>of</strong> localized<br />

neuroblastoma detected by mass<br />

screening. J Clin Oncol 16:1265-1269


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Neuroblastoma: histoprognostic classification and handling <strong>of</strong><br />

material for biology – a challenge for the pathologist<br />

Klaus Beiske<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

Department <strong>of</strong> <strong>Pathology</strong>,<br />

Rikshospitalet,<br />

Oslo, Norway<br />

Abstract<br />

Peripheral neuroblastic tumors originate from immature elements <strong>of</strong> the sympathetic nerve system and are<br />

characterized by a striking morphological, biological and clinical heterogeneity. For about 100 years, pathologists<br />

could not reach an agreement on a comprehensible and prognostic significant classification system.<br />

This paper summarizes the contributions made by Hiroyuki Shimada, who proposed to link morphological<br />

differentiation and cellular turn-over (measured as the number <strong>of</strong> mitotic and karyorrhectic nuclei) to<br />

the patients age, and describes the further development <strong>of</strong> his system into the International Neuroblastoma<br />

<strong>Pathology</strong> Classification (INPC) <strong>of</strong> 1999 with a recent revision <strong>of</strong> 2003. Pathologists play now a key role in<br />

the handling <strong>of</strong> neuroblastic tumors based on macroscopic inspection. They are responsible for an adequate<br />

sampling and saving <strong>of</strong> material for morphological, biological and genetic investigations, while the results <strong>of</strong><br />

their microscopic assessment create the basis for a histoprognostic categorization beyond the subsets which<br />

presently are defined by biological/genetical markers.<br />

Key words: classification, genetical markers, neuroblastoma, pathology<br />

Introduction<br />

Neuroblastomas belong, together with ganglioneuro-blastomas<br />

and ganglioneuromas, to the group <strong>of</strong> peripheral neuroblastic<br />

tumors (pNTs) derived from sympa-thicoblasts (sympathetic<br />

neuroblasts), i.e. immature precursors <strong>of</strong> the sympathetic<br />

nervous system.<br />

They are considered as embryonic tumors originating<br />

either before or after birth from immature neural crest cells which<br />

are “left over” after normal organogenesis. Thus, the primary<br />

tumors are found in the same anatomical localization as their<br />

normal counterparts, i.e. in the adrenal medulla, and in sympathetic<br />

ganglia and paraganglia.<br />

PNTs are the most common extracranial solid malign<br />

tumors <strong>of</strong> childhood (1/10.000 births) and represent 15%<br />

<strong>of</strong> all malignant neoplasms occurring during the first four<br />

years <strong>of</strong> life [22]. 90% are diagnosed under the age <strong>of</strong> five<br />

years [18].<br />

The challenge <strong>of</strong> heterogeneity<br />

Since the sympathetic neuroblasts <strong>of</strong> embryonic origin display<br />

a restricted maturation capacity in individual tumors,<br />

pNTs are typified by an amazing heterogeneity, both with regard<br />

to the specter <strong>of</strong> histological phenotypes, biological properties<br />

and clinical behavior.<br />

Already the very first reports which were published<br />

during the three last decades <strong>of</strong> the 19 th and the beginning <strong>of</strong><br />

the 20 th century, outlined these neoplasms to comprise features<br />

<strong>of</strong> ganglioneuroma [16], neurocytoma/neuroblastoma<br />

[32] and ganglioneuroblastoma [21]. In 1934, Scherer reported<br />

on fully differentiated ganglioneuromas which contained nodules<br />

<strong>of</strong> either undifferentiated neuroblasts or differentiated<br />

ganglion cells, which he called “development centers” [23].<br />

Thirteen years later, Stout confirmed these observations and<br />

suggested the designation “composite” ganglioneuromas/neuroblastomas<br />

for this variant <strong>of</strong> pNT which today would be<br />

categorized as nodular ganglioneuroblastoma [28].<br />

Address for correspondence<br />

Klaus Beiske, MD, PhD<br />

Department <strong>of</strong> <strong>Pathology</strong><br />

Rikshospitalet<br />

Sognsvannsveien 20<br />

N-0027 Oslo, Norway<br />

Phone: +47 23 07 40 76<br />

Fax: +47 23 07 14 10<br />

E-mail: klaus.beiske@labmed.uio.no


74<br />

Biologically, subsets <strong>of</strong> pNTs, like neuroblastomas <strong>of</strong> stage<br />

4S, can show complete spontaneous regression by massive<br />

apoptosis [10], while others, e.g. those with MYCN amplification,<br />

display aggressive proliferation and are frequently diagnosed<br />

at advanced stages <strong>of</strong> disease [4]. A third variant could<br />

be defined as those which grow slowly achieving more or less<br />

complete maturation. As early as in 1926, Cushing and<br />

Wolbach proposed a sympathetic neuroblastoma to be able to<br />

transform into a ganglioneuroma after comparison <strong>of</strong> tumor<br />

material removed from the same patient with a time interval<br />

<strong>of</strong> 10 years [5]. This idea was later supported by a number <strong>of</strong><br />

reports [6, 7, 12, 30]. Several authors pointed out that more<br />

mature lesions were associated with higher age <strong>of</strong> the patient<br />

[9, 31]. It is not difficult to understand that tumors <strong>of</strong> such<br />

great morphological and biological diversity will present with<br />

various clinical pictures.<br />

Histoprognostic classification <strong>of</strong> neuroblastic tumors<br />

Histopathological classification systems are designed to define<br />

categories and subtypes <strong>of</strong> diseases on the basis <strong>of</strong> commonly<br />

accepted and recognizable morphological features. To<br />

make them applicable to surgical pathology, these categories/<br />

subtypes should ideally be equivalent to clinicopathological<br />

entities implying essential clinical information about prognosis<br />

and appropriate treatment. Considering the above outlined<br />

heterogeneity <strong>of</strong> pNTs, it is obvious that a comprehensible<br />

histoprognostical classification system, based on reproducible<br />

morphological criteria and conveying valuable clinical<br />

information, is not created in a trice for these tumors.<br />

The dilemma <strong>of</strong> ganglioneuroblastomas. Previous attempts<br />

to delimit prognostic categories <strong>of</strong> neuroblastomas and<br />

ganglioneuroblastomas, focused on the differentiation <strong>of</strong><br />

neuroblasts and proposed to list parameters as nuclear and<br />

nucleolar size, relative amount <strong>of</strong> eosinophilic cytoplasm, and<br />

formation <strong>of</strong> clumps, rosettes and/or cytoplasmic (nerve) processes<br />

in an either quantitative [2] or qualitative manner [17].<br />

Although ganglioneuromas were widely accepted as the most<br />

mature form within the specter <strong>of</strong> neuroblastic tumors, the<br />

relative amount <strong>of</strong> Schwannian stroma was not appreciated as<br />

a parameter <strong>of</strong> differentiation for less mature tumors like<br />

ganglioneuroblastomas before the early eighties <strong>of</strong> the 20th<br />

century. Nodules <strong>of</strong> undifferentiated neuroblasts or differentiated<br />

ganglion cells had indeed been recognized in<br />

ganglioneuroblastomas (see above), but rather vague designations<br />

as “development centers” or “composite” tumors indicated<br />

that the maturational relationship <strong>of</strong> these nodules to<br />

the surrounding ganglioneuromatous tissue was poorly understood.<br />

Some were even interpreted as a sign <strong>of</strong> focal partial<br />

or focal complete differentiation [2]. The search for a basic<br />

agreement to the entity <strong>of</strong> ganglioneuroblastoma was further<br />

hampered by a lack <strong>of</strong> commonly accepted histological<br />

criteria for this tumor: Some included neuroblastomas with a<br />

high degree <strong>of</strong> gangliocytic differentiation, while others required<br />

a substantial amount <strong>of</strong> ganglioneuromatous tissue<br />

between foci <strong>of</strong> differentiating neuroblasts [12]. Thus, a<br />

histoprognostic classification <strong>of</strong> ganglioneuroblastomas fell<br />

between two stools, i.e. immature neuroblastomas on one side<br />

and mature ganglioneuromas on the other.<br />

The Shimada Classification<br />

The credit for the first histoprognostical classification system<br />

which includes all peripheral neuroblastic tumors, goes to<br />

Hiroyuki Shimada [26].<br />

Histomorphological differentiation and MKI<br />

Shimada recognized the amount <strong>of</strong> Schwannian stroma in neuroblastic<br />

tumors as a major criterium to distinguish between<br />

immature (i.e. Schwannian stroma-poor) and mature (i.e.<br />

Schwannian stroma-rich) lesions.<br />

The stroma-poor group was further subdivided into undifferentiated<br />

and differentiating tumors depending on whether<br />

the tumor contains less or more than 5% gangliocytic differentiated<br />

neuroblasts. This distinction represented a modification<br />

<strong>of</strong> the prognostic system originally proposed by Beckwith<br />

and Martin for neuroblastic tumors. As a measure <strong>of</strong> aggressive<br />

behavior <strong>of</strong> tumor cells in the stroma-poor group, Shimada<br />

proposed to analyse the mitosis-karyorrhexis index (MKI),<br />

i.e. the percentage <strong>of</strong> mitotic and karyorrhectic nuclei per 5000<br />

tumor cells. He found that an MKI <strong>of</strong> 4% as high. Mitotic and<br />

karyorrhectic cells were summed up in one index because it<br />

was not always possible to distinguish between both phenomena<br />

on light microscopic grounds (Fig. 1).<br />

The stroma-rich group contained three subgroups: (a)<br />

stroma-rich well differentiated tumors consisting mainly <strong>of</strong><br />

ganglioneuromatous tissue with few immature neuroblastic<br />

cells, (b) stroma-rich intermixed tumors with nests <strong>of</strong> variably<br />

differentiated neuroblastic cells, and (c) stroma-rich nodular<br />

tumors with <strong>of</strong>ten grossly visible, hemorrhagic nodules <strong>of</strong><br />

stroma-poor neuroblastoma displaying microscopically pushing<br />

borders towards the surrounding stroma-rich tissue.<br />

Shimada pointed out that his stroma-poor group overlaps with<br />

tumors called by others “classical neuroblastoma” and “diffuse<br />

ganglioneuroblastoma”, while the stroma-rich group corresponds<br />

to tumors previously called “ganglioneuroblastoma” and<br />

“immature ganglioneuroma”.<br />

Fig. 1 Undifferentiated neuroblastoma with area <strong>of</strong> high MKI. Karyorrhectic<br />

cells <strong>of</strong>ten show condensed eosinophilic cytoplasm and hyperchromatic,<br />

fragmented nuclei. Round nuclei with a regular outer border might be lymphocytes<br />

and are not accepted as karyorrhectic.


75<br />

Age<br />

In his original paper <strong>of</strong> 1984, Shimada applied these morphological<br />

criteria on histological sections from 295 neuroblastomas<br />

and ganglioneuroblastomas recruited from two American,<br />

one Japanese and one British center. When looking at the<br />

distribution <strong>of</strong> the stroma-poor and stroma-rich groups and<br />

subgroups according to the patients age at diagnosis, he found<br />

that stroma-rich tumors had their debut at a relatively higher<br />

age, i.e. the stroma-rich intermixed and nodular ones mainly<br />

between two and five years, and stroma-rich well differentiated<br />

ones at the age <strong>of</strong> five years and older. These findings<br />

confirmed previous observations (see above) and supported<br />

the idea that the immature elements in the non-aggressive variants<br />

<strong>of</strong> these embryonic tumors have the capability to mature<br />

into benign ganglioneuromas over a certain number <strong>of</strong> years.<br />

On the other hand, a disturbance (e.g. delay) <strong>of</strong> this<br />

“normal” sequence <strong>of</strong> maturation could probably forebode a<br />

tumor which because <strong>of</strong> its restricted maturation capacity will<br />

persist at an immature, biologically more aggressive stage.<br />

Shimada found strong support also for this hypothesis when<br />

he compared the age distribution <strong>of</strong> patients with stroma-poor<br />

tumors to MKI and clinical outcome. The majority <strong>of</strong> undifferentiated<br />

and differentiated stroma-poor tumors with low<br />

MKI diagnosed under the age <strong>of</strong> 1,5 year had a favorable outcome,<br />

while almost all undifferentiated tumors with low MKI<br />

diagnosed at >1,5 years killed the patients. Stroma-poor tumors<br />

with high MKI had a bad outcome, and the majority<br />

occurred in children older than 1,5 year. All children with<br />

stroma-poor tumors diagnosed at >5 years died.<br />

The stroma-rich intermixed and well differentiated subgroups<br />

occurred in children older than 2 and 5 years, respectively,<br />

and almost all showed a favorable outcome. In contrast,<br />

most <strong>of</strong> the stroma-rich nodular tumors had an unfavorable<br />

prognosis, irrespective <strong>of</strong> their occurrence at the “correct”<br />

age <strong>of</strong> 2-5 years. It seemed probable that the stromapoor<br />

nodules could represent a malignant outgrowth <strong>of</strong> biologically<br />

more aggressive clones within an otherwise maturing<br />

stroma-rich tumor.<br />

Taken together, Shimada was the first to prove the usefulness<br />

<strong>of</strong> a classification based on a combination <strong>of</strong><br />

histomorphology (i.e. the relative amount <strong>of</strong> Schwannian<br />

stroma and gangliocytic differentiation), MKI and age at diagnosis<br />

and concluded that<br />

“1) the prognosis <strong>of</strong> the patients under 1,5 years old is directly<br />

influenced by the MKI regardless <strong>of</strong> the degree <strong>of</strong> maturation,<br />

2) the prognosis <strong>of</strong> the patients between 1,5 and 5 years old is<br />

influenced both by the degree <strong>of</strong> maturation and the MKI,<br />

3) the prognosis <strong>of</strong> the patients over 5 years old is poor regardless<br />

<strong>of</strong> the degree <strong>of</strong> maturation and/or the MKI.”<br />

Modifications <strong>of</strong> the Shimada classification<br />

In 1992, Joshi [12] searched to combine the conventional (pre-<br />

Shimada) terminology <strong>of</strong> neuroblastic tumors with the prognostic<br />

categories <strong>of</strong> the Shimada classification and suggested<br />

the following changes/additions:<br />

(1) The stroma-poor tumors <strong>of</strong> Shimada should be called<br />

neuroblastomas and defined as tumors with a neuroblastic<br />

component <strong>of</strong> >50% <strong>of</strong> the total tumor area.<br />

(2) Shimadas stroma-poor differentiated tumors should be<br />

splitted up into poorly differentiated neuroblastomas<br />

(containing neuropil and 5% differentiated<br />

neuroblasts).<br />

(3) Shimadas stroma-rich tumors should be called<br />

ganglioneuroblastoma, nodular, intermixed and borderline<br />

and should be defined as tumors containing >50%<br />

ganglioneuromatous tissue. The borderline type was<br />

meant to replace Shimadas stroma-rich well differentiated<br />

type.<br />

(4) Tumors with equal amounts <strong>of</strong> neuroblastic and<br />

ganglioneuromatous components should be called “transitional<br />

neuroblastic tumors”. The term “unclassifiable<br />

neuroblastic tumor” should be reserved for tumors where<br />

the histological assessment was hampered by necrosis,<br />

hemorrhage, calcification, crush artifacts, cystic changes,<br />

and poor fixation/processing.<br />

Interestingly, Joshi also performed a histoprognostic categorization<br />

<strong>of</strong> the nodules in nine nodular ganglioneuroblastomas<br />

(see below), without being able to show any significant impact<br />

on the survival <strong>of</strong> these few patients.<br />

In other publications, he proposed a histological grading<br />

system based on the presence/absence <strong>of</strong> calcification and<br />

level <strong>of</strong> mitotic rate (MR) [13] which he later modified by<br />

replacing MR with MKI [15]. By combining these (modified)<br />

histological grades with the patients age, he defined (modified)<br />

low and high risk groups independent <strong>of</strong> the morphological<br />

differentiation.<br />

The International Neuroblastoma <strong>Pathology</strong><br />

Classification (INPC)<br />

In 1994, the International Neuroblastoma <strong>Pathology</strong> Committee<br />

was established with the goal to achieve a “standardization<br />

<strong>of</strong> terminology and morphologic criteria <strong>of</strong> neuroblastic<br />

tumors and establishment <strong>of</strong> a morphologic classification that<br />

is prognostically significant, biologically relevant, and reproducible”<br />

[24]. Both Shimada and Joshi were members <strong>of</strong> this<br />

committee, and the proposed classification represents by and<br />

large the original Shimada system <strong>of</strong> 1984 with a few modifications,<br />

some proposed previously by Joshi (see above), and<br />

some performed by the committee.<br />

Morphological categorization<br />

The following categories and subtypes <strong>of</strong> peripheral neuroblastic<br />

tumors were listed:<br />

1. Neuroblastoma (Schwannian Stroma-Poor)<br />

Definition: PNT with 50% or less Schwannian stroma<br />

a. undifferentiated subtype<br />

consists <strong>of</strong> small-to-medium sized neuroblasts without<br />

neuritic processes (neuropil) or gangliocytic<br />

differentiation (Fig. 2). Scattered or foci <strong>of</strong> large or<br />

pleomorphic-anaplastic cells occur infrequently.


76<br />

The diagnosis <strong>of</strong> this subtype <strong>of</strong> pNT may pend on ancillary<br />

techniques as e.g. immunohistology (NSE, CD56, tyrosine<br />

hydroxylase, synaptophysin, chromogranin A and markers to<br />

exclude myogenic tumors, primitive neuroectodermal tumors/<br />

Ewing sarcoma and lymphomas), cytogenetics and elevated<br />

urinary levels <strong>of</strong> metabolites <strong>of</strong> catecholamine synthesis.<br />

b. poorly differentiated subtype<br />

consists <strong>of</strong> neuroblasts and neuropil. Less than 5% <strong>of</strong><br />

neuroblasts show gangliocytic differentiation (Fig. 3).<br />

Scattered or foci <strong>of</strong> large or pleomorphic-anaplastic<br />

cells occur infrequently.<br />

c. differentiating subtype<br />

consists <strong>of</strong> neuroblasts and neuropil. 5% or more <strong>of</strong><br />

neuroblasts show synchronous gangliocytic<br />

differentiation (Fig. 4), i.e. enlarged, eccentric nucleus,<br />

vesicular chromatin pattern, prominent nucleolus and<br />

increased amount <strong>of</strong> cytoplasm (total cell diameter<br />

larger than twice the nuclear diameter). Areas <strong>of</strong><br />

Schwannian stroma can be seen at the periphery <strong>of</strong> tumor<br />

(transitional zone), but do not exceed 50% <strong>of</strong> the<br />

tumor area.<br />

2. Ganglioneuroblastoma, intermixed (Schwannian<br />

Stroma-Rich)<br />

Definition: pNT with >50% ganglioneuromatous tissue which<br />

includes sharply defined nests <strong>of</strong> neuroblastic tissue (Fig. 5)<br />

containing neuropil and a mixture <strong>of</strong> differentiating neuroblasts<br />

and and maturing ganglion cells (Fig. 6).<br />

3. Ganglioneuroma (Schwannian Stroma-Dominant)<br />

a. Ganglioneuroma, maturing subtype<br />

consists <strong>of</strong> >50% ganglioneuromatous tissue with scattered<br />

differentiating neuroblasts, maturing and mature<br />

ganglion cells which never lie in sharply demarcated<br />

nests (Fig. 7). This subtype corresponds to Shimadas<br />

stroma-rich, well differentiated [26] and Joshis<br />

ganglioneuroblastoma, borderline.<br />

b. Ganglioneuroma, mature subtype<br />

consists <strong>of</strong> mature Schwannian stroma with fascicular<br />

neuritic processes and mature ganglion cells<br />

(Fig. 8). Neuroblasts are no longer present.<br />

Fig. 2 The undifferentiated subtype <strong>of</strong> neuroblastoma consists <strong>of</strong> small-tomedium<br />

sized cells without neuritic processes or gangliocytic differentiation.<br />

Fig. 3 In the poorly differentiated subtype, the neuroblasts are surrounded by<br />

a fibrillary meshwork <strong>of</strong> neuritic processes (neuropil). A few cells (50% Schwannian<br />

stroma which includes sharply defined nests <strong>of</strong> neuroblastic tissue.


77<br />

4. Ganglioneuroblastoma, Nodular (Composite Schwannian<br />

Stroma-Rich/Stroma-Dominant and Stroma-<br />

Poor)<br />

Definition: pNT with a stroma-rich (like in ganglioneuro-blastoma,<br />

intermixed) or strom-dominant (like in ganglioneuroma,<br />

maturing) component which surrounds macroscopically visible,<br />

<strong>of</strong>ten hemorrhagic nodules <strong>of</strong> a neuroblastic, stroma-poor<br />

component (Fig. 9 a and b), regarded as a more aggressive<br />

clone. The nodule(s) show either pushing borders (Fig. 10) or<br />

a more infiltrative pattern towards the stroma-rich component.<br />

It is important to be aware <strong>of</strong> the possibility that a surgical<br />

biopsy may only contain a small rim <strong>of</strong> stroma-rich tissue<br />

around the nodule, i.e. the stroma-rich part will not necessarily<br />

make up >50% <strong>of</strong> the visible tumor area.<br />

5. Terminology for not completely classifiable pNTs<br />

a. Neuroblastoma (Schwannian Stroma-Poor), NOS<br />

is reserverd for tumors which clearly belong to the category<br />

<strong>of</strong> Schwannian stroma-poor pNT, but which can<br />

not be allocated to any subtype because <strong>of</strong> poor quality<br />

<strong>of</strong> the sections, bleeding, cystic degeneration, necrosis,<br />

crush artifact or diffuse calcification.<br />

Fig. 6 The neuroblastomatous nests <strong>of</strong> ganglioneuroblastoma, intermixed,<br />

contain differentiating neuroblasts, maturing ganglion cells and neuropil.<br />

b. Ganglioneuroblastoma, NOS<br />

is applied to a stroma-rich tumor which extensive calcification<br />

which may obscure a stroma-poor nodule.<br />

c. Neuroblastic tumor, unclassifiable<br />

is recommended for a pNT which cannot be allocated<br />

to any <strong>of</strong> the categories listed above. This may e.g apply<br />

to very small biopsies.<br />

Prognostic categorization<br />

In order to (1) test the consensus between the committee members<br />

to recognize the above listed morphological categories<br />

and (2) identify the most powerful system for prognostic<br />

categorization, the committee reviewed 227 cases <strong>of</strong> pNTs<br />

[25] and compared the original Shimada system (based on<br />

morphology, MKI and age) with Joshi’s histological grades<br />

(based on MR and calcification), risk groups (based on histological<br />

grade and age) and their modifications replacing MR<br />

with MKI, which can only be used for prognostic analysis <strong>of</strong><br />

stroma-poor tumors. The combination <strong>of</strong> morphology, MKI<br />

and age, originally proposed by Shimada, had the strongest<br />

impact on prognosis, measured as event free survival (EFS).<br />

The statistical analysis also confirmed the age <strong>of</strong> 1,5 years as<br />

the borderline <strong>of</strong> greatest prognostic significance. Beyond the<br />

age <strong>of</strong> 5 years, all stroma-poor tumors had a bad outcome.<br />

Consequently, the International Neuroblastoma <strong>Pathology</strong><br />

Committee included age and MKI, but not MR and calcification<br />

as relevant prognostic data for stroma-poor tumors<br />

into the new classification.<br />

The MKI can be determined applying a method previously<br />

proposed by Joshi [14] It is based on the initial estimation<br />

<strong>of</strong> cell density in different areas <strong>of</strong> a given tumor (Low:<br />

100-300, Moderate: 300-600, High: 600-900 cells per high<br />

power field (HPF) <strong>of</strong> 400x magnification) and the subsequent<br />

definition <strong>of</strong> the number HPFs which are required to evaluate<br />

5000 tumor cells. The MKI is reported as one <strong>of</strong> three classes<br />

(low, intermediate, high) according to the definitions originally<br />

given by Shimada [26].<br />

Fig. 7 Ganglioneuroma, maturing subtype, contains >50% Schwannian stroma<br />

and single or small aggregates <strong>of</strong> differentiating neuroblasts, and maturing/mature<br />

ganglion cells, but no neuropil.<br />

Fig. 8 Ganglioneuroma, mature subtype, consists <strong>of</strong> abundant Schwannian<br />

stroma and scattered mature ganglion cells.


78<br />

Fig. 9 In ganglioneuroblastoma,<br />

nodular, a ganglioneuromatous<br />

component surrounds nodule(s) <strong>of</strong><br />

neuroblastic stroma-poor tumor<br />

tissue (a). Note lack <strong>of</strong> protein S-<br />

100-positive Schwannian stroma<br />

innside the nodule (b).<br />

Fig. 10 The neuroblastic nodule (left) shows pushing borders by compressing<br />

the surrounding ganglioneuromatous tissue (right).<br />

Table 1 shows the prognostic allocation <strong>of</strong> the morphological<br />

categories and subtypes <strong>of</strong> pNTs as recommended in the INPC.<br />

The process <strong>of</strong> reaching a final histoprognostic conclusion<br />

according to differentiation, MKI and age may appear confusing<br />

to the inexperienced. For newcomers, it can be recommended<br />

to learn the favorable categories by heart because their<br />

number is quite low: poorly differentiated and differentiating<br />

tumors with low and intermediate MKI below 1,5 years, and<br />

differentiating neuroblastoma with low MKI between 1,5 and<br />

5 years. All other neuroblastomas are actually unfavorable.<br />

Similar to the Shimada classification <strong>of</strong> 1984, Schwannian<br />

stroma-rich and dominant tumors have a favorable prognosis<br />

unrelated to the patient’s age, except for nodular<br />

ganglioneuroblastomas which all are ascribed an unfavorable<br />

outcome.<br />

Revision <strong>of</strong> the International Neuroblastoma <strong>Pathology</strong><br />

Classification (INPC)<br />

Both Joshi [12] and the International Neuroblastoma <strong>Pathology</strong><br />

Committee [24, 25] mentioned the possibility <strong>of</strong> performing<br />

a prognostic grading <strong>of</strong> nodular ganglioneuroblastoma<br />

(GNBn) on the basis <strong>of</strong> the stroma-poor nodule(s) since this<br />

component <strong>of</strong> the tumor had been regarded as the malignant<br />

one already many years ago [28]. However, the number <strong>of</strong><br />

GNBns in their reviews was too small.<br />

Umehara et al. [29] were the first to review 70 GNBns<br />

and to perform a prognostic categorization by analyzing the<br />

nodule(s) according to the above detailed criteria <strong>of</strong> the INPC<br />

for neuroblastomatous tumors (morphology and MKI) and<br />

relating their data to the patients’ age. These authors also described<br />

GNBns with single or multiple nodules and variant<br />

forms which (1) consisted mainly <strong>of</strong> the stroma-poor component<br />

surrounded only by a thin, microscopically visible rim<br />

<strong>of</strong> stroma-rich/-dominant tissue, and (2) only showed<br />

ganglioneuromatous tissue in the primary tumor (probably due<br />

to poor sampling), but a neuroblastomatous component at the<br />

metastatic site. Depending on the presence <strong>of</strong> nodule(s) <strong>of</strong> the<br />

favorable or unfavorable category, favorable (32,4%) and unfavorable<br />

(67,7%) subsets <strong>of</strong> GNBn were established which<br />

showed statistically significant differences in event free survival<br />

and survival, quite similar to neuroblastic tumors.<br />

In 2003, The International Neuroblastoma <strong>Pathology</strong> Committee<br />

reviewed the same cohort <strong>of</strong> tumors and confirmed these<br />

results [20]. A revision <strong>of</strong> the INPC was proposed including<br />

the following terms: GNBn, classic, containing one nodule;<br />

and variant forms containing either multiple nodules, large<br />

nodules or no nodule, but a metastatic stroma-poor tumor.<br />

Unfavorable prognosis was defined by the demonstration <strong>of</strong><br />

at least one nodule displaying features <strong>of</strong> the unfavorable INPC<br />

categories <strong>of</strong> neuroblastomatous tumors.<br />

The acceptance <strong>of</strong> a large nodule as a variant <strong>of</strong> GNBn<br />

touches on one <strong>of</strong> the principles <strong>of</strong> the former INPC, which<br />

claimed that a tumor belonging to the stroma-rich or -dominant<br />

category has to be made up by >50% Schwannian stroma<br />

(see above). It is important for pathologists to be aware <strong>of</strong> all<br />

variants <strong>of</strong> GNBn because they were two times more frequent<br />

than the classic form among the cases reviewed by Umehara<br />

and Peuchmaur.<br />

The greatest advantage <strong>of</strong> this INPC revision, however,<br />

is brought by the recognition <strong>of</strong> a subset (about 30%)<br />

<strong>of</strong> nodular ganglioneuroblastomas which, in contrast to previous<br />

thinking, belong to a prognostic favorable category<br />

which might benefit from a milder therapeutic regime.


79<br />

Table 1<br />

International Neuroblastoma <strong>Pathology</strong> Classification (INPC). Assignment <strong>of</strong> morphological categories/subtypes to prognostic<br />

categories according to MKI and age (modified from Shimada et al. [25])<br />

Neuroblastoma<br />

Schwannian stroma-poor<br />

favorable<br />

5 yrs all tumors<br />

Ganglioneuroblastoma, intermixed<br />

favorable independent from age<br />

Ganglioneuroma<br />

maturing<br />

mature<br />

favorable independent from age<br />

Ganglioneuroblastoma, nodular<br />

Figure 11 shows the morphologic and prognostic categories<br />

<strong>of</strong> the revised INPC.<br />

Handling <strong>of</strong> tumor material for biological investigations<br />

Schwannian stroma-rich<br />

Schwannian stroma-dominant<br />

composite Schwannian stroma-rich/<br />

stroma-dominant and stroma-poor<br />

Independent from the above outlined endeavor <strong>of</strong> pathologists<br />

towards a clinically informative histoprognostic classification<br />

system for pNTs, a growing number <strong>of</strong> biological and genetic<br />

alterations has been discovered in these tumors during the past<br />

two decades which today allow an assignment to low-, intermediate-<br />

and high-risk groups [3]. The prognostic most important<br />

ones are still MYCN/1p status and ploidy. It is not<br />

possible to enter a child with a neuroblastic tumor into any <strong>of</strong><br />

the current European clinical protocols without at least knowledge<br />

about MYCN, analyzed by Southern blot and/or fluorescence<br />

in situ hybridization (FISH).<br />

Since many <strong>of</strong> these investigations either require or<br />

are most easily performed on unfixed tumor tissue, additional<br />

tasks have to be assumed by pathologists who usually are the<br />

first in the hospital to receive fresh tumor tissue from the operating<br />

theatre.<br />

The various procedures for handling samples <strong>of</strong> neuroblastic<br />

tumors were in detail described by Ambros PF and<br />

Ambros IM [1], and will not be repeated here. The most central<br />

functions <strong>of</strong> the pathologist can be summed up in the following<br />

three key words: sampling, saving and microscopical<br />

evaluation.<br />

Sampling. Taking into account the great morphological<br />

heterogeneity <strong>of</strong> these tumors, it is obvious that the pathologist,<br />

prior to any fixation, should cut the tumor into slices<br />

searching for e.g. nodules or hemorrhagic areas. It is important<br />

to take samples from every macroscopic variant, and this<br />

should proceed under sterile conditions if some <strong>of</strong> the material<br />

is planned to be used for culturing.<br />

The further sub-sampling <strong>of</strong> any macroscopic area,<br />

being <strong>of</strong> interest for special investigations, follows a back-toback<br />

principle, i.e. a representative piece designated for paraffin<br />

embedding should be used to produce at least 10 imprints<br />

from a fresh cut surface (for e.g. FISH or static ploidy<br />

analysis) before fixation in formalin. The two most closely<br />

neighbored (“opposite”) tissue slices should be snap frozen<br />

(for e.g. extraction <strong>of</strong> DNA, RNA and proteins, immunohistology<br />

requiring unfixed tissue and interphase cytogenetics if<br />

imprints were not freshly taken) and put into a sterile culture<br />

medium (for e.g flow cytometry, conventional cytogenetics<br />

and culturing), respectively.<br />

Saving. The adequate saving <strong>of</strong> the samples depends<br />

on the individual pathologists knowledge about the techniques<br />

which are required for the most essential biological investigations.<br />

Thus, imprints should be air dried over night and then<br />

kept frozen in air-tight containers at -80º. Snap frozen tissue<br />

should also be stored at -80º, or in liquid nitrogen if preservation<br />

<strong>of</strong> RNA is intended. Frozen tissue cannot be used for<br />

conventional cytogenetics. Tissue planned to be analyzed by<br />

cytogenetics and flow cytometry, should be brought as soon<br />

as possible in phosphate buffered saline or culture medium to<br />

analysis, although it is possible to recover nuclei for FISH<br />

and flow cytometry from paraffin embedded tissue if fresh<br />

material is not available.


80<br />

Fig. 11 International Neuroblastoma <strong>Pathology</strong> Classification (INPC) (revised form). Morphological and prognostic categories. FH: favorable histology; UH:<br />

unfavorable histology; GNBn: ganglioneuroblastoma, nodular; MKI: mitosis-karyorrhexis index. The prognostic categorization <strong>of</strong> GNBn is performed by<br />

evaluating the neuroblastomatous component in the nodule(s) according to the criteria for neuroblastomas (modified from Peuchmaur et al., 2003).<br />

Microscopical evaluation. The pathologist should not only<br />

analyze morphology and MKI for the histoprognostic classification,<br />

but also use the microscope for estimation <strong>of</strong> the tumor<br />

cell content in the material designed for biological investigations.<br />

Thus, microscoping a paraffin section from the tissue<br />

sample which was used for imprinting prior to fixation,<br />

will provide important information about the possible percentage<br />

<strong>of</strong> tumor cells, non-tumor cells (e.g. lymphocytes or<br />

stroma) and necrosis which might influence the representativity<br />

<strong>of</strong> the imprint. A microscopic analysis <strong>of</strong> the tumor cell content<br />

performed on frozen sections from the material which is<br />

intended to be used e.g. for PCR <strong>of</strong> the 1p36.3 region, is even<br />

more demanded because the correct interpretation <strong>of</strong> the PCR<br />

result requires a tumor cell content <strong>of</strong> at least 70%.<br />

It is obvious that the pathologist who is in charge <strong>of</strong><br />

handling samples from neuroblastic tumors, plays a central<br />

role in providing adequate material for biological analyses.<br />

Prognostic categorization by INPC and biological<br />

markers – competing or complementary systems<br />

It has been known for many years that MYCN amplification<br />

in neuroblastomas is a strong indicator <strong>of</strong> aggressive tumor<br />

behavior [4], and <strong>of</strong>ten associated with undifferentiated morphology<br />

and increased MKI [27]. One might therefore be<br />

tempted to ask whether the histoprognostic categorization provided<br />

by the INPC could be replaced by the analysis <strong>of</strong> MYCN.<br />

The correlation between INPC categorization and<br />

MYCN status was investigated by Goto et al. [8] in a large<br />

cohort <strong>of</strong> 628 patients with neuroblastic tumors. As expected,<br />

97,7% <strong>of</strong> tumors with favorable histology (FH) were MYCN<br />

non-amplified, while 93,2% <strong>of</strong> MYCN amplified tumors<br />

showed an unfavorable histology (UH). However, the comparison<br />

<strong>of</strong> MYCN status and INPC categories revealed a third<br />

and surprisingly large group <strong>of</strong> tumors with UH but without<br />

MYCN amplification. As shown in Fig. 12, the prognosis <strong>of</strong><br />

this group was intermediate to FH/non-amplified and UH/<br />

amplified tumors, and the difference in EFS between all three<br />

groups was statistically significant.<br />

The majority <strong>of</strong> UH, non-amplified tumors were observed<br />

at stage 3 and 4 in children older than one year. The<br />

results <strong>of</strong> this investigation demonstrate that the<br />

histoprognostic INPC categorization is able to identify a substantial<br />

number <strong>of</strong> patients who, in spite <strong>of</strong> normal MYCN<br />

status, belong to a group <strong>of</strong> inferior prognosis, and it appears<br />

relevant to ask whether this category <strong>of</strong> patients should be<br />

<strong>of</strong>fered a modified form <strong>of</strong> therapy.<br />

Further support to this notion comes from the recently<br />

completed histopathological review <strong>of</strong> 124 stage 2A and 2B<br />

MYCN non-amplified trial patients who were treated with<br />

surgery alone according to the LNESG (Localised Neuroblastoma<br />

European Study Group) protocol 94.01 (Navarro et al.,<br />

in preparation). The review was performed according to the<br />

INPC by the seven members <strong>of</strong> the SIOP European Neuroblastoma<br />

(SIOPEN) <strong>Pathology</strong> Reference Panel (Gabriele<br />

Amann, Klaus Beiske, Catherine Cullinane, Emanuele<br />

D’Amore, Claudio Gambini, Sam Navarro, Michel<br />

Peuchmaur), and assigned 20% <strong>of</strong> these MYCN-negative tumors<br />

to the unfavorable category. The difference in event free<br />

and overall survival between the favorable and unfavorable<br />

subset was statistically significant and underlines the importance<br />

<strong>of</strong> the INPC as a valuable tool for prognostic categorization<br />

<strong>of</strong> localized, MYCN non-amplified tumors.<br />

In the forthcoming LNESG2 study, these results have<br />

already influenced the therapeutic design in case <strong>of</strong> relapse <strong>of</strong><br />

the localized tumor. It has been decided that the relapse will


81<br />

receive chemotherapy if the histoprognostic categorization <strong>of</strong><br />

the primary tumor was unfavorable, while the relapse <strong>of</strong> a<br />

histologically favorable tumor will be treated with surgery<br />

alone. However, according to the new protocol, the primary<br />

tumor’s histoprognostic category will only be implied into<br />

therapeutic decisions for a relapse, if the histological slides <strong>of</strong><br />

the primary have been reviewed by one <strong>of</strong> the members <strong>of</strong> the<br />

SIOPEN <strong>Pathology</strong> Reference Panel within six weeks after<br />

the local pathologist finished his/her report. It is mandatory<br />

that the decision about favorable or unfavorable histoprognosis<br />

<strong>of</strong> the localized tumor is based on at least two pathologists<br />

from different institutions. If the local pathologist can not agree<br />

to the conclusion made by the first reviewer, another member<br />

<strong>of</strong> the SIOPEN <strong>Pathology</strong> Reference Panel will be contacted<br />

for a third opinion.<br />

Summary<br />

Pathologists are today ascribed a key role in the diagnostic<br />

work up <strong>of</strong> pNTs:<br />

1. After macroscopic inspection, pathologists are responsible<br />

for an adequate sampling, saving and quality control <strong>of</strong> tumor<br />

material for morphological, biological and genetic investigations.<br />

2. The result <strong>of</strong> the pathologist’s microscopic analysis assigns<br />

a tumor to one <strong>of</strong> five morphological categories which in case<br />

<strong>of</strong> stroma-rich/-dominant tumors (except for nodular<br />

ganglioneuroblastomas) render the analysis <strong>of</strong> biological markers<br />

irrelevant.<br />

The determination <strong>of</strong> morphological differentiation and<br />

MKI enables the pathologist to categorize the<br />

neuroblastomatous tumors and nodular ganglioneuroblastoma<br />

into favorable or unfavorable subsets which will influence the<br />

choice <strong>of</strong> therapy <strong>of</strong> a possible relapse in those patients who<br />

are enrolled into the forthcoming European protocol for localized,<br />

non- MYCN amplified peripheral neuroblastic tumors.<br />

Finally, the great importance <strong>of</strong> obtaining biopsy material,<br />

which is adequate for histoprognostic assessment, cannot<br />

be stressed enough. Cytological specimens <strong>of</strong> tumor cells,<br />

produced from fine needle aspirates, can be used for biological<br />

analyses - provided that a pathologist has controlled their<br />

tumor cell content -, but such samples preclude any<br />

histoprognostic evaluation. The biological information <strong>of</strong> a<br />

MYCN non-amplified status, obtained from cytological<br />

samples, should not make the clinician live in the belief that<br />

this is a tumor <strong>of</strong> good prognosis. Data by Goto et al [8] and<br />

Navarro et al [19], summarized in this paper, provide clear<br />

evidence that MYCN non-amplified tumors may “hide” a<br />

considerable proportion <strong>of</strong> cases with unfavorable outcome.<br />

Fig. 12 Event free survival for patients grouped according to FH (favorable<br />

histology), UH (unfavorable histology) and MYCN status (modified from<br />

Goto et al., 2001).<br />

References<br />

1. Ambros PF, Ambros IM (2001) <strong>Pathology</strong><br />

and biology guidelines for respectable<br />

and unresectable neuroblastic<br />

tumors and bone marrow examination<br />

guidelines. Med Pediatr Oncol<br />

37:492-504<br />

2. Beckwith JB, Martin RF(1968) Observations<br />

on the histopathology <strong>of</strong> neuroblastoma.<br />

J Pediatr Surg 3:106-110<br />

3. Brodeur GM (2003) Neuroblastoma:<br />

biological insights into a clinical<br />

enigma. Nat Rev Cancer 3(3):203-216<br />

4. Brodeur GM, Seeger RC, Schwab M,<br />

Varmus HE, Bishop JM (1984) Amplification<br />

<strong>of</strong> N-myc in untreated human<br />

neuroblastomas correlates with advanced<br />

stage <strong>of</strong> disease. Science<br />

224:1121-1124<br />

5. Cushing H, Wolbach SB (1927) Transformation<br />

<strong>of</strong> malignant paravertebral<br />

sympathicoblastoma into benign ganglioneuroma.<br />

Am J Pathol 3:203-216<br />

6. Everson TC (1964) Spontaneous regression<br />

<strong>of</strong> cancer. Ann N Y Acad Sci<br />

114:767-770<br />

7. Fox F, Davidson J, Thomas LB (1959)<br />

Maturation <strong>of</strong> sympathicoblastoma into<br />

ganglioneuroma. Report <strong>of</strong> two patients<br />

with 20- and 46-year survicals, respectively.<br />

Cancer 12:108-116<br />

8. Goto S, Umehara S, Gerbing RB, et al<br />

(2001) Histopathology (International<br />

Neuroblastoma <strong>Pathology</strong> classification)<br />

and MYCN status in patients with<br />

peripheral neuroblastic tumors. Cancer<br />

92:2699-2708<br />

9. Grosfeld JL, Skinner MA, Rescorla FJ,<br />

West KW, Scherer LR (1994) Mediastinal<br />

tumors in children: experience with<br />

196 cases. Ann Surg Oncol 1:121-127<br />

10. Haas D, Albin AR, Miller C, Zoger S,<br />

Matthay KK (1988) Complete pathologic<br />

maturation and regression <strong>of</strong> stage<br />

IV-S neuroblastoma without treatment.<br />

Cancer 62:818-825<br />

11. Horn RC, Koop CE, Kiesenwetter WB<br />

(1956) Neuroblastoma in childhood.<br />

Lab Invest 5:106-119<br />

12. Joshi VV, Cantor AB, Altshuler G, Larkin<br />

EW, Neill JSA, Shuster JJ, Holbrook CT,<br />

Hayes FA, Castleberry RP (1992) Recommendations<br />

for modification <strong>of</strong> terminology<br />

<strong>of</strong> neuroblastic tumors and prognostic<br />

significance <strong>of</strong> Shimada classification:<br />

a clinicopathologic study <strong>of</strong> 213<br />

cases from the Pediatric Oncology<br />

Group. Cancer 69:2183-2196


13. Joshi VV, Cantor AB, Altshuler G,<br />

Karkin EW, Neill JSA, Shuster JJ,<br />

Holbrook CT, Hayes FA, Nitschke R,<br />

Duncan MH, Shochat SJ, Talbert J,<br />

Smith EI, Castleberry RP (1992) Agelinked<br />

prognostic categorization based<br />

on a new histologic grading system <strong>of</strong><br />

neuroblastomas. Cancer 69:2197-221<br />

14. Joshi VV, Chatten J, Sather HN,<br />

Shimada H (1991) Evaluation <strong>of</strong><br />

Shimada classification in advanced neuroblastoma<br />

with special reference to the<br />

mitosis karyorrhexis index: a report<br />

from the Children’s Cancer Study<br />

Group. Mod Pathol 4:139-147<br />

15. Joshi VV, Rao PV, Cantor AB, Altshuler<br />

G, Shuster JJ, Castleberry RP (1996)<br />

Modified histologic grading <strong>of</strong> neuroblastoma<br />

by replacement <strong>of</strong> mitotic rate<br />

with mitosis karyorrhexis index: a clinicopathologic<br />

study <strong>of</strong> 223 cases from<br />

the Pediatric Oncology Group. Cancer<br />

77:1582-1588<br />

16. Loretz W. Ein Fall von gangliosem<br />

Neurom (Gangliom) (1870) Arch Pathol<br />

Anat Physiol Klin Med 49:435-437<br />

17. Mäkinen J (1972) Microscopic patterns<br />

as a guide to prognosis <strong>of</strong> neuroblastomas<br />

in childhood. Cancer 29:1637-1646<br />

18. Monforte-Munoz H, Kawakami T,<br />

Stram D, Gerbing R, Matthay KK,<br />

Lukens J et al (1997) Neuroblastoma in<br />

children over 5 years <strong>of</strong> age: recognition<br />

<strong>of</strong> a rare and aggressive subset with<br />

“unconventional” morphology. Mod<br />

Pathol 10:4P<br />

19. Navarro S, Amann G, Beiske K,<br />

Cullinane C, D’Amore E, Gambini C,<br />

Mosseri V, De Bernardi B, Michon J,<br />

Peuchmaur M (2004) Prognostic value<br />

<strong>of</strong> International Neuroblastoma <strong>Pathology</strong><br />

Committee Classification in<br />

localised resectable peripheral neuroblastic<br />

tumors. In preparation<br />

20. Peuchmaur M, d’Amore ESG, Joshi VV,<br />

et al (2003) Revisión <strong>of</strong> the International<br />

Neuroblastoma <strong>Pathology</strong> Classification.<br />

Confirmation <strong>of</strong> favorable and unfavorable<br />

prognostic subsets in<br />

Ganglioneuroblastoma nodular. Cancer<br />

298:2274-2281<br />

21. Robertson HE (1915) Das Ganglioneuroblastom,<br />

ein besonderer Typus im<br />

System der Neurone. Virchows Arch<br />

[Pathol Anat] 63:147-168<br />

22. Ross JA, Severson RK, Pollock BH,<br />

Robison LL (1996) Childhood cancer in<br />

the United States: a geographic analysis<br />

<strong>of</strong> cases from the Pediatric Cooperative<br />

Clinical Trials Group. Cancer<br />

77:201-207<br />

23. Scherer HJ (1934) Bemerkungen zur<br />

Empfindlichkeit der Ganglienzelle.<br />

Virchows Arch Path Anat 293:429-437<br />

24. Shimada H, Ambros IM, Dehner LP,<br />

Hata J, Joshi VV, Roald B (1999) Terminology<br />

and morphologic criteria <strong>of</strong><br />

neuroblastic tumors: recommendations<br />

by the International Neuroblastoma <strong>Pathology</strong><br />

Committee. Cancer 86:349-363<br />

25. Shimada H, Ambros IM, Dehner LP, et<br />

al (1999) The International Neuroblastoma<br />

<strong>Pathology</strong> Classification (the<br />

Shimada system). Cancer 86:364-372<br />

26. Shimada H, Chatten J, Newton WA Jr,<br />

Sachs N, Hamoudi AB, Chiba T,<br />

Marsden HB, Misugi K (1984) Histopathologic<br />

prognostic factors in neuroblastic<br />

tumors: definition <strong>of</strong> subtypes <strong>of</strong><br />

ganglioneuroblastoma and an age-linked<br />

classification <strong>of</strong> neuroblastomas. J Natl<br />

Cancer Inst 73:405-416<br />

27. Shimada H, Stram DO, Chatten J, et al<br />

(1995) Identification <strong>of</strong> subsets <strong>of</strong> neuroblastomas<br />

by combined histopathologic<br />

and N-myc analysis. J Natl Cancer<br />

Inst 187:1470-1476<br />

28. Stout AP (1947) Ganglioneuroma <strong>of</strong> the<br />

sympathetic nervous system. Surg<br />

Gynecol Obstet 84:101-110<br />

29. Umehara S, Nakagava A, Matthay KK,<br />

Lukens JN, Seeger RC, Stram DO,<br />

Gerbing RB, Shimada H (2000) Histopathology<br />

defines prognostic subsets <strong>of</strong><br />

ganglioneuroblastoma, nodular. Cancer<br />

89:1150-1161<br />

30. Visfeldt J (1963) Transformation <strong>of</strong><br />

sympathicoblastoma into ganglioneuroma.<br />

Acta Path Microbiol Scand<br />

158:414-428<br />

31. Wahl HR (1914) Neuroblastoma: with<br />

study <strong>of</strong> a case illustrating the three types<br />

that arise from the sympathetic nervous<br />

system. J Med Res 30:205-260<br />

32. Wright JH (1910) Neurocytoma or neuroblastoma,<br />

a kind <strong>of</strong> tumor not generally<br />

recognized. J Exper Med xii:556


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Intracranial ependymomas in children: correlation <strong>of</strong> selected<br />

factors and event-free survival in the group <strong>of</strong> 142 consecutive<br />

patients treated at one institution.<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

S³awomir Barszcz 1 , Wies³awa Grajkowska 2 , Danuta Perek 3 , Marcin Roszkowski 1 , Monika<br />

Drogosiewicz 3 , Marta Perek-Polnik 3 , El¿bieta Jurkiewicz 4 , Pawe³ Daszkiewicz 1<br />

1<br />

Department <strong>of</strong> Neurosurgery,<br />

2<br />

Department <strong>of</strong> <strong>Pathology</strong>,<br />

3<br />

Department <strong>of</strong> Oncology,<br />

4<br />

Department <strong>of</strong> Radiology,<br />

The Children’s Memorial Health Institute,<br />

Warsaw, Poland<br />

Abstract<br />

Ependymomas are among the most frequent intracranial tumors in children. Nevertheless, the identification<br />

<strong>of</strong> risk factors and the role <strong>of</strong> various treatment modalities are still far from standarization. The aim <strong>of</strong> this<br />

study was to evaluate the impact <strong>of</strong> patients’ age, tumor location, extent <strong>of</strong> surgical resection, degree <strong>of</strong><br />

histological malignancy, type <strong>of</strong> radiotherapy (RT), and chemotherapy (CHT) on event free survival in the<br />

group <strong>of</strong> 142 consecutive patients treated at one institution. Retrospective analysis and comparisons <strong>of</strong> eventfree<br />

survival (EFS) probabilities were done in subgroups <strong>of</strong> patients selected on the basis <strong>of</strong> various treatemt<br />

protocols. For the entire group, age <strong>of</strong> patients, extent <strong>of</strong> surgical resection, and histological malignancy<br />

significantly affected EFS. Extent <strong>of</strong> surgical resection significantly influenced EFS in benign tumors and<br />

did not in anaplastic ependymomas. Neuroaxis RT did not improve EFS for anaplastic lesions. Treatment<br />

results were better after 1997 in anaplastic ependymomas (potential role <strong>of</strong> CHT) and finally CHT significantly<br />

improved control <strong>of</strong> small tumor residues<br />

Key words: chemotherapy, ependymoma, prognosis, radiotherapy, surgery<br />

Introduction<br />

Ependymomas are among the most frequent intracranial tumors<br />

in children, accounting for approximately 10% <strong>of</strong> primary<br />

central nervous system neoplasms [35]. Treatment <strong>of</strong><br />

patients with ependymomas includes neurosurgical resection,<br />

radiotherapy (RT), chemotherapy (CHT), and radiosurgery<br />

[16, 33]. Despite introduction <strong>of</strong> various treatment regimens,<br />

treatment outcomes are still unsatisfactory [27, 31, 36].<br />

The aim <strong>of</strong> the study was to analyse the influence <strong>of</strong><br />

selected factors on event-free survival (EFS) in the group <strong>of</strong><br />

intracranial ependymomas treated at our institution.<br />

Material and methods<br />

In the years since 1981 thru 2003, a total number <strong>of</strong> 154 children<br />

suffering from intracranial ependymomas were treated<br />

at the Children’s Memorial Health Institute in Warsaw. Twelve<br />

patients were lost to follow-up, and the remaining group <strong>of</strong>142<br />

children (74 boys and 68 girls) were used for further analysis.<br />

Retrospective analysis <strong>of</strong> the data was performed to<br />

evaluate the influence <strong>of</strong> patients’age, tumor location, extent<br />

<strong>of</strong> surgery, tumor malignancy, type <strong>of</strong> radiotherapy (RT) and<br />

use <strong>of</strong> chemotherapy (CHT) on event-free survival (EFS),<br />

according to various treatmet protocols. The group <strong>of</strong> patients<br />

was heterogenous (various treatment protocols, mainly before<br />

and after 1997). Age <strong>of</strong> patients, extent <strong>of</strong> resection, and tumor<br />

malignancy were the main factors influencing selection<br />

Address for correspondence<br />

S³awomir Barszcz, MD, PhD<br />

Department <strong>of</strong> Neurosurgery<br />

Children's Memorial Health Institute<br />

Al. Dzieci Polskich 20<br />

04-853 Warsaw, Poland<br />

Phone: +48 22 8157560<br />

E-mail: slawomirbarszcz@wp.pl


84<br />

<strong>of</strong> treatment options. In such circumstances the mentioned<br />

factors with potential influence on survival were not independent<br />

verieties, and multivariate analysis could not be performed.<br />

We made a 2-step statistical analysis. First, comparability<br />

<strong>of</strong> the subgroups <strong>of</strong> benign and anaplastic ependymomas<br />

was ascertained, according to age, extent <strong>of</strong> surgery, type<br />

<strong>of</strong> RT, and use <strong>of</strong> CHT. Then, we made a few separate comparisons<br />

for comparable subgroups <strong>of</strong> patients in order to<br />

analyse the potential influence <strong>of</strong> risk factors on actuarial EFS.<br />

EFS probability curves were drawn, and their stratification<br />

was studied using the log-rank test.<br />

We did not study overall survival (OS), because recurrences<br />

<strong>of</strong> tumors as well as their treatments were not analysed.<br />

Results<br />

Age <strong>of</strong> the patients ranged from 5 months to 18 years and 6<br />

months (median 5 years 0 months). In 63 patients ependymomas<br />

were located in the infratentorial space, and in 59 children<br />

supratentorial tumors were found. In 61 patients the tumors<br />

were totally, in 38 subtotally, and in 38 children partially<br />

resected. In the remaining 5 cases only biopsies were<br />

performed. In the years since 1981 thru 1999, the extent <strong>of</strong><br />

surgical tumor removal was evaluated on the basis <strong>of</strong> surgical<br />

reports and postoperative control CTs. Since 1999, the intraoperative<br />

estimation as well as postoperative MRI studies have<br />

been used to determine the extent <strong>of</strong> surgical resection. All<br />

pathological specimens were reviewed and devided into two<br />

subgroups <strong>of</strong> 89 benign and 53 malignant (anaplastic) ependymomas.<br />

Anaplastic tumors matched the following critera:<br />

marked nuclear pleomorphism, high mitotic activity, necrosis,<br />

vascular proliferation. 118 patients were treated with radiotherapy.<br />

Radiotherapy significanty changed over the described<br />

period <strong>of</strong> time, due to the developement <strong>of</strong> cobalttherapy<br />

and linac technology as well as introduction <strong>of</strong> new<br />

treatment protocols. To simplify the variety <strong>of</strong> problems concerning<br />

radiotherapy, for further analysis we defined only two<br />

main subgroups: 1. “local” radiotherapy (29 patients) and 2.<br />

neuroaxis radiotherapy (89 patients). Untill 1996, irrespective<br />

<strong>of</strong> the extent <strong>of</strong> surgical resection, all patients underwent<br />

whole neuroaxis irradiation. Since1996 thru 2000, patients<br />

after total removal <strong>of</strong> benign ependymomas were treated with<br />

“local” radiotherapy. In 2000 the “wait and see” approach was<br />

introduced after total removal <strong>of</strong> benign tumors. Partial resections<br />

<strong>of</strong> benign ependymomas were followed by chemotherapy<br />

and “local” RT. Anaplastic ependymomas after surgery were<br />

treated with adjuvant chemotherapy followed by tumor bed<br />

and ventricles irradiation, and maintenance chemotherapy. Two<br />

different treatment approaches were tailored for children under<br />

3 years <strong>of</strong> age. Total resection <strong>of</strong> benign ependymoma<br />

was the only treatment modality. The rest <strong>of</strong> patients below 3<br />

years <strong>of</strong> age (partial resection <strong>of</strong> benign tumor and all patients<br />

with anaplastic ependymomas) were treated with 18-months’<br />

chemotherapy without RT. The use <strong>of</strong> the whole neuroaxis<br />

RT was reserved only for disseminated tumors. Chemotherapy<br />

was used for the treatment <strong>of</strong> 58 patients. The median followup<br />

period was 3 years and 2 months (ranging from 2 months<br />

to 18 years and 6 months).<br />

In the study group there were no significant differences<br />

between patients with benign and anaplastic tumors according<br />

to age and extent <strong>of</strong> resection. The use <strong>of</strong> neuroaxis vs. local RT<br />

were close to the limit <strong>of</strong> signifficance (Table 1). The entire<br />

study group was devided into 3 age-dependent subgroups. There<br />

were 41 children below 3 years <strong>of</strong> age. In 75 patients, age ranged<br />

from 3 to 10 years, and 26 patients were older than 10 years.<br />

The list <strong>of</strong> subsequent comparisons <strong>of</strong> potential risk factors,<br />

outcomes and statistical analysis are shown in Table 2.<br />

The study revealed that age <strong>of</strong> patients significantly influenced<br />

EFS.The lower the age, the worse the prognosis (Fig.<br />

1). No differences were found concernig event-free survival<br />

probabillities for the group <strong>of</strong> 83 patients with infratentorial<br />

tumors vs. 59 patients with supratentorial ependymomas. Primary<br />

tumor location did not influence EFS. Diagnosis <strong>of</strong> malignant<br />

(anaplastic) ependymoma according to the mentioned<br />

morphologic criteria was a marker <strong>of</strong> poor prognosis in the study<br />

group (Fig. 2). Event-free survival probability for the group <strong>of</strong><br />

99/142 patients after total and subtotal tumor resection was better<br />

than for the group <strong>of</strong> 43/142 patients after partial resection and<br />

biopsy. Extent <strong>of</strong> surgical resection significantly influenced EFS<br />

for the entire study group. Extent <strong>of</strong> surgical removal significantly<br />

influenced EFS for benign tumors. The prognosis was<br />

better for patients after total and subtotal resection <strong>of</strong> benign<br />

ependymomas (Fig. 3). Event-free survival probability after total<br />

and subtotal resection <strong>of</strong> grade III ependymomas was slightly<br />

better than after less radical resection, but stratification <strong>of</strong> survival<br />

curves was not significant. The comparison <strong>of</strong> EFS probabilities<br />

for benign ependymomas treated with neuroaxis and<br />

“local” RT revealed that EFS was better for the subgroup after<br />

whole neuroaxis irradiation. It is rather confusing, taking into<br />

account everyday clinical practice. Before 1996, all the cases<br />

<strong>of</strong> intracranial ependymomas irrespective histological malignancy<br />

and extent <strong>of</strong> surgical resection received whole neuroaxis<br />

RT, whereas after 1996 neuroaxis irradiation was reserved for<br />

disseminated lesions only. There was no statistically significant<br />

difference between EFS probabilities for neuroaxis vs. “local”<br />

(tumor bed or tumor bed + ventricles) RT for the patients<br />

with malignant ependymomas. It was shown that introduction<br />

(1997) <strong>of</strong> the standardized treatment protocols including: surgery,<br />

adjuvant chemotherapy, tumor bed or tumor bed + ventricles<br />

radiotherapy, and finally maintenance chemotherapy significantly<br />

improved EFS probability for malignant ependymomas<br />

(Fig. 4). Statistically significant stratification <strong>of</strong> EFS curves<br />

for the patients treated with chemotherapy vs. patients treated<br />

without chemotherapy after total or subtotal tumor removals<br />

indicates that chemotherapy improved control <strong>of</strong> small tumor<br />

residues (Fig. 5).<br />

Discussion<br />

Identification <strong>of</strong> clinical and pathological factors influencing<br />

survival has a key role in improvement <strong>of</strong> treatment outcomes<br />

in children suffering from intracranial ependymomas. Various<br />

risk factors were proposed, and because <strong>of</strong> relatively small<br />

and heterogenous groups <strong>of</strong> patients and different treatemt<br />

protocols, the results <strong>of</strong> numerous studies differ considerably


85<br />

Table 1<br />

Comparison between ependymoma grade I - II and ependymoma grade III patients according to: age, extent <strong>of</strong> surgery, type <strong>of</strong><br />

RT, and use <strong>of</strong> CHT (NS=not significant)<br />

Table 2<br />

Feature Benign vs. anaplastic (ch 2 ) Significance<br />

Age P > 0.05 NS<br />

Surgery P> 0.05 NS<br />

RT (CNS vs. local) P = 0.06 +/-<br />

CHT P < 0.0005 ∗∗∗<br />

List <strong>of</strong> comparisons <strong>of</strong> the studied factors, and their significance (NS=not significance, * , ** , *** = degrees <strong>of</strong> statistical<br />

significance)<br />

Comparisons <strong>of</strong> EFS probabilities for the studied parameters Log-rank Significance<br />

Three age groups p= 0.0039 ∗∗<br />

Infratentorial vs. supratentorial ependymomas p=0.37 NS<br />

Benign vs. anaplastic ependymomas. p=0.0037 ∗∗<br />

Total and subtotal resections vs. partial resections and biopsies<br />

p=0.0001<br />

∗∗∗<br />

(entire study group).<br />

Total and subtotal resections vs. partial resections and biopsies<br />

p=0.00005<br />

∗∗∗<br />

(benign ependymomas).<br />

Total and subtotal resections vs. partial resections and biopsies<br />

p=0.15<br />

NS<br />

(anaplastic ependymomas).<br />

Neuroaxis RT vs. “local” RT<br />

(benign ependymomas)<br />

p=0.007<br />

∗∗<br />

Neuroaxis RT vs. “local” RT<br />

(anaplastic ependymomas)<br />

p=0.6326<br />

Anaplastic ependymomas treated before 1997 vs. after 1997. p=0.043 ∗<br />

Totally and subtotally removed tumors CHT (+) vs. CHT (-) p=0.047 ∗<br />

NS<br />

[2, 6, 9, 19, 25]. Lower age <strong>of</strong> patients was found to be a poor<br />

prognostic factor in most series [3]. It was not clear wheather<br />

poorer prognosis in very young children was due to different<br />

tumor biology, different treatement protocols, higher morbidity<br />

and poorer response to various treatment modalities especially<br />

radiotherapy [34]. Our results confirmed that observation.<br />

EFS probabilities were lower for the subgroups <strong>of</strong><br />

younger children. The influence <strong>of</strong> primary tumor location on<br />

survival was difficult to evaluate reliably. Predominance <strong>of</strong><br />

infratentorial ependymomas in the first decade <strong>of</strong> life and<br />

higher frequency <strong>of</strong> supratentorial tumors in adolescents and<br />

adults as well as precise tumor location have led to different<br />

treatment outcomes in particular subgroups <strong>of</strong> patients. No<br />

differences were found in the study group concerning two main<br />

localization groups <strong>of</strong> supra- and infratentorial ependymomas.<br />

However, it should be remembered that surgical mortality and<br />

morbidity as well as extent <strong>of</strong> surgical resection are more favorable<br />

in supratentorial extraaxial and midline posterior fossa<br />

ependymomas as compared to midline supratentorial and posterior<br />

fossa paramedian (cerebellopontine angle) tumors [4,<br />

20]. Extent <strong>of</strong> surgical resection was the most consistent and<br />

strongest prognostic factor in almost all <strong>of</strong> series as well as in<br />

our group <strong>of</strong> patients [24, 35]. The main problem in tailoring<br />

treatment and predicting prognosis is to define reliable features<br />

in the diagnosis <strong>of</strong> malignant (anaplastic) ependymomas.<br />

Uncritical use <strong>of</strong> the morfologic criteria <strong>of</strong> histologic<br />

malignancy accepted for astrocytomas is doubtful. In our group<br />

<strong>of</strong> patients, actuarial event-free survival was significantly better<br />

for the so called benign ependymomas, but a lot <strong>of</strong> authors<br />

indicate that there is no strict correlation between tumor morphology<br />

and prognosis in ependymomas [11, 29, 30]. Much<br />

time and effort were spent to study various immunohistochemical<br />

and genetic features as well as their correlations with tumor<br />

biology, but there are still no reliable markers <strong>of</strong> malig-


86<br />

Fig. 1 Statistically significant stratification <strong>of</strong> event-free survival probability<br />

curves for various age groups <strong>of</strong> patients (log-rank: p= 0.0039)<br />

Fig. 2 Significant stratification <strong>of</strong> EFS probabnility curves for benign (B - 89<br />

patients) and anaplastic (A- 53 patients) tumors (log-rank: p=0.0037)<br />

Fig. 3 Significantly higher EFS after total and subtotal resections (T+ST) vs.<br />

partial resections and biopsies (P+B) <strong>of</strong> benign ependymomas (log-rank:<br />

p=0.00005)<br />

Fig. 4 Significant improvement <strong>of</strong> treatment outcome <strong>of</strong> anaplastic ependymomas<br />

after 1997 (introduction <strong>of</strong> complex treatment protocols: surgery +<br />

adjuvant CHT + RT + maintenance CHT; log-rank: p=0.043)<br />

Fig. 5 Better EFS after chemotherapy (CH +) in patients in whom total or<br />

subtotalal tumor resections were performed (log-rank: p=0.047)


87<br />

nancy in ependymomas [7, 21]. Opinions on adjuvant therapy<br />

differ from series to series. It is generally accepted that radiotherapy<br />

and chemotherapy can delay tumor recurrence but their<br />

influence on overall survival is still unclear [1, 5, 23, 28]. The<br />

rationale for agressive removal <strong>of</strong> ependymomas in certain<br />

locations is debatable. Devastating late complications <strong>of</strong><br />

neuroaxis radiotherapy, especially in the youngest age group<br />

are well known [17, 18, 32]. That is why there is a trend to<br />

eliminate radiotherapy in children under 3 years <strong>of</strong> age and<br />

after total removal <strong>of</strong> histologically benign tumors. The fields<br />

<strong>of</strong> irradiation were restricted to tumor bed or tumor bed and<br />

ventricles for partially resected benign and anaplastic lesions<br />

respectively. The neuroaxis irradiation is usually reserved for<br />

disseminated ependymomas [12, 26]. This tendency is supported<br />

by the observations that reccurences are limited to primary<br />

tumor location in almost all cases and CSF dissemination<br />

is relatively rare [13]. Patterns <strong>of</strong> failure indicate, that<br />

final treatment outcome depends mostly on the local control<br />

<strong>of</strong> disease. These statements were the reason for the concept<br />

<strong>of</strong> “second-look” surgery, whenever reoperation was possible<br />

in cases <strong>of</strong> residual disease [8]. Chemotherapy was introduced<br />

in the treatment <strong>of</strong> children under 3 years to delay or<br />

eliminate the need for radiotherapy as well as in the treatment<br />

<strong>of</strong> malignant lesions and tumor recurrences and disseminations<br />

and finally in the treatment <strong>of</strong> partially resected benign<br />

lesions [15, 37]. Variable response rates to the same treatment<br />

protocols created the need for potential identification <strong>of</strong> subpopulations<br />

sensitive or resistent to chemotherapy,<br />

developement <strong>of</strong> novel drugs and intensification <strong>of</strong> treatment<br />

with subsequent bone marrow reconstruction [10, 14, 22]. As<br />

mentioned above, in our series neuroaxis irradiation correleated<br />

with better actuarial EFS for benign ependymomas. Significant<br />

change <strong>of</strong> treatment protocols over the years resulted in<br />

fairly confusing data. Before 1996, all cases were treated with<br />

neuroaxis irradiation; after 1996, RT was reserved only for<br />

disseminated ependymomas. It was clearly shown that<br />

neuroaxis RT did not improve EFS in anaplastic ependymomas<br />

in our series. An introduction <strong>of</strong> standarized treatment<br />

protocols (including chemotherapy) in 1997 improved treatment<br />

outcomes in malignant tumors. Chemotherapy also improved<br />

control <strong>of</strong> small tumor residues after surgery. Treatment<br />

response <strong>of</strong> tiny remnants seems to be an interesting<br />

clinical model for evaluation <strong>of</strong> CHT efficacy in the treatment<br />

<strong>of</strong> ependymomas.<br />

Conclusions<br />

1. For the entire group <strong>of</strong> patients, age, extent <strong>of</strong> surgical<br />

resection, and histological malignancy, significantly influenced<br />

EFS;<br />

2. Extent <strong>of</strong> surgical resection significantly influenced EFS in<br />

benign tumors and did not influence EFS in anaplastic<br />

ependymomas;<br />

3. Neuroaxis RT did not influence EFS in patients with anaplastic<br />

ependymomas;<br />

4. Treatment results in anaplastic ependymomas improved after<br />

1997 (a potential role <strong>of</strong> CHT);<br />

5. CHT significantly improved control <strong>of</strong> small tumor residues.<br />

References<br />

1. Bouffet E, Foreman N (1999) Chemotherapy<br />

for intracranial ependymomas<br />

(review). Child’s Nerv Syst 15:563-323<br />

2. Bouffet E, Perilongo G, Canete A, et al<br />

(1998) Intracranial ependymomas in<br />

children: a critical review <strong>of</strong> prognostic<br />

factors and a plea for cooperation. Medical<br />

and Pediatric Oncology 30:319-331<br />

3. Duffner PK, Krischer JP, Sanford RA,<br />

et al (1998) Prognostic factors in infants<br />

and very young children with intracranial<br />

ependymomas. Pediatr Neurosurg<br />

28:215-222<br />

4. Ernestus R, Schroder R, Stutzer H, et al<br />

(1996) Prognostic relevance <strong>of</strong> localization<br />

and grading in intracranial ependymomas<br />

<strong>of</strong> childhood. Child’s Nerv Syst<br />

12:522-526<br />

5. Evans AE, Anderson JR, Lefkowitz-<br />

Boudreaux IB, et al (1996) Adjuvant<br />

chemotherapy <strong>of</strong> childhood posterior<br />

fossa ependymoma: caraniospinal irradiation<br />

with or without adjuvant CCNU,<br />

vincristine, and prednisone: a Childrens<br />

Cancer Group study. Med Pediatr Oncol<br />

27:8-14<br />

6. Figarella-Branger D, Civatte M, Bouvier-Labit<br />

C, et al (2000) Prognostic factors<br />

in intracranial ependymomas in<br />

children. J Neurosurg 93:605-613<br />

7. Figarella-Branger D, Gambarelli D,<br />

Dollo C, et al (1991) Infratentorial<br />

ependymomas <strong>of</strong> childhood. Correlation<br />

between histological features, immunohistological<br />

fenotype, silver nucleolar<br />

staining values and post-operative survival<br />

in 16 cases; Acta Neuropathol<br />

82:208-216<br />

8. Foreman NK, Love S, Gill SS, Coakham<br />

HB (1997) Second-look surgery for incompletely<br />

resected fourth ventricle<br />

ependymomas: technical case report.<br />

Neurosurgery 40:856-860<br />

9. Foreman NK, Love S, Thorne R (1996)<br />

Intracranial ependymomas: analysis <strong>of</strong><br />

prognostic factors in a population-based<br />

series. Pediatr Neurosurg 24:119-125<br />

10. Geddes JF, Vowles GH, Ashmore SM<br />

(1994) Detection <strong>of</strong> multidrug resistance<br />

gene product (P-glycoprotein) expression<br />

in ependymomas. Neuropathol<br />

Appl Neurobiol 20(2):118-121<br />

11. Gerszten PC, Pollack IF, Martinez AJ<br />

(1996) Intracranial ependymomas <strong>of</strong><br />

childhood. Lack <strong>of</strong> correlation <strong>of</strong> histopathology<br />

and clinical outcome. Path<br />

Res Pract 192:515-522<br />

12. Goldwein JW, Corn BW, Finlay JL, et<br />

al (1991) Is craniospinal irradiation required<br />

to cure children with malignant<br />

(anaplastic) intractranial ependymomas<br />

Cancer 67:2766-2771<br />

13. Goldwein JW, Glause TA, Packer RJ,<br />

et al (1990) Recurrent intracranial<br />

ependymomas in children. Survival,<br />

patterns <strong>of</strong> feilure, and prognostic factors.<br />

Cancer 66: 557-563<br />

14. Grill J, Kalifa C, Doz F, et al (1996) A<br />

high-dose busulfan thiotepa combination<br />

followed by autologous bone marrow<br />

transplantation in childhood recurrent<br />

ependymoma. A phase II study.<br />

Pediatr Neurosurg 25:7-12<br />

15. Grill J, LeDeley MC, Gambarelli D, et<br />

al (2001) Postoperative chemotherapy<br />

without irradiation for ependymoma in<br />

children under 5 years <strong>of</strong> age: a<br />

multicenter trial <strong>of</strong> the French Society<br />

<strong>of</strong> Pediatric Oncology. J Clin Oncol<br />

19:1288-1296


16. Grill J, Pascal C, Chantal K (2003)<br />

Childhood ependymoma a systemic review<br />

<strong>of</strong> treatment options and strategies.<br />

Paediatr Drugs 5(8):533-543<br />

17. Grill J, Renaux VK, Bulteau C, et al<br />

(1999) Long-term intellectual outcome<br />

in children with posterior fossa tumors<br />

according to radiation dose and volumes.<br />

Int J Radiation Oncology Biol<br />

Phys 45:137-145<br />

18. Hoppe-Hirsch E, Brunet L, Laroussinie<br />

F, et al (1995) Intellectual outcome in<br />

children with malignant tumors <strong>of</strong> posterior<br />

fossa: influence <strong>of</strong> the field <strong>of</strong> irradiation<br />

and quality <strong>of</strong> surgery. Child’s<br />

Nerv Syst 11(6):340-345<br />

19. Horn B, Heideman R, Geyer R, et al<br />

(1999) A multiple-institutional retrospective<br />

study <strong>of</strong> intracranial ependymomas<br />

in children: identification <strong>of</strong> risk<br />

factors. J Pediatr Hematol Oncol<br />

21:203-211<br />

20. Ikezaki K, Matsushima T, Inoue T, et al<br />

(1993) Correlation <strong>of</strong> microanatomical<br />

localization with postoperative survival<br />

in posterior fossa ependymomas. Neurosurgery<br />

32(1):38-44<br />

21. Kordek R, Liberski PP (2001)<br />

Nowotwory gleju wyœció³kowego. Pol<br />

J Pathol 53, 4 (suppl):39-53 (in Polish)<br />

22. Mason WP, Goldman S, Yates AJ, et al<br />

(1998) Survival following intensive chemotherapy<br />

with bone marrow reconstruction<br />

for children with recurrent intracranial<br />

ependymoma – a report <strong>of</strong> the<br />

Children’s Cancer Group. J Neurooncol<br />

37:135-143<br />

23. McLaughlin MP, Marcus RB, Buatti<br />

JM, et al (1998) Ependymoma: results<br />

prognostic factors and treatment recommendations.<br />

Int J Radiation Oncology<br />

Biol Phys 40:845-850<br />

24. Palma L, Celli P, Mariottini A (2000)<br />

The importance <strong>of</strong> surgery in supratentorial<br />

ependymomas. Child’s Nerv Syst<br />

16:170-175<br />

25. Paulino AC, Bouffet E, Perilongo G, et<br />

al (1998) Intracranial ependymomas in<br />

children: a critical review <strong>of</strong> prognostic<br />

factors and a plea for cooperation. Medical<br />

and Pediatric Oncology 30:319-331<br />

26. Paulino AC (2001) The local field in<br />

infratentorial ependymoma: does the<br />

entire posterior fossa need to be treated<br />

Int J Radiation Oncology Biol Phys<br />

49:757-761<br />

27. Pollack IF, Gerszten PC, Martinez AJ,<br />

et al (1995) Intracranial ependymomas<br />

<strong>of</strong> childhood: long-term outcome and<br />

prognostic factors. Neurosurgery<br />

37(4):655-666<br />

28. Robertson PL, Zeltzer PM, Boyett JM,<br />

et al (1998) Survival and prognostic factors<br />

following radiation therapy and chemotherapy<br />

for ependymomas in children:<br />

a report <strong>of</strong> the Children’s Cancer Group.<br />

J Neurosurg. 88:695-703<br />

29. Rorke LB (1987) Relationship <strong>of</strong> morphology<br />

<strong>of</strong> ependymomas in children to<br />

prognosis. Prog Exp Tumor Res 30:170-<br />

174<br />

30. Ross GW, Rubinstein LJ (1989) Lack<br />

<strong>of</strong> histopathological correlation <strong>of</strong> malignant<br />

ependymomas with postoperative<br />

survival. J Neurosurg 70:31-36<br />

31. Schiffer D, Giordana MT (1998) Prognosis<br />

<strong>of</strong> ependymoma. Child’s Nerv<br />

Syst 14:357-361<br />

32. Seaver E, Geyer R, Sulzbacher S, et al<br />

(1994) Psychosocial adjustment in longterm<br />

survivors <strong>of</strong> childhood<br />

meduloblastoma and ependymoma<br />

treated with craniospinal irradiation.<br />

Pediatr Neurosurg 20(4):248-253<br />

33. Smyth MD, Horn BN, Russo C, et al<br />

(2000) Intracranial ependymomas <strong>of</strong><br />

childhood: current management strategies.<br />

Pediatr Neurosurg 33:138-150<br />

34. Suc E, Kalifa C, Brauner R, et al (1990)<br />

Brain tumors under the age <strong>of</strong> tree. The<br />

price <strong>of</strong> survival. Acta Neurochir<br />

106:93-98<br />

35. Sutton LN, Goldwein JW, Schwartz D<br />

(1999) Ependymoma. In: Albright AL,<br />

Pollack IF, Adelson PD (eds) Principles<br />

and practice <strong>of</strong> pediatric neurosurgery.<br />

Thieme, New York, Stuttgart, pp 609-<br />

628<br />

36. Teo Ch, Nakaji P, Symons P, et al (2003)<br />

Ependymoma. Child’s Nerv Syst<br />

19:270-285<br />

37. White L, Johnston H, Jones R, et al<br />

(1993) Postoperative chemotherapy<br />

without radiation in young children with<br />

malignant non-astrocytic brain tumors.<br />

A report from the Australia and New<br />

Zeland Childhood Cancer Study Group<br />

(ANZCCSG). Cancer Chemoter<br />

Pharmacol 32(5):403-406


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

<strong>Pathology</strong> <strong>of</strong> fatty liver in children with LCHAD deficiency<br />

Katarzyna Iwanicka 1 , Janusz Ksi¹¿yk 2 , Monika Pohorecka 3 , Maciej Pronicki 1<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

1<br />

Department <strong>of</strong> <strong>Pathology</strong>,<br />

2<br />

The Clinic <strong>of</strong> Pediatrics,<br />

3<br />

Department <strong>of</strong> Metabolic Diseases,<br />

The Children’s Memorial Health Institute,<br />

Warsaw, Poland<br />

Abstract<br />

Deficiency <strong>of</strong> long chain 3-hydroksyacyl-CoA dehydrogenase (LCHADD) impairs the process <strong>of</strong> mitochondrial<br />

beta-oxidation <strong>of</strong> fatty acids. LCHADD may be clinically benign up to the moment <strong>of</strong> first symptoms,<br />

i.e. fasting inducing the first acute life-threatening episode (ALTE). Patients present vomiting, lethargy, convulsions,<br />

arrhytmia and hypoketotic hypoglycemia, which is essential for established a diagnosis. Thus, after<br />

fasting, healthy looking child may die with serious symptoms <strong>of</strong> acute illness. The aim <strong>of</strong> this study was the<br />

histopathological assessment <strong>of</strong> liver in our own LCHADD patients with particular attention to characteristic<br />

changes including intensity, type and localization <strong>of</strong> liver steatosis. Seven liver samples were assessed: 3<br />

biopsies and 4 autopsy specimens obtained from 6 patients, aged from 3 months to one year. In 3 autopsies<br />

we found severe macrovacuolar fatty liver, in one biopsy moderate mixed steatosis, and in remaining 3 cases<br />

mild fatty liver <strong>of</strong> macrovacuolar and mixed character. All samples but one displayed diffuse distribution <strong>of</strong><br />

steatotic hepatocytes. In 4 cases steatosis was accompanied by fibrosis and cirrhosis, in 3 by inflammatory<br />

infiltrates. Intensity <strong>of</strong> steatosis was related to the severity <strong>of</strong> clinical symptoms. Fibrosis was another morphological<br />

feature characteristic for LCHAD deficiency. The results showed that liver steatosis found during<br />

autopsy <strong>of</strong> patients who died <strong>of</strong> unexplained reason should attract attention to LCHADD as a possible cause<br />

<strong>of</strong> death.<br />

Key words: LCHAD deficiency, liver steatosis<br />

Introduction<br />

Clinical symptoms <strong>of</strong> long chain 3-hydroksyacyl-CoA dehydrogenase<br />

(LCHAD) deficiency usually appear after prolonged<br />

fasting or could be evoked by upper respiratory tract infections,<br />

vaccination, physical exercises and other stress. Clinically<br />

healthy child presents hypoketotic hypoglycemia, which<br />

may be accompanied by vomiting, abdominal pain, convulsion,<br />

muscle weakness and cardiomegaly. In the course <strong>of</strong> the<br />

disease sensorimotor neuropathy and pigmentary retinopathy<br />

develop [5, 9, 11]. The age <strong>of</strong> onset ranges from one day to 40<br />

months. Treatment involves the diet with long chain fatty acids<br />

restriction and increased supply <strong>of</strong> carbohydrates.<br />

Fatty acids are the most important source <strong>of</strong> energy<br />

during fasting [5, 11]. They are transported into the inner<br />

membrane <strong>of</strong> mitochondria to be used in beta-oxidation process<br />

[5, 9, 11]. This process has especially important value<br />

for muscle, heart and liver cells as their own source <strong>of</strong> energy.<br />

Beta-oxidation process has two steps: transport and following<br />

oxidation. Three enzymes take parts in transport <strong>of</strong> fatty<br />

acids into the mitochondria, creating the pathway called carnitine<br />

transporting system [2, 3, 5, 12]. Proper beta-oxidation<br />

pathway depends on activity <strong>of</strong> several enzymes (four for long,<br />

four <strong>of</strong> medium and four for short chain fatty acids). The names<br />

<strong>of</strong> appropriate enzymes differ only by the name <strong>of</strong> length <strong>of</strong><br />

the chain. The basis <strong>of</strong> beta-oxidation process is shortening <strong>of</strong><br />

fatty acid chain by cutting acetyl-CoA to introduce it to tricarboxylic<br />

acids cycle. Deficiency <strong>of</strong> 3-hydroksyacyl-CoA dehydrogenase<br />

(LCHAD) impairs the process and leads to toxic<br />

accumulation <strong>of</strong> fatty acids and their acylcarnitines derivatives<br />

[2, 5, 7, 9, 11]. Accumulation <strong>of</strong> lipids in the cells may<br />

be a site effect <strong>of</strong> this metabolic decompensation.<br />

Address for correspondence<br />

Iwanicka Katarzyna<br />

Department <strong>of</strong> <strong>Pathology</strong><br />

The Children's Memorial Health Institute<br />

Aleja Dzieci Polskich 20,<br />

04-736 Warsaw, Poland<br />

Phone: +48 22 815 16 14<br />

E-mail: kasiai@yahoo.com


90<br />

Since 1994, twenty two unrelated children with LCHAD deficiency<br />

have been identified in Poland. Diagnosis was done<br />

on the analysis <strong>of</strong> GC-MS organic acid pr<strong>of</strong>ile in urine and<br />

than confirmed by genetic investigation. Common protocol<br />

<strong>of</strong> treatment and monitoring (CPTM) has been introduced in<br />

2000. There were 8 LCHAD-deficient children identified before<br />

CPTM. Diagnosis was markedly delayed in all cases,<br />

rarely established at first acute life-threatening episode<br />

(ALTE), frequently post mortem. Five <strong>of</strong> them died, only three<br />

patients were in good condition. In second period (since 2000)<br />

14 new LCHAD deficient cases were identified. The diagnosis<br />

was established usually at first ALTE, only in 4 patients<br />

too late (post mortem or post-episodic neurological sequels).<br />

As in the literature there are only few reports concerning<br />

histological pattern <strong>of</strong> the liver from patients with LCHAD<br />

deficiency [5, 7, 9, 11], the aim <strong>of</strong> our study was to analyse<br />

morphological findings in the liver <strong>of</strong> 6 patients with LCHAD<br />

deficiency to establish the spectrum <strong>of</strong> morphological changes<br />

and compare our results with the described reports.<br />

Results<br />

Case No 1<br />

The liver specimen has been obtained on autopsy. It revealed<br />

the diffused localization <strong>of</strong> mixed pattern <strong>of</strong> steatosis concerning<br />

<strong>of</strong> 100 % <strong>of</strong> hepatocytes. Apart <strong>of</strong> congection any<br />

other pathological change was not detected in the liver. Steatosis<br />

<strong>of</strong> cardiomyocytes and lipid accumulation in renal proximal<br />

tubules was observed as well (Fig. 1).<br />

Material and methods<br />

Material consists <strong>of</strong> seven liver samples obtained from six<br />

LCHAD deficient patients (4 boys and 2 girls). One <strong>of</strong> the<br />

patients underwent liver biopsy at the age <strong>of</strong> 3 months and<br />

post mortem examination at 6 months. There were three biopsy<br />

samples obtained from the LCHAD-deficient boys and<br />

four autopsy specimens derived from affected patients (two<br />

boys and two girls). Patients were aged from 3 to 12 months.<br />

Clinical diagnosis <strong>of</strong> LCHAD deficiency was based on urine<br />

organic acid pr<strong>of</strong>ile analysis using GC-MS method (Dept <strong>of</strong><br />

Laboratory <strong>Diagnostic</strong>s, CMHI), and confirmed by molecular<br />

study.<br />

The liver samples obtained during biopsies were prepared<br />

for light and electron microscopy. Slides were stained<br />

with hematoxylin and eosin, PAS, diastase digested PAS, for<br />

collagen and reticulin fibres – AZAN and Gomori stains, respectively.<br />

Semi-thin slides <strong>of</strong> plastic embedded (epon) liver<br />

samples post fixed in osmium tetroxide and stained with toluidine<br />

blue were used for steatosis detection. Liver tissue samples<br />

obtained during autopsies were processed routinely for histology<br />

and stained with hematoxylin and eosin.<br />

During microscopical assessment we assumed three<br />

grades <strong>of</strong> steatosis intensity: mild, moderate and severe, when<br />

lipid droplets comprised up to 30%, 30-70%, and over 70%<br />

<strong>of</strong> liver parenchyma, respectively. Two types <strong>of</strong> distribution<br />

<strong>of</strong> steatotic hepatocytes were recorded: diffuse or focal. Type<br />

<strong>of</strong> steatosis included three categories: macrovacuolar,<br />

microvacuolar, and mixed. Semi-thin slides are very helpful<br />

in correct lipid detection, especially <strong>of</strong> microvacuolar type.<br />

Accompanying inflammatory infiltrates and fibrosis were assessed<br />

according to routine grading and staging system.<br />

Fig. 1 Liver biopsy <strong>of</strong> patient no 1 reveals severe mixed steatosis. Presence<br />

<strong>of</strong> lipid accumulation in renal proximal tubules (<strong>of</strong> patient no 1).<br />

Case No 2<br />

The liver specimen has been obtained on autopsy. It revealed<br />

the typical pattern <strong>of</strong> fatty liver. Macrovesicular lipid droplets<br />

are present in almost all hepatocytes. Mild fibrosis was present<br />

only in periportal areas. Inflammatory infiltrates and<br />

cholestasis were not detected. There was found vacuolisation<br />

corresponding probably to lipid accumulation in proximal renal<br />

tubules (Fig. 2).


91<br />

Fig. 2 Liver biopsy <strong>of</strong> patient no 2 shows severe diffuse macrovesicular steatosis.<br />

Liver biopsy <strong>of</strong> patient no 3 shows diffuse, moderate, mixed steatosis.<br />

Mild intralobular inflamatory infiltrates <strong>of</strong> lymphoid cells and single necrotic<br />

hepatocytes are observed.<br />

Case No 3<br />

The liver tissue obtained during biopsy showed fatty changes<br />

in approximately 60 % <strong>of</strong> hepatocytes. They revealed diffuse<br />

distribution. Lipid droplets in hepatocytes were <strong>of</strong> mixed pattern:<br />

macrovacuoles and microvacuoles were present. The mild<br />

intralobular and portal inflamatory infiltrates <strong>of</strong> lymphoid cells<br />

were found. Single necrotic hepatocytes were observed. Fibrosis<br />

and cholestasis were not detected (Fig. 2).<br />

Case No 4<br />

The liver biopsy specimen obtained prior to autopsy revealed<br />

precirrhotic pattern <strong>of</strong> liver injury. Macrovesicular steatotis were<br />

seen in 30 % <strong>of</strong> hepatocytes. Their localization was rather focal,<br />

within some <strong>of</strong> the nodules. Severe portal inflammatory<br />

infiltrates were present. Cholestasis was not found (Fig. 3).<br />

Case No 5 (the same patient as above)<br />

Subsequent liver specimen obtained during autopsy revealed<br />

cirrhosis. Severe macrovesicular fatty changes concerned all<br />

hepatocytes. Mild portal inflammatory infiltrates remained<br />

similar to those <strong>of</strong> the biopsy specimen. No cholestasis was<br />

present (Fig. 3).<br />

Fig. 3 Precihrrotic pattern <strong>of</strong> moderate, focal, macrovesicular steatosis <strong>of</strong><br />

patient 4. Subsequent, post mortem examination <strong>of</strong> the same patient reveals<br />

severe, macrovesicular fatty changes.<br />

Case No 6<br />

The liver biopsy differed substantially from other patients.<br />

Diffuse, mixed steatotic changes were seen only in about 5-<br />

10% <strong>of</strong> hepatocytes. No inflammatory infiltrates, fibrosis or<br />

cholestasis were detected in the liver.<br />

Case No 7<br />

The girl was referred to autopsy examination with diagnosis<br />

<strong>of</strong> cardiac insufficiency due to hypertrophied cardiomyopathy.<br />

Appropriate diagnosis was established retrospectively,<br />

some years after death on the basis on genetic family study. It<br />

was performed after her younger brother was diagnosed to<br />

have LCHAD deficiency. On autopsy examination features <strong>of</strong><br />

excentric hypertrophy <strong>of</strong> left cardiac ventricle with accompanying<br />

signs <strong>of</strong> mitral insufficiency and endocardial thickening<br />

was observed. Steatosis <strong>of</strong> the liver was also reported in<br />

autopsy protocol, but no morphological characteristic was<br />

made. Microscopically, macrovesicular steatosis <strong>of</strong> diffuse localization<br />

consisted <strong>of</strong> 10 % <strong>of</strong> hepatocytes accompanied by<br />

mild fibrosis was found. No inflammation and cholestasis was<br />

observed.


92<br />

Summary<br />

Liver steatosis <strong>of</strong> severe degree involving 100% <strong>of</strong> liver parenchyma<br />

was found in three patients (No 1, 2, and 5) who<br />

died during ALTE. Moderate steatosis involving approximately<br />

60% <strong>of</strong> liver was present in one biopsy ( No 3). Mild steatosis<br />

affecting 30% and 15% <strong>of</strong> liver parenchyma was found in two<br />

biopsies coming from cases 4 and 6, respectively. In one autopsy<br />

sample liver steatosis involved 10% <strong>of</strong> parenchyma (No<br />

7). Steatosis was macrovacuolar or mixed. The first type was<br />

found in two sequential examinations <strong>of</strong> the same patient (biopsy<br />

– No 4 and autopsy – No 5) and in two autopsies (samples<br />

No 2 and 7). Macrovacuolar steatosis was severe in two cases<br />

(No 2 and 5) and mild in two others (No 4 and 7). Mixed<br />

steatosis was seen in two biopsies (No 3 and 6) and one autopsy<br />

(No 1). Its degeree was mild (sample No 6), moderate<br />

(sample No 3), and severe (sample No 1). None <strong>of</strong> the patients<br />

had pure microvacuolar fatty liver. In six cases distribution<br />

<strong>of</strong> steatotic hepatocytes was diffuse, in one focal (sample<br />

No 4). Inflammation was present in three cases. It was mild<br />

(cases No 3 and 5) or severe (case No 4). Fibrosis was disclosed<br />

in four examined samples derived from three patients:<br />

in two cases its intensity was mild (No 2 and 7) in two severe<br />

(samples No 4 and 5). Liver biopsy samples available for assessment<br />

from the same patient disclosed progression from<br />

precirrhosis to cirrhosis. Data concerning patients and their<br />

liver morphology are presenting in Table 1.<br />

Discussion<br />

It is known that the process <strong>of</strong> mitochondrial fatty acid oxidation<br />

can be divided in two stages. First involves transport<br />

through mitochondrial membrane, the second consists <strong>of</strong> cutting<br />

<strong>of</strong>f two-carbon moieties from fatty acid chain by several<br />

enzymes for long, medium, and short chain fatty acids, respectively<br />

[5, 9, 11]. Fatty liver may develop in both patients<br />

with defects <strong>of</strong> the first stage involving carnitine dependent<br />

transport [2, 3, 12], and with defects <strong>of</strong> the proper beta-oxidation<br />

[2, 4, 6-8, 10, 11]. Baldridge in his study <strong>of</strong> 82 patients<br />

with liver steatosis reported only one child in whom the<br />

LCHAD deficiency was established [1]. Our group comprises<br />

six patients with this diagnosis. Roe and Coates provided general<br />

description <strong>of</strong> liver morphology in LCHAD deficiency<br />

[9]. They pointed lipid accumulation and sometimes accompanying<br />

fibrosis as characteristic features <strong>of</strong> this disorder. Our<br />

findings are consistent with this observation. However, they<br />

have not provided particulars concerning intensity, type, and<br />

distribution <strong>of</strong> steatosis. Gilbert-Barness and Barness have<br />

presented more detailed description <strong>of</strong> liver microscopical<br />

pathology in LCHAD deficient patients [5]. They considered<br />

diffuse or zonal macrovacuolar severe fatty liver to be characteristic<br />

<strong>of</strong> the majority <strong>of</strong> LCHAD-deficient patients. In our<br />

group this type <strong>of</strong> morphological pattern was found in three<br />

patients always when the liver biopsy was performed at the<br />

acute clinical presentation.<br />

In a number <strong>of</strong> described patients with LCHAD deficiency<br />

the liver discloses only focal steatosis <strong>of</strong> mild intensity<br />

[5]. We observed focal fatty changes only in one case out <strong>of</strong><br />

seven (sample No 4). In remaining patients the degree <strong>of</strong> steatosis<br />

was mild to moderate and was almost invariably diffusely<br />

distributed.<br />

Moore described morphology <strong>of</strong> the liver in six months<br />

old girl with LCHAD deficiency [7]. There were mixed steatosis<br />

accompanied by mild cholestasis. Author has not provided<br />

data concerning intensity and localization <strong>of</strong> fatty<br />

changes. In our series <strong>of</strong> seven cases we did not find any case<br />

with the features <strong>of</strong> cholestasis.<br />

Tyni described liver morphology including microscopical<br />

findings <strong>of</strong> 16 patients coming from 12 families, who died<br />

with diagnosis <strong>of</strong> LCHAD deficiency [11]. All patients in her<br />

study had features <strong>of</strong> macrovacuolar fatty liver. In our patients<br />

steatosis was not only macrovacuolar, but also mixed. Tyni<br />

concluded that steatosis was typical for fatty acid beta oxidation<br />

defects, and emphasized that accompanying fibrosis was<br />

Table 1<br />

Details <strong>of</strong> morphological changes <strong>of</strong> the liver in patients with LCHADD<br />

Type <strong>of</strong><br />

examination<br />

Sample<br />

No<br />

Age<br />

(mo)<br />

Intensity <strong>of</strong><br />

steatosis (%)<br />

Type <strong>of</strong><br />

steatosis<br />

Localization<br />

<strong>of</strong> steatosis<br />

Inflammation<br />

Fibrosis Cholesta<br />

A 3299 4 100 mixed diffuse no no no<br />

A 3289 3 100 macro diffuse no mild no<br />

B 30573 12 60 mixed diffuse mild no no<br />

B 30760 3 30 macro focal severe precirrhosis no<br />

A 2701 6 100 macro diffuse mild cirrhosis no<br />

B 51605 7 15 mixed diffuse no no no<br />

A 3070 6 10 macro diffuse no mild no


93<br />

especially typical for LCHAD deficiency [11]. In our series<br />

the fibrosis was found in four out <strong>of</strong> seven liver samples (obtained<br />

from three children). Tyni suggested presence <strong>of</strong> relationship<br />

between the severity <strong>of</strong> clinical course and the intensity<br />

<strong>of</strong> morphological changes in the liver [11]. Our observations<br />

are in agreement with this statement.<br />

Our observations strongly underline that liver steatosis<br />

<strong>of</strong> any degree detected at autopsy <strong>of</strong> a child died without<br />

clearly established diagnosis obliges to include fatty acids<br />

oxidation disorders into the differential diagnosis [2]. One <strong>of</strong><br />

our patient, died many years ago with the diagnosis <strong>of</strong> hypertrophied<br />

cardiomyopathy, was detected just recently as<br />

LCHAD-deficient during genetic family study, after establishing<br />

LCHAD deficiency in her younger brother. Lack <strong>of</strong> acute<br />

life threatening episode in her medical history and only mild<br />

degree <strong>of</strong> liver steatosis in her post mortem examination, preliminarily<br />

did not paid our attention and missed from correct<br />

diagnosis.<br />

In general our results are consistent with observations <strong>of</strong> other<br />

authors and warrant drawing the following conclusions:<br />

1. Fatty liver is characteristic <strong>of</strong> LCHAD deficiency.<br />

2. Intensity <strong>of</strong> steatosis is related to severity <strong>of</strong> clinical symptoms<br />

3. In patients with mitochondrial beta-oxidation defects liver<br />

steatosis may be accompanied by fibrosis which is suggestive<br />

<strong>of</strong> LCHAD deficiency.<br />

4. Fatty liver found in a patient who died <strong>of</strong> undiagnosed disease<br />

should suggest the possibility <strong>of</strong> LCHAD deficiency<br />

as a cause <strong>of</strong> death.<br />

5. Semi-thin plastic embedded tissue section postfixed in osmium<br />

tetroxide and stained with toluidine blue is very helpful<br />

in reliable assessment <strong>of</strong> liver steatosis<br />

Acknowledgments<br />

The study was supported by a statutory grant No S 86 <strong>of</strong> The<br />

Children’s Memorial Health Institute<br />

References<br />

1. Baldridge AD, Perez-Atayde AR<br />

Graeme-Cook F, Higgins L, Lavine JE<br />

(1995) Idiopathic steatohepatitis in<br />

childchood A multicenter study. J<br />

Pediatr 127:700-704<br />

2. Chace DH, DiPerna JC, Mitchell BL,<br />

Sgroi B, H<strong>of</strong>man LF, Naylor EW (2001)<br />

Electrospray Tandem Mass Spectrometry<br />

or Analysis <strong>of</strong> Acylcarnitines in Dried<br />

Postmortem Blood Spcimens Collcted at<br />

Autopsy from Infants with Unexplained<br />

Cause <strong>of</strong> Death. Clin Chemist<br />

47(7):1166-1182<br />

3. Chalmers RA, Stanley CA, English N,<br />

Wigglesworth JS (1997) Mitochondrial<br />

carnitine-acylcarnitine translocase deficiency<br />

presenting as sudden neonatal<br />

death. J Pediatr 131:220-225<br />

4. Dawson DB, Waber L, Hale DE,<br />

Bennett MJ (1995) Transient organic<br />

aciduria and persistent lacticacidemia in<br />

a patient with short-chain acyl-coenzyme<br />

A dehydrogenase deficiency. J<br />

Pediatr 126:69-71<br />

5. Gilbert-Barness and Barness (2000)<br />

Fatty acids beta-oxidation defects. In:<br />

Gilbert-Barness and Barness (eds)<br />

Metabolic Diseases Eaton Publishing,<br />

Natick, pp113-136<br />

6. Iafolla AK, Thompson RJ, Roe CR<br />

(1994) Medium-chain acyl-coenzyme A<br />

dehydrogenase deficiency: Clinical<br />

course in 120 affected children. J Pediatr<br />

124:409-415<br />

7. Moore R, Glasgow JFT, Bingham MA,<br />

Dode JA, Pollitt RJ,Olpin SE, Middleton<br />

B, Carpentier K (1993) Long chain 3-<br />

hydroxyacyl-coenzyme A dehydrogenase<br />

deficiency – diagnosis,<br />

plasmacarnitine fractions and management<br />

in a further patient. Eur J Pediatr<br />

152:433-6<br />

8. Roe CR Millington DS, Maltby DA<br />

Kinnebrew P (1986) Recognition <strong>of</strong> medium-chain<br />

acyl-CoA dehydrogenase<br />

deficiency in asymptomatic siblings <strong>of</strong><br />

children dying <strong>of</strong> sudden infant death<br />

or Reye-like syndromes. J Pediatr<br />

108:13-18<br />

9. Roe CR, Coates PM (1995) Mitochondrial<br />

fatty acids oxidation disorders. In:<br />

Scriver CR, Beaudet AL, Sly WS, Valle<br />

D (eds) The metabolic and molecular<br />

basis <strong>of</strong> inherited disease. McGraw-Hill,<br />

New York, pp 1501-1535<br />

10. Santer R, Schmidt-Sommerfeld E,<br />

Leung YK, Fischer JE, Lebental E<br />

(1990) Medium-chain acyl-CoA dehydrogenase<br />

deficiency: electron microscopic<br />

differentiation from Reye syndrome.<br />

Eur J Pediatr 150:111-114<br />

11. Tyni T, Rapola J, Paetau A, Palotie A,<br />

Pihko H (1997) Patology <strong>of</strong> long chain<br />

3-hydroxyacyl-CoA dehydrogenase deficiency<br />

causes by the G1528C mutation.<br />

Ped Path Lab Med 17:427-447<br />

12. Winter SC, Szabo-Aczel S, Curry CJR,<br />

Hutchinson HT, Hogue R, Shug A<br />

(1987) Plasma carnitine deficiency<br />

Clinical Observations in 51 Pediatric<br />

Patients. AJDC 141:660-665


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Isolation <strong>of</strong> urothelial cells for tissue engineered<br />

bladder augmentation<br />

Tomasz Drewa 1, 2 , Przemys³aw Galazka 1, 3 , Zbigniew Wolski 2 , Andrzej I. Prokurat 3 , Jan Sir 4<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

1<br />

Department <strong>of</strong> Biology,<br />

2<br />

Department <strong>of</strong> Urology,<br />

3<br />

Department <strong>of</strong> Pediatric Surgery,<br />

The L. Rydygier Medical University in Bydgoszcz,<br />

Bydgoszcz, Poland<br />

4<br />

Department <strong>of</strong> <strong>Pathology</strong>,<br />

Oncological Center in Bydgoszcz,<br />

Bydgoszcz, Poland<br />

Abstract<br />

Objectives. In case <strong>of</strong> certain human congenital anomalies such as bladder extrophies, there may not be<br />

enough residual bladder for closure. Tissue engineering is a promising method for bladder augmentation.<br />

The aim <strong>of</strong> our study was to compare isolation efficiency <strong>of</strong> various digestive mixtures and establish isolation<br />

and cell culture methodology <strong>of</strong> urothelium. Different variants <strong>of</strong> growth media and their influence on proliferative<br />

capacity was assessed. Methods. Primary Rabbit Urothelial Cells (PRUC) culture was established<br />

from 8 months old male rabbit urinary bladder wall. 5 different digesting mixtures for isolation <strong>of</strong> urothelial<br />

cells were tested. Cells obtained from 0.1% collagenase I digestion after 1 and 4 hours were checked for their<br />

growth potential in 3 different media. Results. The best digestion solution for urothelial cells isolation was<br />

0.1% collagenase I. It can be noticed that the cell number <strong>of</strong> all secondary cultures was higher after 4 h<br />

digestion than after 1 h. The medium substrates also influenced total cell number in urothelial subcultures.<br />

The best foetal bovine serum (FBS) supplementation was 5%, when the medium was additionally enriched<br />

with micro-supplements. Conclusions. Despite <strong>of</strong> enzymes used for isolation, the digestion time and medium<br />

enriched with properly amount <strong>of</strong> serum both deeply influence on the outcome <strong>of</strong> cell therapy. Autologous<br />

cells could be used for urinary tract reconstruction using tissue engineering methods.<br />

Key words: tissue engineering, urinary tract reconstruction, urothelial cells<br />

Introduction<br />

In case <strong>of</strong> certain human congenital anomalies such as bladder<br />

extrophies, there may not be enough residual bladder for closure<br />

[7]. The autologous materials and techniques have been<br />

proposed for bladder augmentation with small and large bowel,<br />

stomach [1, 13]. Unfortunately none <strong>of</strong> used materials has been<br />

enough satisfactory [9, 17]. A number <strong>of</strong> biomaterials have been<br />

used in experimental models including acellular tissue matrix<br />

grafts, synthetic biodegradable or non-biodegradable polymers<br />

[2]. Many reports revealed failure <strong>of</strong> these attempts because <strong>of</strong><br />

recurrent urinary tract infections, marked contracture <strong>of</strong> the graft<br />

or graft rejection and stones formation [5, 12, 15, 17].<br />

Tissue engineering is a promising method for bladder augmentation<br />

[7]. The use <strong>of</strong> an autologous cultured urothelium for<br />

bladder reconstruction could prevent many <strong>of</strong> the problems associated<br />

with applied materials [4]. Tissue engineered bladder<br />

wall consisting <strong>of</strong> a adequate shaped biodegradable polymer<br />

used as a scaffold, can deliver autologous urothelial cells [11].<br />

This device may restore bladder capacity more effective than<br />

biomaterials alone. Atala et al demonstrated that urothelial cells<br />

seeding on the bioabsorbable scaffold can allow creation <strong>of</strong> new<br />

functional urinary bladder wall [3].<br />

Address for correspondence<br />

Tomasz Drewa, MD, PhD<br />

Dpt. <strong>of</strong> Medical Biology,<br />

the L. Rydygier Medical University in Bydgoszcz<br />

Ul. Karlowicza 24<br />

85-094 Bydgoszcz, Poland<br />

E-mail: tomaszdrewa@wp.pl


96<br />

The aim <strong>of</strong> our study was to compare isolation efficiency <strong>of</strong><br />

various digestive mixtures and establish proper isolation methodology<br />

<strong>of</strong> urothelial cells. Also different variants <strong>of</strong> growth<br />

medium and its influence on proliferative capacity <strong>of</strong> growing<br />

cells was assessed. In the future these cells can be used for urinary<br />

tract reconstruction using tissue engineering methods.<br />

Methods<br />

Primary Rabbit Urothelial Cells (PRUC) culture was established<br />

from 8 months old male rabbit urinary bladder wall.<br />

After ketamine (25 mg/kg m.c. i.m.) and scoline (50 mg/kg)<br />

overdosage bladder wall sample (5 x 5 cm) was obtained. Fragment<br />

was immersed in sterile Phosphate Buffered Saline (PBS,<br />

Biomed). In the next step urinary mucosa was mechanically<br />

separated from muscle layer. At this stage samples were cut<br />

into 12 equal sized parts (1 cm 2 each). Mucosa samples were<br />

cut into 2 mm 3 pieces.<br />

First <strong>of</strong> all 5 different digesting mixtures for isolation<br />

<strong>of</strong> urothelial cells were tested. Incubation time was 4 hours.<br />

Digesting solutions were as follow:<br />

1. 0.125% trypsin and 0.01% EDTA<br />

2. 0.08 % trypsin and 0.006% EDTA<br />

3. 0.06 % trypsin and 0.005% EDTA<br />

4. 0.05% trypsin and 0.02% EDTA<br />

5. 0.1% collagenase-I<br />

After incubation time the cell pellet was centrifuged<br />

(800 g/5 min), resuspended in each <strong>of</strong> the tested culture media<br />

and seeded on 25 cm 2 culture dishes (Greiner). Cells were<br />

identified as epithelial cells by morphological criteria as well<br />

as presence <strong>of</strong> cytokeratins (Anti-Cytokeratin, Clone: MNF<br />

116, DAKO), after three passages.<br />

In the next step 2 nd PRUC was established using the<br />

most efficient digestive solution: 0.1% collagenase I. Digestion<br />

efficiency was assessed using two incubation times (1<br />

and 4 hours). Cells obtained from 0.1% collagenase I digestion<br />

after 1 and 4 hours were checked for their growth potential<br />

in 3 different media:<br />

I- DMEM supplemented with 10% FBS and antibiotics: penicillin<br />

(100 IU/ml) (Polfa) and streptomycin (100 ug/ml) (Polfa)<br />

II- 3:1 DMEM and F-12 mixture supplemented with 5% FBS,<br />

epidermal growth factor (EGF, Sigma), bovine pituary extract<br />

(Sigma), cholera toxin (30 ng/ml) (Sigma), penicillin (100<br />

IU/ml) (Polfa) and streptomycin (100 ug/ml) (Polfa)<br />

III- 1:1 DMEM and F-12 mixture supplemented with 2.5%<br />

FBS, epidermal growth factor (EGF, Sigma), bovine pituary<br />

extract (Sigma), cholera toxin (30 ng/ml) (Sigma), penicillin<br />

(100 IU/ml) (Polfa) and streptomycin (100 ug/ml) (Polfa).<br />

Cells were seeded on 25 cm 2 culture dishes (Greiner)<br />

and grown in 37 o C in a humidified atmosphere with 5% CO 2<br />

.<br />

Cells were counted using trypan blue exclusion test after four<br />

days <strong>of</strong> cultivation. Neubauer«s cytologic chamber was used<br />

for counting. Morphology was examined under inverted microscope.<br />

Photographic documentation was done.<br />

Mean values (+/-SD) from 10 counts for each culture were<br />

compared to each other. The Student test was used for statistical<br />

analysis. A p value


97<br />

3<br />

2<br />

1<br />

0<br />

x10 6 cells<br />

1 hour 4 hours<br />

Fig. 2 Digestion efficiency using 0.1% collagenase I solution was assessed<br />

using two incubation times (1 and 4 hours)<br />

4<br />

3<br />

2<br />

1<br />

0<br />

x10 6 cells<br />

A B C D E F<br />

Fig. 3 Cells obtained from 0.1% collagenase I digestion after 1 hour (a,b,c)<br />

and 4 hours (d,e,f) were checked for their growth potential in 3 different<br />

media as: - DMEM with 10% FBS (a,d); - 3:1 DMEM and F-12 mixture<br />

enriched with microsupplements and 5% FBS (b,e); - 1:1 DMEM and F-12<br />

mixture enriched with microsupplements and 2.5% FBS (c,f). Cells were<br />

counted after 4 days <strong>of</strong> culturing (A vs D, p


References<br />

1. Adams MC, Mitchell ME, Rink RC<br />

(1882) Gastrocystoplasty: an alternative<br />

solution to the problem <strong>of</strong> urological reconstruction<br />

in the severely compromised<br />

patient. J Urol 140:1152-1156<br />

2. Atala A (1997) Tissue engineering in<br />

the genitourinary system. In: Tissue engineering,<br />

Atala A, Mooney D (eds)<br />

Birhauser Press, Boston, MA, pp. 149-<br />

164<br />

3. Atala A (2002) Future trends in bladder<br />

reconstructive surgery. Semin Pediatr<br />

Surg 11:134-142<br />

4. Atala A, Vacanti JP, Peters CA, Mandell<br />

J, Retik AB, Freeman MR (1992) Formation<br />

<strong>of</strong> urothelial structures in vivo<br />

from dissociated cells attached to biodegradable<br />

polymer scaffolds in vitro.J<br />

Urol 148:658-662<br />

5. Cain MP, Casale AJ, Kaefer M, Yerkes<br />

E, Rink RC (2002) Percutaneous<br />

cystolithotomy in the pediatric augmented<br />

bladder. J Urol 168(4 Pt<br />

2):1881-1882<br />

6. Drewa T, Wolski Z, Galazka P, Sir J,<br />

WoŸniak A, Czaplewski A (2004) Preliminary<br />

study on alginate scaffold application<br />

for chondrocytes transplantation<br />

into the rat bladder wall.<br />

Biotechnologia 3(66):213-224<br />

7. Fauza DO, Fishmann SJ, Mehegan K,<br />

Atala A (1998) Videoscopically assisted<br />

fetal tissue engineering: bladder augmentation.<br />

J Ped Surg 33(1):7-12<br />

8. Follmann W, Guhe C, Weber S, Birkner<br />

S, Mahler S (2000) Cultured porcine urinary<br />

bladder epithelial cells as a screening<br />

model for genotoxic effects <strong>of</strong> aromatic<br />

amines: characterisation and application<br />

<strong>of</strong> the cell culture model. Altern<br />

Lab Anim 28(6):833-854<br />

9. Gough DC (2001) Enterocystoplasty.<br />

BJU 88(7):739-743<br />

10. Hobbiesiefken J, Ehlers ME, Behrens<br />

P, Wunsch L (2003) Urothelial mesh- a<br />

new technique <strong>of</strong> cell culture on<br />

biomaterials. Eur J Pediatr Surg<br />

13(6):361-366<br />

11. Kim BS, Baez CE, Atala A (2000)<br />

Biomaterials for tissue engineering.<br />

World J Urol 18(1):2-9<br />

12. Mingin GC, Stock JA, Hanna MK<br />

(1999) Gastrocystoplasty: long-term<br />

complications in 22 patients. J Urol<br />

162(3 Pt 2):1122-1125<br />

13. Motley RC, Montgomery BT, Zollman<br />

PE, Holley KE, Kramer SA (1990) Augmentation<br />

cystoplasty utilizing de-epithelialized<br />

sigmoid colon: a preliminary<br />

study. J Urol 143:1257-1260<br />

14. Rebel JM, De Boer WI, Thijssen CD,<br />

Vermey M, Zwarth<strong>of</strong>f EC, Van der<br />

Kwast TH (1994) An in vitro model <strong>of</strong><br />

urothelial regeneration: effects <strong>of</strong><br />

growth factors and extracellular matrix<br />

proteins. J Pathol. 173(3):283-291<br />

15. Sugasi S, Lesbros Y, Bisson I, Zhang YY,<br />

Kucera P, Frey P (2000) In vitro engineering<br />

<strong>of</strong> human stratified urothelium: analysis<br />

<strong>of</strong> its morphology and function. J Urol<br />

164(3 Pt 2):951-957<br />

16. Wechselberger G, Schoeller T, Stenzl A,<br />

Ninkovic M, Lille S, Russell RC (1998)<br />

Fibrin glue as a delivery vehicle for autologous<br />

urothelial cell transplantation<br />

onto a prefabricated pouch. J Urol<br />

160(2):583-586<br />

17. Woodhouse CR, Redgrave NG (1996)<br />

Late failure <strong>of</strong> the reconstructed exstrophy<br />

bladder. Br J Urol 77(4):590-592<br />

18. Zhang YY, Ludwikowski B, Hurst R,<br />

Frey P (2001) Expansion and long-term<br />

culture <strong>of</strong> differentiated normal rat<br />

urothelial cells in vitro. In Vitro Cell Dev<br />

Biol Anim 37(7):419-429


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Status <strong>of</strong> oral hygiene and dentition in children<br />

exposed to anticancer treatment<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

Dorota Olczak-Kowalczyk 1 , Marta Daszkiewicz 1 , Barbara Adamowicz-Klepalska 2 , El¿bieta Czarnowska 3 ,<br />

Agnieszka Sowiñska 3 , Bo¿ena Dembowska-Bagiñska 4 , Danuta Perek 4 , Pawe³ Daszkiewicz 5<br />

1<br />

Department <strong>of</strong> Oral <strong>Pathology</strong>,<br />

3<br />

Department <strong>of</strong> <strong>Pathology</strong>,<br />

4<br />

Department. <strong>of</strong> Oncology,<br />

5<br />

Department <strong>of</strong> Neurosurgery,<br />

The Children’s Memorial Health Institute,<br />

Warsaw, Poland<br />

2<br />

Department <strong>of</strong> Developmental Age Stomatology,<br />

Medical Academy,<br />

Gdañsk, Poland<br />

Abstract<br />

Background: Deleterious effects <strong>of</strong> radiation and chemotherapy on oral cavity tissues are well known, but<br />

little data exist on the effect <strong>of</strong> combined anticancer treatment on developing teeth. Additionally, there are no<br />

established standards for dental care in children undergoing complex anticancer treatment. The aim <strong>of</strong> study:<br />

Evaluation <strong>of</strong> oral hygiene and dentition status in children subjected to combined anticancer treatment. Material<br />

and method: 88 children (age range 3-18 years), who underwent combined chemo-radiotherapy or<br />

chemotherapy alone due to a malignant neoplasm, were included to the study. Follow-up time was from 6<br />

months to 5 years. Patients were assessed in the dental out-patients’ clinic and their medical documentation<br />

reviewed. Results and conclusions: An inadequate oral hygiene was found in all patients as well as a severe<br />

caries and an increased incidence <strong>of</strong> non-caries-mediated enamel defects. A deleterious effect <strong>of</strong> combined<br />

chemo-radiotherapy on mineralized tooth tissues was found, both in primary and in permanent teeth. Ionizing<br />

radiation had a stronger negative impact than chemotherapy alone. In all children analyzed, there was a<br />

discrepancy between actual requirements for dental care and currently performed dental treatment.<br />

Key words: children, chemotherapy, enamel structure, oral hygiene, permanent dentition, primary dentition,<br />

radiotherapy<br />

Introduction<br />

Anticancer treatment, including chemotherapy and radiotherapy<br />

<strong>of</strong> the facial region, may enhance both the development<br />

<strong>of</strong> dental caries and also <strong>of</strong> non-caries dependant lesions<br />

on smooth surfaces <strong>of</strong> teeth [4-6, 10, 11]. The etiology <strong>of</strong> these<br />

phenomena is multi-factorial and still poorly understood. Both<br />

chemo- and radiotherapy increase the susceptibility <strong>of</strong> teeth<br />

to noxious factors and affect adversely the ecosystem <strong>of</strong> oral<br />

cavity [9, 10]. In patients undergoing combined anticancer<br />

treatment, these adverse effects may enhance each other [7].<br />

In general opinion, the direct effect <strong>of</strong> ionizing radiation<br />

on teeth leads to denaturation <strong>of</strong> protein matrix <strong>of</strong> the dentin<br />

and to damage <strong>of</strong> its structure due to degeneration and dysfunction<br />

<strong>of</strong> dental pulp. On the other hand, there is much less<br />

information concerning the pathogenesis <strong>of</strong> enamel lesions in<br />

Address for correspondence<br />

Dorota Olczak-Kowalczyk<br />

Department <strong>of</strong> Oral <strong>Pathology</strong><br />

The Children's Memorial Health Institute<br />

Al. Dzieci Polskich 20<br />

04-736 Warsaw, Poland<br />

Phone : +4822 815 13 15; +4822 815 16 31<br />

E-mail : dok.@bobas.czd.pl


100<br />

available literature. Enamel is described as poorly mineralized,<br />

less hard and more brittle, or as more susceptible to damage by<br />

acids while preserving normal hardness. Independent on the<br />

mechanism <strong>of</strong> radiation-induced lesions in mineralized tissues<br />

<strong>of</strong> teeth, they become more susceptible to harmful factors [5].<br />

There is little data concerning the effects <strong>of</strong> chemotherapy not<br />

combined with radiotherapy on teeth. Nevertheless, they show<br />

its deleterious effect on the function <strong>of</strong> dental pulp, thereby<br />

increasing susceptibility <strong>of</strong> teeth to various pathologic processes<br />

[10]. The effect <strong>of</strong> particular cytostatics on tooth pulp is difficult<br />

to ascertain. This is due to general use <strong>of</strong> multi-drug chemotherapy<br />

regimens. Toxic effects <strong>of</strong> vincristin are known the<br />

best. Histopathologic examination <strong>of</strong> teeth extracted from patients<br />

undergoing multi-drug chemotherapy revealed linear lesions,<br />

most probably resulting from disturbed production <strong>of</strong><br />

collagen matrix by odontoblasts. An important point is finding<br />

<strong>of</strong> correlation between the number and distribution <strong>of</strong> these lines<br />

and chemotherapy cycles, when one <strong>of</strong> administered drugs was<br />

vincristin [6]. It is also well known that colchicin and vinblastin<br />

block dentin formation in incisors in rats, while trethanomelamin<br />

and cyclophosphamide delay cutting <strong>of</strong> molars in these animals<br />

[6, 7].<br />

Adverse change <strong>of</strong> oral eco-system in patients undergoing<br />

anticancer treatment is caused mainly by qualitative and<br />

quantitative disturbances <strong>of</strong> salivary secretion [9]. Facial irradiation<br />

<strong>of</strong>ten leads to dysfunction <strong>of</strong> serous cells <strong>of</strong> parotid<br />

gland. Depending on total absorbed dose <strong>of</strong> radiation (over<br />

60 Gy), this may lead to total (irreversible) or partial (reversible)<br />

xerostomia and to qualitative change <strong>of</strong> saliva, such as<br />

increased viscosity, decreased buffering properties and decreased<br />

IgA level. A decreased volume <strong>of</strong> secreted saliva may<br />

accompany administration <strong>of</strong> many cytostatic drugs [2, 4, 9].<br />

Increased acidity <strong>of</strong> oral cavity may be further enhanced by<br />

vomiting, which <strong>of</strong>ten accompanies chemotherapy. An extremely<br />

unfavorable aspect <strong>of</strong> chemotherapy is its prolonged<br />

administration, ranging from 6 months to 3 years, depending<br />

on the kind <strong>of</strong> neoplasm.<br />

Unfavorable alteration <strong>of</strong> oral eco-system in patients undergoing<br />

oncologic treatment may enhance demineralization<br />

<strong>of</strong> teeth, thus increasing the susceptibility <strong>of</strong> teeth to cariespromoting<br />

bacteria, leading to the development <strong>of</strong> caries and<br />

erosion <strong>of</strong> enamel. Particular form <strong>of</strong> pathologic process (caries<br />

and/or erosion) depends mainly on acid pH and duration <strong>of</strong><br />

its contact with mineralized tissues <strong>of</strong> teeth [2]. Increased susceptibility<br />

<strong>of</strong> teeth to the influence <strong>of</strong> deleterious factors (chemical,<br />

bacterial and mechanical) may be an important factor particularly<br />

in children and adolescents, where it is caused not only<br />

by the neoplasm itself and anti-cancer therapy, but also by morphologic<br />

and functional immaturity <strong>of</strong> teeth [15, 16].<br />

When discussing the etiology <strong>of</strong> damage to mineralized<br />

tissues <strong>of</strong> teeth observed in patients undergoing anti-cancer<br />

treatment, we must also take into account the effect <strong>of</strong><br />

neurologic deficits, e.g. weakness <strong>of</strong> chewing muscles leading<br />

to “lazy” chewing and compromising self-purification <strong>of</strong><br />

oral cavity. Other important factors are: co-existing stomatitis,<br />

disturbed taste, lack <strong>of</strong> appetite and general malaise, leading<br />

to hygienic neglect and dietary mistakes.<br />

There is no published data concerning the pr<strong>of</strong>ile <strong>of</strong> structural<br />

changes in mineralized tissues <strong>of</strong> teeth, developing in children<br />

undergoing chemotherapy combined with facial irradiation.<br />

Aim <strong>of</strong> the study<br />

The aim <strong>of</strong> this study was clinical assessment <strong>of</strong> lesions in<br />

mineralized tissues <strong>of</strong> teeth in children subjected to chemotherapy<br />

combined with radiotherapy to the facial region and<br />

an electron-microscopic analysis <strong>of</strong> structure <strong>of</strong> atypical defects<br />

present on smooth surface <strong>of</strong> primary teeth.<br />

Material and method<br />

Patients. The study included 88 children (57 boys and 31 girls)<br />

aged from 3 to 18 years, who underwent combined anti-cancer<br />

treatment. Time since completion <strong>of</strong> oncologic treatment<br />

ranged from 6 months to 5 years. Patients’ characteristics concerning<br />

basic diagnosis, total dose <strong>of</strong> radiation and location<br />

<strong>of</strong> irradiated area (face or neurocranium), are presented in Table<br />

1. Children included in the study have been subdivided into 3<br />

subgroups: those with primary dentition, those with mixed<br />

dentition and those with permanent dentition (Table 2).<br />

Stomatologic assessment <strong>of</strong> dentition. Dental examinations<br />

were performed in the setting <strong>of</strong> a dental <strong>of</strong>fice, using<br />

a shadowless lamp, dental probe and dental mirror. Primary<br />

assessed parameters included: oral hygiene and dental status,<br />

presence <strong>of</strong> caries, fillings, non-caries-mediated enamel defects<br />

and missing teeth. At the same time, prophylactic and<br />

therapeutic requirements were defined and secondary endpoints<br />

were assessed, such as frequency and severity <strong>of</strong> caries,<br />

treatment index and incidence <strong>of</strong> atypical non-caries-mediated<br />

defects.<br />

Oral hygiene. Oral hygiene was assessed using the<br />

“Oral Hygiene Index” according to Green and Vermillion<br />

(OHI-S). Dental plaque stained with eosin was evaluated on<br />

buccal/labial and palatal/lingual surfaces <strong>of</strong> 6 representative<br />

teeth: 55 (15), 53 (13), 51 (11), 75 (36), 73 (13), 71 (31). If<br />

the desired tooth was missing, the adjacent tooth was examined.<br />

On the basis <strong>of</strong> mean values <strong>of</strong> the OHI-S index, oral<br />

hygiene was considered good if the score was 0-1,0; adequate<br />

if the score was >1,0-2,0 and poor if the score was >2,0-3,0.<br />

Severity <strong>of</strong> caries. Evaluation was performed according<br />

to the DMF index for permanent teeth and the dmf index for<br />

primary teeth. The following lesions <strong>of</strong> mineralized tooth tissues<br />

were considered as atypical: white smooth stains, white or<br />

rusty rough stains, enamel defect within a white or a rusty stain.<br />

Structure <strong>of</strong> defect surface. Teeth were cut along the longitudinal<br />

axis and transversely at the defect level. They were<br />

fixed in 4 % glutaraldehyde solution (Serva) in 0.1 M cacodyle<br />

buffer with pH value <strong>of</strong> 7.3 (BDH Chemicals Ltd, Poole, England)<br />

and next they were dehydrated using increasing concentrations<br />

<strong>of</strong> ethyl alcohol and acetone. Tooth fragments were attached<br />

to brass rods with silver paste (AGAR, England) and<br />

vacuum-coated with charcoal and gold powder and finally analyzed<br />

using a scanning electron microscope (JSM 35C, Jeol).


101<br />

Table 1<br />

Diagnosis, mean total dose and field <strong>of</strong> radiation in patients subjected to combined chemo-radiotherapy. Radiation fields<br />

included teeth and teeth buds<br />

Diagnosis<br />

Number <strong>of</strong><br />

children (N)<br />

Mean dose <strong>of</strong><br />

radiation (cGy)<br />

Irradiated field<br />

Medulloblastoma 18 3500<br />

4500<br />

Neural axis (brain and spinal cord)<br />

Posterior fossa<br />

Brainstem tumors 7 5400 Tumor site<br />

Ependymoma 5 5400 Tumor site<br />

Astrocytoma III 5 5400 Tumor site<br />

Glioblastoma multiforme 2 5400 Tumor site<br />

Carcinoma plexus chorioidei 3 5400 Tumor site<br />

Nasopharyngeal<br />

9 5400 Tumor site<br />

rhabdomyosarcoma<br />

Non-Hodgkin lymphoma 1 2400 Neural axis<br />

Neuroblastoma stage IV 1 4500 Brain metastases<br />

Retinoblastoma 1 4500 Lymph node metastases in head and<br />

Table 2<br />

Distribution <strong>of</strong> dentition types in the analyzed population <strong>of</strong><br />

children subjected to anti-cancer treatment<br />

Dentition type<br />

Primary dentition 10<br />

Mixed dentition 30<br />

Permanent dentition 25<br />

Total 65<br />

Results<br />

Number <strong>of</strong> children (N)<br />

Clinical evaluation<br />

The incidence <strong>of</strong> caries in the studied population <strong>of</strong> children<br />

after combined chemo-radiotherapy was high and approached<br />

100 %. Oral hygiene in the whole group studied was considered<br />

adequate (mean OHI-S score = 1,88), while in the subgroup<br />

<strong>of</strong> children with primary teeth oral hygiene was poor<br />

(mean OHI-S score = 2,25) (Table 3).<br />

An analysis <strong>of</strong> severity <strong>of</strong> caries is presented in Table<br />

4. The mean dmf score was 14,6; 7,06 and 6,0 in subgroups <strong>of</strong><br />

children with primary, mixed and permanent teeth respectively,<br />

indicating extremely severe caries both in primary and in permanent<br />

dentition. In all subgroups, the proportion <strong>of</strong> primary<br />

and permanent teeth with active caries were a significant component<br />

<strong>of</strong> the mean score <strong>of</strong> dmf and DMF indices.<br />

An analysis <strong>of</strong> treatment indices, both for primary and<br />

for permanent teeth, showed significant neglect concerning<br />

both treatment and prevention (Table 5). Obtained values were<br />

very small, ranging from 0,02 to 0,43, depending on sex and<br />

type <strong>of</strong> dentition.<br />

In the area <strong>of</strong> dental therapeutic requirements (Table 6), the<br />

need for conservative treatment significantly outnumbered all<br />

other parameters. On the basis <strong>of</strong> proportion <strong>of</strong> teeth with active<br />

caries, need for conservative treatment was observed in<br />

71,42 % <strong>of</strong> teeth in the subgroup <strong>of</strong> children with primary dentition,<br />

in 48,82 % <strong>of</strong> primary teeth and 89.,10 % <strong>of</strong> permanent<br />

teeth in the subgroup with mixed dentition and in 95,22 % <strong>of</strong><br />

teeth in the subgroup <strong>of</strong> children with permanent dentition.<br />

In the subgroup <strong>of</strong> children with primary dentition, the<br />

mean number <strong>of</strong> teeth with active caries was 13,3 out <strong>of</strong> 20,0<br />

teeth examined, where the mean number <strong>of</strong> teeth qualifying<br />

for extraction was 3,8 (Table 7). In children with permanent<br />

dentition, the mean number <strong>of</strong> teeth with caries was 10,88 out<br />

<strong>of</strong> 26,88 teeth examined and the mean number <strong>of</strong> teeth qualified<br />

for extraction was 0,52. In the subgroup <strong>of</strong> children with<br />

mixed dentition, mean numbers <strong>of</strong> teeth with active caries and<br />

teeth qualified for extraction were elevated too. Interpretation<br />

<strong>of</strong> results obtained in this subgroup is difficult due to on-going<br />

process <strong>of</strong> exchange <strong>of</strong> dentition.<br />

Non-caries-mediated enamel defects were noted in 49<br />

children (75,38 % <strong>of</strong> the whole group) (Table 8). The proportion<br />

<strong>of</strong> children with non-caries-mediated enamel defects were<br />

70,0 %, 76,6 % and 76,0 % in the subgroups <strong>of</strong> children with<br />

primary, mixed and permanent dentition, respectively. Noncaries-mediated<br />

enamel defects were located on buccal or labial<br />

surfaces, in pericervical area or encompassed more than<br />

one tooth surface. However, most non-caries-dependant defects<br />

were localized on labial or buccal surfaces, taking the<br />

form <strong>of</strong> white-smooth (173 out <strong>of</strong> 229 lesions seen in 39 subjects)<br />

or rusty-rough patches (156 out <strong>of</strong> 254 lesions seen in<br />

33 subjects). Enamel defects were noticed in 22 patients. Labial<br />

or buccal location <strong>of</strong> lesions was seen in 104 out <strong>of</strong> 182<br />

lesions (Table 8).


102<br />

Table 3<br />

Correlation <strong>of</strong> oral hygiene with age and sex in children subjected to chemio- and radiotherapy (OHI-S mean value)<br />

Dentition type Sex N (children) OHI-S (mean value)<br />

Primary dentition Boys 7 2,51<br />

Girls 3 1,63<br />

Total 10 2,25<br />

Mixed dentition Boys 19 1,90<br />

Girls 11 2,01<br />

Total 30 1,94<br />

Permanent dentition Boys 17 1,70<br />

Girls 8 1,55<br />

Total 25 1,65<br />

Total Boys 43 1,92<br />

Table 4<br />

Girls 22 1,79<br />

Total 65 1,88<br />

Correlation <strong>of</strong> Severity <strong>of</strong> caries, sex and type <strong>of</strong> dentition in children subjected to chemotherapy combined with radiotherapy<br />

(mean scores <strong>of</strong> dmf and DMF indices)<br />

N (teeth)<br />

Indem sc<br />

Dentition<br />

type<br />

Sex<br />

with caries removed filled dmf D<br />

N<br />

(children) d D m M f F<br />

primary boys 7 105 - 0 - 2 - 15,20 -<br />

girls 3 28 - 0 - 11 - 13.0 -<br />

total 10 133 - 0 - 13 - 14.6 -<br />

mixed boys 19 104 102 13 0 20 15 7,21 6.<br />

girls 11 66 54 2 0 7 9 6,81 5,<br />

total 30 170 156 15 0 27 24 7,06 6,<br />

permanent boys 17 - 191 - 1 - 106 - 17<br />

girls 8 - 81 - 3 - 62 - 20<br />

total 25 - 272 - 4 - 168 - 17<br />

d/D - caries in primary /permanent dentition<br />

m/M - removed primary teeth/permanent<br />

f/F - filled primary teeth/permanent<br />

duf/DUF - dental caries severity index in primary/permanent dentition


103<br />

Table 5<br />

Correlation <strong>of</strong> teeth-treatment index, dentition type and sex in children subjected to combined chemo-radiotherapy<br />

Teeth-treatment index<br />

Dentition type Sex N (children) Primary Permanent<br />

Primary boys 7 0,02 -<br />

girls 3 0,28 -<br />

total 10 0,09 -<br />

Mixed boys 19 0,16 0,12<br />

girls 11 0,09 0,14<br />

total 30 0,14 0,13<br />

permanent boys 17 - 0,36<br />

girls 8 - 0,43<br />

total 25 - 0,38<br />

Table 6<br />

Correlation <strong>of</strong> requirement for conservative and surgical treatment with sex and dentition type in children subjected to chemotherapy<br />

combined with radiotherapy<br />

Dentition<br />

type<br />

Sex<br />

N<br />

(children)<br />

examined<br />

N (teeth)<br />

with caries<br />

n=100% For conservative treatment For extraction<br />

M S M S M S M S<br />

n % n % n % n %<br />

Primary boys 7 140 - 105 - 71 67,6 - - 34 32,4 - -<br />

girls 3 60 - 28 - 24 85,7 - - 4 14,28 - -<br />

total 10 200 - 133 - 95 71,42 - - 38 28,57 - -<br />

Mixed boys 19 163 271 104 102 53 50,96 94 92,15 51 49,03 8 7,84<br />

girls 11 102 155 66 54 30 45,45 45 83,33 36 54,54 9 16,66<br />

total 30 265 426 170 156 83 48,82 139 89,10 87 51,17 17 10,89<br />

Perma-nent boys 17 - 451 - 191 - - 180 94,24 - - 11 5,75<br />

girls 8 - 221 - 81 - - 79 97,53 - - 2 2,46<br />

total 25 - 672 - 272 - - 259 95,22 - - 13 4,77<br />

M- primary teeth<br />

S- permanent teeth


104<br />

Table 7<br />

Total mean number <strong>of</strong> teeth with caries and total mean number <strong>of</strong> teeth requiring conservative and surgical treatment depending<br />

on dentition type<br />

Dentition<br />

type N Examined with caries<br />

N (teeth) (mean value)<br />

requiring<br />

conservative<br />

treatment<br />

requiring<br />

extraction<br />

M S M S M S M S<br />

Primary 10 20 - 13,30 - 9,50 - 3,80 -<br />

Mixed 30 8,83 14,20 5,66 5,20 2,76 4,63 2,90 0,56<br />

Permanent 25 - 26,88 - 10,88 - 10,36 - 0,52<br />

Table 8<br />

Non-caries-dependent enamel defects in children subjected to chemo- and radiotherapy dependent on dentition type (w-labial/<br />

buccal surface, sz-cervical area)<br />

White smooth stain Rough stain Enamel defects within s<br />

Dentition<br />

Nt Nt Nt<br />

type Nc Nc w sz >1 total Nc w sz >1 total Nc w sz >1<br />

Primary 7 2 2 4 - 6 6 35 8 - 43 5 49 - 15<br />

Mixed 23 21 106 1 27 134 16 77 3 35 115 6 19 - 6<br />

Perma-nent 19 16 65 1 24 89 13 40 4 58 96 11 36 36 21<br />

Total 49 39 173 6 51 229 33 156 15 93 254 22 104 36 42<br />

Nc = number <strong>of</strong> children<br />

Nt = number <strong>of</strong> teeth<br />

Fig. 1 Scanning electron microscopic view <strong>of</strong> tooth surface with cracked<br />

enamel and altered enamel prisms within an area <strong>of</strong> enamel defect. Microscopical<br />

magnification x 1100<br />

Fig. 2 Scanning electron microscopic view <strong>of</strong> the tooth dentin canals. Microscopical<br />

magnification x 1500


105<br />

Electron-microscopic examination<br />

Scanning electron microscopical analysis <strong>of</strong> the enamel <strong>of</strong> teeth<br />

showed its lack or crack in number <strong>of</strong> areas. Derangement <strong>of</strong><br />

enamel prisms and their clump was observed (Fig.1). No alterations<br />

<strong>of</strong> the pattern <strong>of</strong> dentin canals were noticed (Fig.2).<br />

Discussion<br />

Intensification <strong>of</strong> chemotherapy and it’s combination with<br />

surgery and/or radiotherapy have made possible an increasingly<br />

effective treatment <strong>of</strong> head and neck malignancies [1,<br />

2]. However, such an aggressive treatment is associated with<br />

an increased risk <strong>of</strong> many adverse side-effects, concerning<br />

many different components <strong>of</strong> the chewing apparatus. Particularly<br />

severe complications may accompany anticancer<br />

treatment <strong>of</strong> children and adolescents. Anticancer treatment<br />

proves effective in a growing number <strong>of</strong> patients, resulting in<br />

longer survival time. This in turn contributes to an increased<br />

incidence <strong>of</strong> late adverse effects <strong>of</strong> therapy, e.g. destruction <strong>of</strong><br />

mineralized tissues <strong>of</strong> teeth and various developmental abnormalities<br />

<strong>of</strong> chewing apparatus [3, 4, 5]. Therefore, oral care<br />

pr<strong>of</strong>essionals will be increasingly <strong>of</strong>ten confronted by such<br />

problems. Unfortunately, available literature provides little data<br />

concerning dental problems in patients undergoing anticancer<br />

treatment in childhood or guidelines for their effective<br />

prophylactic, therapeutic and rehabilitative management.<br />

Results <strong>of</strong> clinical trials indicate significant hygienic<br />

neglect and poor dentition status <strong>of</strong> children subjected to chemotherapy<br />

combined with radiotherapy. We noticed alarmingly<br />

high incidence <strong>of</strong> caries and it’s extreme severity. In our<br />

study, the mean dmf score was 14,6, largely exceeding values<br />

observed in children in the risk group for infectious endocarditis,<br />

where dmf score was 9,25 and DMF score was 10,42, as<br />

well as in children undergoing chemotherapy alone, where it<br />

equaled to 2,5 and 14,1 respectively [13]. We have noticed a<br />

large disproportion between requirements for dental treatment<br />

and dental treatment actually carried out. Values <strong>of</strong> treatment<br />

indices in children after anticancer treatment and those in the<br />

risk group for infectious endocarditis were similar, in the range<br />

<strong>of</strong> 0-0,34 and 0,03-0,34 respectively.<br />

An analysis <strong>of</strong> the scope <strong>of</strong> required dental treatment<br />

in children after chemotherapy combined with radiotherapy<br />

revealed significant needs in the area <strong>of</strong> conservative treatment<br />

and relatively smaller needs in the field <strong>of</strong> surgical treatment,<br />

possibly indicating high speed <strong>of</strong> development <strong>of</strong> new<br />

caries-dependent defects.<br />

In 69 % <strong>of</strong> children analyzed we observed atypical noncaries<br />

dependent enamel lesions in the form <strong>of</strong> patches and<br />

enamel defects. They occurred twice as <strong>of</strong>ten than in children<br />

undergoing chemotherapy alone. Children after chemotherapy<br />

combined with radiotherapy presented mostly white, rusty or<br />

rough patches, while children after chemotherapy alone had<br />

usually white lesions.<br />

Deleterious effect <strong>of</strong> radiotherapy on dentition is well known<br />

since a long time. The so-called “post-radiation caries” in patients<br />

undergoing radiotherapy in the area <strong>of</strong> head and neck,<br />

is a phenomenon well established in the literature [2, 3, 5, 8,<br />

11, 12, 14]. Observed thereby enamel defects are at the beginning<br />

probably non-bacterial in nature and are caused by chemical<br />

and mechanical factors acting upon a primarily more susceptible<br />

enamel. It would seem, that subsequent development<br />

<strong>of</strong> caries in these patients is secondary to the above described<br />

phenomena.<br />

Conclusions<br />

1. High incidence and severity <strong>of</strong> caries and atypical non-caries<br />

mediated enamel defects observed in patients subjected to<br />

chemotherapy combined with radiotherapy in the facial region,<br />

support the thesis about deleterious effect <strong>of</strong> anti-cancer<br />

on dentition.<br />

2. Significant disproportion between requirements for dental<br />

treatment and actually performed conservative and surgical<br />

treatment indicate, that dental prophylactic and therapeutic<br />

management should be instituted as early as possible in these<br />

patients.<br />

References<br />

1. Cioch M (2001) Uszkodzenie bariery<br />

sluzówkowej (Mucosal barier injury-<br />

MBI) w nastêpstwie intensywnego<br />

leczenia cytostatycznego. Onkol Pol 4,2:<br />

85-89 (in Polish)<br />

2. Darczuk D (1999) Zmiany w jamie<br />

ustnej wywo³ane napromienianiem<br />

nowotworów g³owy i szyi. Stomat<br />

Wspó³czesna 6,1:23-25 (in Polish)<br />

3. Groetz KA, Wagner W, Duschner H<br />

(2001) Chronische Strahlenfolgen an<br />

den<br />

Zahnhartgeweben<br />

(“Strahlenkaries”). Klassifikation und<br />

Behandlungsansaetze. Strahlentherapie<br />

und Onkologie-Abstract 177,2 :96-10<br />

4. Jaffe N, Toth BB, Hoar RE, Ried HL,<br />

Sullivan MP, McNeese M (1984) Dental<br />

and maxill<strong>of</strong>acial abnormalites in<br />

long-term survivors <strong>of</strong> childhood cancer:<br />

effects <strong>of</strong> treatment with chemotherapy<br />

and radiation to the head and<br />

neck. Pediatrics 73,6:816-823<br />

5. Jankowska K (1992) Próchnica<br />

popromienna (opis przypadku).<br />

Wroc³awska Stomatologia 235-238 (in<br />

Polish)<br />

6. Macleod RI, Welbury RR, Soames J<br />

(1987) Effects <strong>of</strong> cytotoxic chemotherapy<br />

on dental development. J Royal<br />

Soc Med 80:207-209<br />

7. Maguire A, Welbury R (1996) Long-term<br />

Effects <strong>of</strong> antineoplastic chemotherapy<br />

and radiotherapy on dental development.<br />

Dental Update 23 (5):188-194<br />

8. Manguire A, Craft AW, Evans RGB,<br />

Amineddine H, Kehrnahan J, Macleod<br />

RI (1987) Long-term effects <strong>of</strong> treatment<br />

on the dental condition <strong>of</strong> children<br />

surviving malignant disease. Cancer<br />

60:2570-2575<br />

9. Markitziu A, Zafiropolos G, Tsalikis L,<br />

Cohen L (1992) Gingival health and salivary<br />

function in head- and neck–<br />

irradited patients. Oral Surg Oral Med<br />

Oral Pathol 73:427-433


10. Nunn JH, Welbury RR, Gordon PH,<br />

Kernahan J, Craft AW (1991) Dental<br />

caries and dental anomalies in children<br />

treated by chemotherapy for malignant<br />

disease: a study in the north <strong>of</strong> England.<br />

Int J Paediatr Dent 1:131-135<br />

11. Olczak-Kowalczyk D, Dobies K,<br />

Daszkiewicz M (2002) Zastosowanie<br />

lakieru Seal & Protect w pr<strong>of</strong>ilaktyce<br />

próchnicy popromiennej u dzieci po<br />

przebytej terapii przeciwnowotworowej.<br />

Magazyn Stomatologiczny<br />

11,133,12:10-13 (in Polish)<br />

12. Olczak-Kowalczyk D, Daszkiewicz M<br />

(2002) Wspó³czesne metody<br />

pr<strong>of</strong>ilaktyki próchnicy w grupie<br />

wysokiego ryzyka jej wystêpowania.<br />

Standardy Medyczne 4,6 (32):402-411<br />

(in Polish)<br />

13. Olczak-Kowalczyk D, Bedra B (2003)<br />

Stan zdrowia jamy ustnej i<br />

stomatologiczne potrzeby lecznicze w<br />

badanej populacji dzieci z ryzykiem<br />

infekcyjnego zapalenia wsierdzia. Nowa<br />

Stomat 3,25:111-119 (in Polish)<br />

14. Olczak-Kowalczyk D, Perek D,<br />

Daszkiewicz M, Adamowicz-Klepalska<br />

B, Dembowska-Baginska B,<br />

Daszkiewicz P (2003) Problemy<br />

stomatologiczne u dzieci z chorobami<br />

nowotworowymi. Doœwiadczenia<br />

wlasne. Nowa Stomat 4;26:175-179 (in<br />

Polish)<br />

15. Rosenberg SW, Kolodney H, Wong<br />

GY, Murphy ML (1987) Altered dental<br />

root development in long-term survivors<br />

<strong>of</strong> pediatric acute lymphoblastic<br />

leukemia. A review <strong>of</strong> 17 cases.<br />

Cancer 59:1640-1648<br />

16. Welbury RR, Craft AW, Murray JJ,<br />

Kehrnahan J (1984) Dental health <strong>of</strong> survivors<br />

<strong>of</strong> malignant disease. Archiv Dis<br />

Child 59:1186-1188


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4): 106-110<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Preliminary review <strong>of</strong> the treatment results <strong>of</strong> hepatoblastoma and<br />

hepatocarcinoma in children in Poland in the period 1998-2002<br />

Czes³aw Stoba 1 , Katarzyna Nierzwicka 1 , Piotr Czauderna 1 , Barbara Skoczylas-Stoba 2 ,<br />

Stefan Popadiuk 3 , Jolanta Bonar 4 , Krystyna Sawicz-Birkowska 5 , Hanna Kaczmarek-Kanold 6 ,<br />

Przemys³aw Mañkowski 7 , Krzyszt<strong>of</strong> K¹tski 8 , Wojciech Madziara 9 , Magdalena Rych³owska 10 ,<br />

Anna Sitkiewicz 11 , Anna Szo³kiewicz 12 , Sabina Szymik-Kantorowicz 13 , Violetta Œwi¹tkiewicz 14<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

1<br />

Department <strong>of</strong> Pediatric Surgery,<br />

3<br />

Department <strong>of</strong> Pediatrics, Gastroenterology and Oncology<br />

12<br />

Department <strong>of</strong> Pediatric Oncology<br />

Medical University <strong>of</strong> Gdansk, Gdansk,Poland<br />

2<br />

Department <strong>of</strong> Anesthesiology and Intensive Therapy,<br />

Regional Copernicus Hospital, Gdansk, Poland<br />

4<br />

Department <strong>of</strong> Pediatrics, Hematology, Oncology<br />

and Endocrinology,<br />

5<br />

Department <strong>of</strong> Pediatric Surgery,<br />

Medical University <strong>of</strong> Wroclaw, Wroclaw, Poland<br />

6<br />

Department <strong>of</strong> Pediatric Hematology and Oncology,<br />

7<br />

Department <strong>of</strong> Pediatric Surgery,<br />

Medical University <strong>of</strong> Poznan, Poznan, Poland<br />

8<br />

Department <strong>of</strong> Pediatric Hematology and Oncology,<br />

Medical University <strong>of</strong> Lublin, Lublin, Poland<br />

9<br />

Department <strong>of</strong> Pediatric Surgery,<br />

Medical University <strong>of</strong> Katowice, Katowice, Poland<br />

10<br />

Department <strong>of</strong> Pediatric Surgery and Oncology,<br />

Institute <strong>of</strong> Mother and Child, Warszawa, Poland<br />

11<br />

Department <strong>of</strong> Pediatric Surgery and Oncology,<br />

Medical University <strong>of</strong> Lodz, Lodz, Poland<br />

13<br />

Department <strong>of</strong> Pediatric Surgery,<br />

Medical University <strong>of</strong> Krakow, Krakow, Poland<br />

14<br />

Department <strong>of</strong> Hematology and Oncology,<br />

Medical University <strong>of</strong> Bydgoszcz, Bydgoszcz, Poland<br />

Abstract<br />

Address for correspondence<br />

Objectives: Hepatoblastoma (HB) and hepatocellular carcinomas (HCC) are most common <strong>of</strong> all primary<br />

malignant liver tumors in children. This series <strong>of</strong> HB and HCC represents a preliminary report <strong>of</strong> treatment in<br />

11 Polish pediatric oncology centers. Treatment protocols were based on two consecutive SIOPEL group<br />

studies: 2 and 3. Material and methods: From 1998 to 2002 there were 34 children with HB and HCC<br />

diagnosed (28 were HB and 6 were HCC). All HB and HCC patients received preoperative chemotherapy<br />

based on SIOPEL 3 protocols (CDDP alone, PLADO or SuperPLADO). Among HB 23 tumors were unifocal<br />

and 5 multifocal. Eight HB cases were qualified to the high risk group according to the SIOPEL protocol<br />

definition. Alphafetoprotein (AFP) was elevated in all cases. Lung metastases were present in one HB and in<br />

three HCC cases. In 5 cases diagnosis was made on the clinical ground, in most <strong>of</strong> the patients initial biopsy<br />

was performed. Standard risk tumor were treated either Cisplatin monotherapy or PLADO regimen. High<br />

risk group was treated with SuperPLADO regimen. Twenty-one HB cases were operated in a delayed setting.<br />

Five tumor resections were incomplete: 3 macroscopically and 2 microscopically. In 4 cases information on<br />

surgery is missing. Results: Among 28 HB patient 19 are alive with no evidence <strong>of</strong> disease, 2 are alive with<br />

disease, 6 children died and one is lost to follow-up. Thus 3-year overall survival <strong>of</strong> HB patient was 75%,<br />

while 3-year event-free-survival was 64%. Median follow-up time is 40 mouths. Overall survival among<br />

standard risk group was 90% in comparison with 66% in the high risk group. All children with macroscopic<br />

residuum post resection died. Overall survival in HCC cases at 3 years is 33%, but <strong>of</strong> the two alive patients<br />

one currently experienced recurrence. Conclusions: Reported treatment for hepatoblastoma are similar to<br />

those achieved by other study groups and constitute an improvement in comparison with our retrospective<br />

series (OS=75% vs. 44%). High risk HB patients require modification <strong>of</strong> the current treatment strategy due to<br />

inferior survival than in standard risk HB. Treatment results <strong>of</strong> HCC (OS=33%) were clearly inferior to HB<br />

being in line with experience <strong>of</strong> other international study groups. Importance <strong>of</strong> the complete tumor resection,<br />

both in HB and HCC was confirmed.<br />

Keywords: children, hepatoblastoma, hepatocellular carcinoma, surgery, treatment<br />

Piotr Czauderna, PhD<br />

Dept. <strong>of</strong> Pediatric Surgery,<br />

Medical University <strong>of</strong> Gdansk,<br />

Ul. Nowe Ogrody 1-6,<br />

80-803 Gdansk, Poland.<br />

Phone/fax: +48 58 302 64 27<br />

E-mail: pedsurg@amg.gda.pl


108<br />

Introduction<br />

In the last 15 years a steady increase in survival in pediatric<br />

liver tumors has been noted. First observations on improvement<br />

in survival with the use <strong>of</strong> adjuvant chemotherapy came<br />

from Children’s Cancer Study Group (CCSG) and Pediatric<br />

Oncology Group (POG) in the late 70’s. Later in unresectable<br />

cases neoadjuvant chemotherapy was applied [3, 9]. In 1986<br />

Belgian and Canadian centers introduced in hepatoblastoma<br />

standard pre- and postoperative chemotherapy consisting <strong>of</strong><br />

cisplatin and adriamycin. This chemotherapy protocol, called<br />

later PLADO, has become a golden standard <strong>of</strong><br />

hepatoblastoma (HB) treatment in many centers, including<br />

Mother and Child Memorial Hospital in Warsaw. Efficacy <strong>of</strong><br />

further modified protocols was tested by the multinational<br />

SIOPEL group in three subsequent trials: SIOPEL 1-3 [9-11].<br />

It was shown that cisplatin alone is as effective as PLADO in<br />

standard risk HB [9, 10].<br />

Despite chemotherapy progress complete tumor resection<br />

remains a goal <strong>of</strong> treatment and prerequisite for cure. If it<br />

cannot be achieved it is better to abandon any surgical attempt<br />

and resort to another treatment modality and/or consult the<br />

case with referral center. Our remarks on principles <strong>of</strong> the<br />

surgical treatment <strong>of</strong> liver tumors were presented during Surgical<br />

Workshop in Szklarska Porêba, Poland in 2002 [13]. It<br />

was reported that progress in surgical management <strong>of</strong> liver<br />

tumors had been associated with better knowledge <strong>of</strong> its<br />

anatomy, improved surgical technique and equipment (ultrasonic<br />

and water-jet dissectors, argon beam coagulation,<br />

thrombostatic materials), as well as new imaging techniques,<br />

new method <strong>of</strong> anesthesia and improved perioperative care.<br />

Presented results emerged from multicenter cooperation<br />

<strong>of</strong> 13 Polish institutions within the study devoted to surgical<br />

treatment <strong>of</strong> malignant epithelial tumors <strong>of</strong> childhood<br />

including patients’ stratification according to risk groups in<br />

cooperation with the International Childhood Liver Tumors<br />

Strategy Group (SIOPEL). This study is coordinated in Poland<br />

by the Department <strong>of</strong> Pediatric Surgery <strong>of</strong> the Medical<br />

University <strong>of</strong> Gdansk.<br />

Material and methods<br />

In the period 1998-2002 all together 34 cases <strong>of</strong> primary epithelial<br />

liver tumors were treated in 11 Polish paediatric oncology<br />

centers. Twenty-eight were HB and 6 were HCC including<br />

one transitional (HB/HCC) liver tumor. Patients’ characteristics<br />

and stage are shown in Tab.1 and 2. Maximal tumor<br />

diameter ranged from 5 to 18 cm. Among HB 23 tumors (82%)<br />

were unifocal and 6 – multifocal. In HCC multifocal tumors<br />

prevailed: 4 out <strong>of</strong> 5. Eight HB cases (29%) were qualified to<br />

the high risk group according to the SIOPEL protocol definition.<br />

Alphafetoprotein (AFP) was elevated in all but one HB<br />

case (from 150 ng/ml to 2.480.000 ng/ml), in which AFP level<br />

was < 100 ng/ml. In all HCC cases AFP was elevated (from<br />

150 ng/ml to 2.500.000 ng/ml). Lung metastases were present<br />

in one HB case (3,6%) and in 3 HCC cases (50%). In one<br />

HCC case metastases involved also mediastinal and retroperitoneal<br />

lymph nodes.<br />

Treatment protocols were based on consecutive SIOPEL studies:<br />

SIOPEL 2 and 3. Standard imaging methods included:<br />

abdominal ultrasonography (US), chest X-ray (AP and lateral),<br />

computed tomography (CT) <strong>of</strong> the abdomen (with and<br />

without i.v. contrast) and chest (to detect eventual pulmonary<br />

metastases) and/or magnetic resonance imaging (MRI). Also<br />

complete blood count (including platelet level) and serum AFP<br />

were obtained at diagnosis. AFP, if elevated, was used to monitor<br />

response to treatment. The protocol required preoperative<br />

assessment <strong>of</strong> tumor extent according to PRETEXT classification,<br />

which is described in details elsewhere [12]. Closed<br />

needle biopsy was performed in 2 cases, open biopsies were<br />

done in 21 cases (18 – wedge and 3 – open needle biopsies).<br />

In one case liver biopsy was done laparoscopically. In 5 cases<br />

diagnosis was made on the clinical ground. In 4 patients data<br />

on biopsy are missing. Patients with biopsy proven HB and<br />

HCC were qualified to two risk groups on the basis <strong>of</strong> PRE-<br />

TEXT grouping. High risk tumors were those PRETEXT 4<br />

(involving the whole liver) or belonging to any PRETEXT<br />

category with the following features: significant intravascular<br />

involvement (V or P), extrahepatic extension and/or distant<br />

metastases (M). Hence high risk tumors were primarily<br />

unresectable. All others formed standard risk group. In tumors<br />

occurring between 6 months and 3 years <strong>of</strong> age with<br />

unequivocal imaging and increased AFP level diagnosis <strong>of</strong><br />

hepatoblastoma on clinical ground was allowed. Before definite<br />

surgery Doppler US and helical CT were required. All<br />

patients with hepatoblastoma received preoperative chemotherapy<br />

which was dependent on the risk group assignment.<br />

Standard risk tumors were treated either cisplatin monotherapy<br />

or PLADO regimen (those registered in the SIOPEL 3 trial<br />

were randomized), however few patients were not randomized.<br />

High risk patients were treated with SuperPLADO regimen,<br />

which included 2-weekly cisplatin alternating with<br />

doxorubicin administered together with carboplatin. Four triweekly<br />

chemotherapy courses were given. Postoperatively two<br />

more courses <strong>of</strong> the same chemotherapy were administered.<br />

For details <strong>of</strong> the treatment protocol check elsewhere [9, 10,<br />

11]. Cisplatin alone was used in 10 children, PLADO in 7<br />

cases and SuperPLADO in 9 cases. In 2 cases details on preoperative<br />

chemotherapy are missing. Twenty-one HB cases<br />

were operated in a delayed setting. Type and completeness <strong>of</strong><br />

performed surgery is shown in Table 3. Treatment <strong>of</strong> operable<br />

hepatocellular carcinoma (HCC) patients was started with tumor<br />

resection followed by 6 postoperative courses <strong>of</strong><br />

SuperPLADO regimen. Two HCC cases were primarily operated<br />

including one orthotopic liver transplantation Treatment<br />

<strong>of</strong> unresectable and/or metastatic HCC was identical with high<br />

risk HB. One child with HCC was operated in a delayed setting<br />

after partial response to preoperative chemotherapy. In<br />

non-responders (2 cases) chemoembolization was used.


109<br />

Results<br />

Among 28 HB patients 19 are alive with no evidence <strong>of</strong> disease,<br />

2 are alive with disease, 6 children died and one is lost to<br />

follow-up. Thus 3-year overall survival <strong>of</strong> hepatoblastoma<br />

patients was 75%, while 3-year event-free-survival was 64%.<br />

Three deaths resulted from disease progression and 3 from<br />

treatment complications: one was surgery-related (intestinal<br />

necrosis), one resulted from chemotherapy complications (severe<br />

neutropenia and typhlitis) and one was unexplained (possible<br />

anthracycline-related cardiomyopathy). Median followup<br />

time is 40 months (range from 24 to 72 months). Overall<br />

survival among standard risk patients was 90% (17 out <strong>of</strong> 19)<br />

in comparison with 66% (6 out <strong>of</strong> 9) in the high risk group.<br />

Preliminary analysis showed that 75% <strong>of</strong> standard risk<br />

patients (12 out <strong>of</strong> 16; in 3 cases data are not known) responded<br />

to preoperative chemotherapy (CDDP or PLADO), while from<br />

the high risk group 55% (5 out <strong>of</strong> 9) responded to<br />

SuperPLADO protocol.<br />

All together in HB cases 5 incomplete tumor resections<br />

were done: in 3 macroscopic residuum resulted and in<br />

two microscopic residuum was left. All patients with macroscopic<br />

residuum died, while one patients after microscopically<br />

incomplete resection is alive with no evidence <strong>of</strong> disease<br />

while another died due to surgical complications. In 4<br />

cases information on surgery is missing.<br />

Overall survival in HCC cases at 3 years is 33% but<br />

one <strong>of</strong> the two alive patients currently experienced recurrence.<br />

Among 6 patients with HCC in 2 cases primary tumor resection<br />

was done (including one orthotopic liver transplantation)<br />

was performed and 3 patients were treated with induction chemotherapy.<br />

In 2 non-responders chemombolization was used.<br />

Two patients were never made operable and in one data on<br />

surgery are missing.<br />

Table 1<br />

Localisation <strong>of</strong> liver tumors<br />

Localisation<br />

Hepatoblastoma<br />

Both lobem 12 4<br />

Right lobe 11 2<br />

Left lobe 5 -<br />

Table 2<br />

Liver tumors' stages<br />

PRETEXT<br />

Hepatoblastoma<br />

I 1 -<br />

II 14 1<br />

III 7 2<br />

IV 6 3<br />

Hepatocellular<br />

carcinoma<br />

Hepatocellular<br />

carcinoma<br />

Discussion<br />

Reported treatment results for HB are similar to those achieved<br />

by other study groups (OS=75%). Introduction <strong>of</strong> efficient<br />

chemotherapy has contributed to a significant progress in the<br />

treatment <strong>of</strong> hepatoblastoma as nowadays overall survival<br />

reaches 70%. Before this era only 25% <strong>of</strong> HB patients survived<br />

and tumor resection was possible in not more than 50%<br />

<strong>of</strong> patients, while half <strong>of</strong> them died later due to metastases<br />

[3]. Similarly retrospective analysis <strong>of</strong> the Polish treatment<br />

results for the period 1985-1995 showed inferior outcome with<br />

only 44% long term survival in hepatoblastoma due to the<br />

scarcely used preoperative chemotherapy and few extended<br />

liver resections done [1, 6]. In that period as many as 30% <strong>of</strong><br />

analyzed tumors were never made operable. Thus, currently<br />

achieved treatment results <strong>of</strong> hepatoblastoma in Poland constitute<br />

a significant improvement in comparison with our past<br />

series [1]. However despite overall improvement prognosis <strong>of</strong><br />

the high risk patients remains uncertain – with 66% overall<br />

survival vs. 90% for the standard risk group. Similar results<br />

were achieved in the SIOPEL 2 study, in which 3-year eventfree-survival<br />

for high risk patients was 47% vs. 89% for standard<br />

risk [9, 10]. Most failures resulted from tumor’s<br />

unresectability and insufficient or complete lack <strong>of</strong> response<br />

to preoperative chemotherapy. This confirms that high risk<br />

patients require some modifications in treatment strategy,<br />

which should be aimed at increase <strong>of</strong> resection rate. This includes<br />

more efficient induction chemotherapy, as well as more<br />

frequent use <strong>of</strong> liver transplantations in hepatoblastoma cases<br />

unresectable by conventional means [7, 8].<br />

Treatment results <strong>of</strong> hepatocellular carcinoma (33%<br />

overall survival) were clearly inferior to hepatoblastoma is<br />

confirmed by most international study groups, in which survival<br />

remained in the range <strong>of</strong> 20-30% [2, 5]. Complete tumor<br />

resection should be an essential treatment step, both in<br />

HB and HCC. In the reported series all patients after macroscopically<br />

incomplete tumor resections died, while all HB<br />

cases, who underwent complete resection, had been alive and<br />

well. Due to the relatively short follow-up <strong>of</strong> some patients,<br />

incomplete data and limited number <strong>of</strong> cases presented analysis<br />

has a preliminary character only.<br />

Conclusions<br />

1. Overall survival achieved in hepatoblastoma – 75%, as well<br />

as EFS = 64% are similar to other international reports and<br />

constitute a significant improvement in comparison with<br />

the Polish past series (1985-1995): OS = 44%.<br />

2. Complete tumor resection remains an essential step to<br />

achieve cure in primary epithelial liver tumors (HB and<br />

HCC). It is however difficult in high risk HB, as well as in<br />

HCC patients.<br />

3. High risk HB patients require modification <strong>of</strong> the current<br />

treatment strategy due to inferior survival than in standard<br />

risk HB.<br />

4. HCC are associated with poor prognosis and require completely<br />

different therapeutic strategy than HB.


Table 3<br />

Characteristics <strong>of</strong> surgical treatment<br />

Type <strong>of</strong> surgery Hepatoblastoma Hepatocarcinoma Radicality<br />

RT Hemihepatectomy 9 1 7rad. / 2 non-rad.<br />

LT Hemihepatectomy 3 - 3 rad.<br />

Extended Hemihepatectomy 7 - 5 rad. / 2 non-rad.<br />

Atypical 3 - 2 rad. /1 non-rad.<br />

Central resection 1 - 1 rad.<br />

Liver transplantation - 1 1 rad.<br />

Never operable 1 3 -<br />

Missing data 4 1 -<br />

References<br />

1. Czauderna P, Popadiuk S, Korzon M,<br />

et al (2001) Multicenter retrospective<br />

analysis <strong>of</strong> various primary pediatric<br />

malignant hepatic tumours management<br />

in the series <strong>of</strong> 47 Polish patients (1985-<br />

1995). Eur J Pediatr Surg 11:82-85<br />

2. Czauderna P, Mackinlay G, Perilongo<br />

G, et al (2002) Hepatocellular carcinoma<br />

in children – Results <strong>of</strong> the first prospective<br />

study <strong>of</strong> the International Society<br />

<strong>of</strong> <strong>Paediatric</strong> Oncology. J Clin Oncol<br />

20:2798-2804<br />

3. Ehrlich PF, Grereenberg ML, Filler<br />

RM (1997) Improved long-term survival<br />

with preoperative chemotherapy<br />

for hepatoblastoma. J Pediatr Surg<br />

32:999-1002<br />

4. Katzenstein HM, London WB,<br />

Douglass EC, Reynolds M, Plaschkes<br />

J, Finegold MJ, Bowman LC (2002)<br />

Treatment <strong>of</strong> unresectable and metastatic<br />

hepatoblastoma: a pediatric oncology<br />

group phase II study J Clin Oncol<br />

20:3438-3444<br />

5. Katzenstein HM, Krailo MD,<br />

Malogolowkin MH, et al (2002) Hepatocellular<br />

carcinoma in children and<br />

adolescents: results from the Pediatric<br />

Oncology Group and the Children’s<br />

Cancer Group Intergroup Study. J Clin<br />

Oncol 20: 2789-2797<br />

6. Korzon M, Popadiuk S, Stoba C, et al<br />

(1997) Analiza retrospektywna<br />

zlosliwych guzów watroby u dzieci w<br />

Polsce w latach 1985-1995. Pediatr Pol<br />

11 (Suppl):37-42 (in Polish)<br />

7. Ortega JA, Douglass EC, Feusner JH,<br />

et al (2000) Randomized comparison <strong>of</strong><br />

Cisplatin/Vincristine/5-Fluorouracil and<br />

Cisplatin/continuous infusion Doxorubicin<br />

for the treatment <strong>of</strong> paediatric<br />

hepatoblastoma (HB): a report from the<br />

Children’s Cancer Group and the <strong>Paediatric</strong><br />

Oncology Group. J Clin Oncol<br />

18:2665-2675<br />

8. Otte JB, Aronson DC, Brown J,<br />

Czauderna P, et al (2004) Liver transplantation<br />

for hepatoblastoma. results<br />

from the International Society <strong>of</strong> Pediatric<br />

Oncology (SIOP) study siopel-1<br />

and review <strong>of</strong> the world experience.<br />

Pediatr Blood Cancer 42:74-83<br />

9. Perilongo G, Shafford E, Plasches J<br />

(2000) SIOPEL trials using preoperative<br />

chemotherapy in hepatoblastoma. The<br />

Lancet Oncology 1:94-100<br />

10. Perilongo G, Shafford E, Maibach R<br />

(2004) Risk-adapted treatment for childhood<br />

hepatoblastoma. Final report <strong>of</strong> the<br />

second study <strong>of</strong> the International Society<br />

<strong>of</strong> Pediatric Oncology - SIOPEL 2.<br />

Eur J Cancer 40:411-421<br />

11. Pritchard J, Brown J, Shafford E (2000)<br />

Cisplatin, doxorubicin and delayed surgery<br />

for childhood hepato-blastoma: a<br />

succesful approach - Results <strong>of</strong> the first<br />

prospective study <strong>of</strong> the International<br />

Society <strong>of</strong> <strong>Paediatric</strong> Oncology J Clin<br />

Oncol 18:3819-3828<br />

12. Schnater JM, Aronson DC, Plaschkes<br />

J (2002) Surgical view <strong>of</strong> the treatment<br />

<strong>of</strong> patients with hepatoblastoma: results<br />

from the first prospective trial <strong>of</strong><br />

the International Society <strong>of</strong> Pediatric<br />

Oncology Liver Tumor Study Group<br />

(SIOPEL-1). Cancer 94:1111-1120<br />

13. Stoba C, Czauderna P, Narozanski<br />

(2001) Pierwotne nowotwory watroby<br />

u dzieci – aspekty chirurgiczne. In:<br />

Wybrane nowotwory jamy brzusznej<br />

u dzieci. A. Jankowski, P. Mankowski<br />

(Eds) Stowarzyszenie Pomocy<br />

Dzieciom Wymagajacym Leczenia<br />

Chirurgicznego, Poznan, pp. 12-30 (in<br />

Polish)


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Liver transplantation for primary malignant liver tumors<br />

in children – single center experience<br />

Piotr Kaliciñski 1 , Hor Ismail 1 , Andrzej Kamiñski 1 , Danuta Perek 2 , Joanna Paw³owska 3 ,<br />

Przemys³aw Kluge 4 , Joanna Teisserye 1 , Marek Szymczak 1 , Tomasz Drewniak 1 ,<br />

Pawe³ Nachulewicz 1 , Bo¿ena Dembowska-Bagiñska 2 , Marek Stefanowicz 1 ,<br />

Andrzej Byszewski 5 , Ma³gorzata Manowska 5<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

1<br />

Department <strong>of</strong> Pediatric Surgery and Organ Transplantation<br />

2<br />

Department <strong>of</strong> Oncology<br />

3<br />

Department <strong>of</strong> Gastrology, Hepatology and Immunology<br />

4<br />

Department <strong>of</strong> Patomorphology<br />

5<br />

Department <strong>of</strong> Anesthesiologu and Intensive Care<br />

The Children’s Memorial Health Institute,<br />

Warsaw, Poland<br />

Abstract<br />

The aim <strong>of</strong> the study was to analyse results <strong>of</strong> treatment <strong>of</strong> primary liver tumors in children in a center<br />

performing the liver transplant program. Clinical material consisted <strong>of</strong> 42 children treated for hepatoblastoma<br />

or hepatocarcinoma since 1990, since liver transplantant program was established. Retrospective comparison<br />

<strong>of</strong> results was performed according to type <strong>of</strong> the tumor and different treatments protocols. Survival <strong>of</strong><br />

patients with hepatoblastoma is similar after primary conventional treatment (chemotherapy and resection)<br />

and primary transplantation after chemotherapy, however number <strong>of</strong> transplanted patients is to low to draw<br />

any conclusions. Patients with tumor recurrence after resection probably should not be qualified for transplantation<br />

as their prognosis is very poor. In hepatocarcinoma group overall results after chemotherapy and<br />

primary transplantation seem to be much better than after conventional therapy. This observation should be<br />

confirmed however with longer follow-up <strong>of</strong> these patients. In conclusion liver transplantation should be<br />

considered as an option early in the process <strong>of</strong> treatment <strong>of</strong> primary malignant tumors in children particularly<br />

with hepatocarcinoma, and transplantation as a primary surgical treatment in all patients with unresectable or<br />

marginally resectable tumors and without extrahepatic extension.<br />

Key words: children, hepatoblastoma, hepatocellular carcinoma, liver transplantation, primary liver tumors<br />

Introduction<br />

Primary hepatic malignancies are main indication for liver<br />

transplantation in 2-4% pediatric recipients. This is well less<br />

than in adult population, where patients with hepatocellular<br />

carcinoma account for 11-12% <strong>of</strong> liver transplantations [3-5].<br />

In contrast to adult group, there are no established strict criteria<br />

for qualification for transplantation in children with primary<br />

liver tumors, particularly that various types <strong>of</strong> hepatic<br />

tumors may develop in childhood (hepatoblastoma,<br />

hepatocarcinoma, undifferentiated sarcoma, angiosarcoma<br />

etc.) [1, 7, 12]. The aim <strong>of</strong> this study is to summarize our<br />

experience with this particular group <strong>of</strong> liver transplant recipients.<br />

Material and methods<br />

Liver transplant program was initiated in our institution in<br />

march 1990. Since then 222 liver transplants were performed<br />

till July 2004. We have analysed retrospectively data <strong>of</strong> 42<br />

children treated for hepatoblastoma (HBL) or hepatocarcinoma<br />

(HCC) within the same period in our institution.<br />

Patients with HBL<br />

Between 1990 and 2004, 23 patients aged 5 months to 16<br />

years, most with advanced tumors, were treated for HBL.<br />

Twenty children with HBL were treated initially with chemotherapy<br />

and then by conventional surgery – partial liver resection.<br />

In two <strong>of</strong> them recurrence was suspected despite <strong>of</strong> mul-<br />

Address for correspondence<br />

Piotr Kalicinski MD, PhD<br />

Al. Dzieci Polskich 20<br />

04-736 Warsaw, Poland<br />

Phone: +48-22-8151360<br />

Fax: +48-22-8151450<br />

E-mail: piokal@czd.waw.pl


0000000000000000000<br />

0000000000000000000<br />

0000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

000000000000000000<br />

000000000000000000<br />

000000000000000000<br />

000000000000000000<br />

e<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000<br />

0000000000000000000<br />

0000000000000000000<br />

0000000000000000000<br />

ea<br />

00000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000<br />

0000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000<br />

0000000000000<br />

00000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000000<br />

0000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000<br />

00000000000000000<br />

00000000000000000<br />

e<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

0000000000000000000000000000000000000000000000000000<br />

00000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000<br />

00000000000000000<br />

00000000000000000<br />

ea<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

00000000000000000<br />

00000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

000000000000000000000000000000000000000000000000000000000000000000<br />

112<br />

tiple postoperative chemotherapy courses, and were qualified<br />

for total hepatectomy <strong>of</strong> the remaining liver and orthotopic<br />

liver transplantation. Another 2 children were transplanted<br />

immediately after initial chemotherapy. In 1 patient only palliative<br />

treatment (chemotherapy) was administered.<br />

Patients with HCC<br />

There were 19 patients aged 3 to 18 years treated for HCC. In<br />

9 children initial chemotherapy was administered, followed<br />

by partial liver resection, in 2 <strong>of</strong> them despite multiple resections<br />

tumor recurred and salvage transplantation was done in<br />

both <strong>of</strong> them. In 5 children liver transplantation was performed<br />

as primary surgical treatment. All <strong>of</strong> them exceeded Milano<br />

criteria for transplantation in patients with HCC. Initial chemotherapy<br />

was given in 4 <strong>of</strong> them. In 4 pts liver transplantation<br />

was performed due to suspected tumor development in<br />

cirrhotic liver (on the basis <strong>of</strong> CT or ultrasound findings or<br />

growing serum alphafetoprotein concentration). No<br />

pretransplant chemotherapy was administered to these patients.<br />

In 1 child only palliative chemotherapy was given.<br />

reccurence. One patient is alive after extensive resection, however<br />

with poor prognosis due to extrahepatic invasion <strong>of</strong> the<br />

tumor at the time <strong>of</strong> surgery.<br />

Among patients transplanted, 9/11 (81,8%) are alive,<br />

while 2 deaths were not related to tumor reccurence (early<br />

graft failure in 1 and chronic rejection in 1 (Fig. 2).<br />

In all 4 patients with cirrhosis and suspected tumor<br />

small HCC was found in explanted liver. One <strong>of</strong> these patients<br />

died <strong>of</strong> graft primary non-function. One <strong>of</strong> two patients<br />

who underwent multiple resections before transplantation presented<br />

pulmonary metastases almost 2 years after transplantation<br />

and extensive metastasectomy was performed, however<br />

prognosis for this patient is rather poor. No recurrence or<br />

metastatic dissemination was noted until now among 5 patients<br />

who underwent primary liver transplantation after total<br />

hepatectomy.<br />

Results<br />

Hepatoblastoma<br />

Twelve out <strong>of</strong> 23 (52,2%) children with HBL are alive and<br />

without active disease, while 11 patients died (47,8%). Best<br />

results were obtained with standard therapy consisting <strong>of</strong> chemotherapy,<br />

tumor resection and postoperative chemotherapy<br />

with long term survival <strong>of</strong> 11/20 pts (55,0%), including two<br />

patients who died after transplantation performed due to tumor<br />

recurrence. Among 2 patients who were transplanted primarily<br />

- 1 died 6 months after transplantation due to<br />

posttransplant lymphoproliferative disease. Another one is<br />

alive and well 25 months after Tx.Two additional children<br />

transplanted for tumor recurrence after previous resections died<br />

<strong>of</strong> metastatic disease despite <strong>of</strong> posttransplant chemotherapy.<br />

No graft involvement was observed in these patients (Fig. 1).<br />

Hepatocellular carcinoma<br />

Overall survival among 19 children with HCC is 12/19<br />

(63,2%), 42,9% (3/7) children are alive and free <strong>of</strong> tumor<br />

after CHT and resection, while all other died <strong>of</strong> tumor<br />

3<br />

3<br />

00000000000000000<br />

alive&well<br />

e e ea<br />

e a e alive wi<br />

Fig. 2 Hepatocarcinoma - actual results according to type <strong>of</strong> treatment (1990-<br />

2004)<br />

Posttransplant observation is still relatively short in survivors<br />

(range: 5 - 40 months). AFP concentration remains within<br />

normal limits in all, however it is necessary to say that it was<br />

not elevated in 2 <strong>of</strong> 11 before transplantation (Fig. 3). Chemotherapy<br />

(1-3 courses) was given after transplantation to 5<br />

patiens. It had to be stopped in most <strong>of</strong> them after 1-2 courses<br />

due to complications. Liver graft function is good in all but<br />

one patient who developed severe cholangitis after chemotherapy,<br />

further complicated by intrahepatic bile duct strictures.<br />

1<br />

1<br />

11<br />

0000<br />

000<br />

AFP (ng/ml)<br />

0000<br />

000<br />

0000<br />

1 1<br />

1<br />

000<br />

0<br />

before treatment before a t<br />

0000000000000000000<br />

alive&well<br />

e e ea<br />

e a e<br />

Fig. 1 Hepatoblastoma - results according to type <strong>of</strong> treatment (1990-2004)<br />

Fig. 3 Alphafetoprotein (AFP) in 9 patients with HCC after liver transplantation<br />

(follow up 5-40 months)


113<br />

Discussion<br />

Total hepatectomy with liver transplantation became an important<br />

therapeutic option for children with malignant primary<br />

hepatic tumors [1, 5, 9, 12]. Overall long term survival in pediatric<br />

liver transplant recipients is now reaching in leading<br />

centers 90-95% independently on indication for transplantation,<br />

while survival <strong>of</strong> children with hepatoblastoma and<br />

hepatocarcinoma after conventional treatment (chemotherapy<br />

+ resection) is much lower, 60-80% and 30-40% respectively<br />

[2, 4, 5, 11, 12]. Various protocols and trials have been tried<br />

to improve results <strong>of</strong> conventional treatment, and much success<br />

was achieved independently by SIOPEL group, German<br />

group or Japanese group, mostly in HBL, with less optimistic<br />

results in patients with HCC [9, 10]. The place <strong>of</strong> total hepatectomy<br />

and liver transplantation have been not clearly defined<br />

in pediatric population, although many patients benefited<br />

from this type <strong>of</strong> treatment on the individual qualification basis<br />

[1, 9, 12]. Standard qualification criteria for transplantation<br />

in patients with primary hepatic tumors included in most centers:<br />

unresectable tumor, no extrahepatic tumor extension, no<br />

distant metatases (or in case <strong>of</strong> hepatoblastoma pulmonary<br />

metastases which cleared with chemotherapy), [5, 9]. It is<br />

obvious that conventional complex treatment is best for most<br />

patients with hepatoblastoma, however in our opinion transplantation<br />

should be considered as first surgery in these children<br />

in which tumor is unresectable, but also in cases <strong>of</strong> successful<br />

response <strong>of</strong> large, unresectable tumors to chemotherapy<br />

which allows downstaging <strong>of</strong> the tumor and complete extensive<br />

but marginal resection. Although not all agree that narrow<br />

margin free <strong>of</strong> HBL in resected liver specimen is important<br />

for long term survival, but one should keep in mind that<br />

recurrence <strong>of</strong> HBL in these patients indicates almost uniformly<br />

poor prognosis also after transplantation [2, 5, 9, 12].<br />

Conventional treatment can not <strong>of</strong>fer such good results in children<br />

with hepatocarcinoma despite intensive chemotherapy<br />

and improvements in resection techniques. In adult population<br />

large groups <strong>of</strong> patients with HCC were qualified to liver<br />

transplantation since 25 years, however no criteria for transplantation<br />

in these patients existed until study done by<br />

Mazzaferro et all in 1996 [7, 8]. They defined so called Milano<br />

criteria which have been accepted by most adult transplant<br />

centers. According to these criteria liver transplantation should<br />

be <strong>of</strong>fered to patients with: one single tumor with diameter<br />

less than 5 cm, maximum number <strong>of</strong> 3 tumors with diameters<br />

less than 3 cm each no vascular invasion and no extrahepatic<br />

metastases. With these criteria 4 years survival after liver transplantation<br />

in adults achieve 75%, which is impossible to<br />

achieve by conventional treatment. One can not directly transfer<br />

Milano criteria to pediatric population, as tumor behaviour<br />

and response to chemotherapy in children are different in children<br />

and adults. There are no large enough groups to perform<br />

such analyses in pediatric patients however. Since all our patients<br />

with “non-accidentaloma” did not fit into Milano criteria<br />

one should expect very early tumor recurrence and dissemination<br />

in patients on immunosuppression. We did not<br />

observe such events in any patient till now with medium length<br />

follow-up. Multicenter analysis should be performed probably<br />

to try to define pediatric criteria for liver transplantation<br />

in patients with HCC, as single center can not collect number<br />

<strong>of</strong> patients necessary for such analysis.<br />

In conclusion, liver transplantation should be considered<br />

as an option early in the process <strong>of</strong> treatment <strong>of</strong> primary<br />

malignant tumors in children, particularly with<br />

hepatocarcinoma, and transplantation should be considered<br />

as a primary surgical treatment in all patients with unresectable<br />

or marginally resectable tumors and without extrahepatic extension.<br />

Living related donor transplantation is one <strong>of</strong> important<br />

solution in best timing <strong>of</strong> transplantation within treatment<br />

protocol [6].<br />

References<br />

1. Cillo U, Ciarleglio FA, Bassanello M,<br />

et al (2003) Liver transplantation for the<br />

management <strong>of</strong> hepatoblastoma. Transplant<br />

Proceed 35:2983-2985<br />

2. Dicken BJ, Bigam DL, Lees GM<br />

(2004) Association between surgical<br />

margins and long-term outcome in advanced<br />

hepatoblastoma. J Pediatr Surg<br />

39:721-725<br />

3. European Registry <strong>of</strong> Liver Transplantation<br />

Report (ELTR), 2003.<br />

4. Kaliciñski P (1997) Przeszczep w¹troby.<br />

Wybrane zagadnienia. Fundacja Polski<br />

Przegl¹d Chirurgiczny (Ed), pp 1-138<br />

5. Kamiñski A, Kaliciñski , Ismail H, et<br />

al (2003) Liver transplantation in children<br />

with hepatic tumors. Pediatr Transplant<br />

7(suppl):124<br />

6. Lo CM, Fan ST, Liu CL, Chan SC,<br />

Wong J (2004) The role and limitation<br />

<strong>of</strong> living donor liver transplantation for<br />

hepatocellular carcinoma. Liver Transpl<br />

10:440-447<br />

7. Mazzaferro V, Regalia E, Doci R, et al<br />

(1996) Liver transplantation for the<br />

treatment <strong>of</strong> small hepatocellular carcinomas<br />

in patients with cirrhosis. N Engl<br />

J Med 334:693-699<br />

8. Nart D, Arikan C, Akyildiz M, et al<br />

(2003) Hepatocellular carcinoma in liver<br />

transplant era: a clinicopathologic analysis.<br />

Transplant Proceed 35:2986-2990<br />

9. Otte JB, Pritchard J, Aronson DC, et al<br />

(2004) Liver transplantation for<br />

hepatoblastoma: results from the International<br />

Society <strong>of</strong> Pediatric Oncology<br />

(SIOP) study SIOPEL-1 and review <strong>of</strong><br />

the world experience. Pediatr Blood<br />

Cancer 42:74-83<br />

10. Perilongo G, Shafford E, Maibach R, et<br />

al (2004) Risk-adapted treatment for<br />

childhood hepatoblastoma. Final report<br />

<strong>of</strong> the second study <strong>of</strong> the International<br />

Society <strong>of</strong> <strong>Paediatric</strong> Oncology –<br />

SIOPEL 2. Eur J Cancer 40:411-421<br />

11. Prokurat A, Kluge P, Chrupek M,<br />

Koœciesza A (2002) Possibilities and<br />

limitations in the treatment <strong>of</strong> hepatocellular<br />

carcinoma in children. Ann<br />

Diagn Paediatr Pathol 6:51-57<br />

12. Srinivasan P, McCall J, Pritchard J, et al<br />

(2002) Ortotopic liver transplantation<br />

for unresectable hepatoblastoma. Transplantation<br />

74:652-655


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Hepatocyte ultrastructure in non-hemolytic hyperbilirubinemias<br />

El¿bieta Czarnowska, Bogdan WoŸniewicz<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

Departament <strong>of</strong> <strong>Pathology</strong>,<br />

The Children’s Memory Health Institute,<br />

Warsaw, Poland<br />

Abstract<br />

The non-hemolytic hyperbilirubienias comprise a group <strong>of</strong> syndromes that are clinically and some in cases<br />

biochemically and genetically well characterized, and ultrastructural investigations <strong>of</strong> biopsy samples have<br />

still diagnostic applications. In this study utrastructural analysis <strong>of</strong> biopsy tissues in order to identify pattern<br />

<strong>of</strong> features characteristic for different non-hemolytic hyperbirubinemias among group <strong>of</strong> patients with clinical<br />

presentation <strong>of</strong> hyperbilirubinemia and normal histopathology were done. A group <strong>of</strong> 62 patients was<br />

selected on the basis <strong>of</strong> mild unconjugated hyperbilirubinemia and, in some cases, on the basis <strong>of</strong> a mild<br />

unconjugated or conjugated hyperbilirubinemias in the presence <strong>of</strong> repeatedly normal liver function tests and<br />

absence <strong>of</strong> hyperhemolysis. Electron microscopic investigation distinguished between patients in three groups<br />

<strong>of</strong> primary hyperbilirubiemia: Gilbert syndrome (21 pts), Dubin-Johnson syndrome (5 pts), Rotor syndrome<br />

(3 pts) and a group with non specific changes (33 pts). The ultrastructural features <strong>of</strong> hepatocytes that univocally<br />

distinguish primary hyperbilirubinemias from secondary diseases were presented. Electron microscopic<br />

methods permit on recognition <strong>of</strong> non-specific changes in the ultrastructure <strong>of</strong> liver tissue among samples<br />

obtained from patients with non-hemolytic hyperbilirubinemias and can be still usefull in diagnosis <strong>of</strong><br />

jaundince.<br />

Key words: congenital hyperbilirubinemia, Dubin-Johnson disease, Gilbert disease, Rotor disease, ultrastructure<br />

Introduction<br />

Idiopathic hyperbilirubinemias may encompass several primary<br />

and secondary diseases <strong>of</strong> bilirubin metabolism [2]. Bilirubin,<br />

an oxidative product <strong>of</strong> the hem group <strong>of</strong> hemoglobin,<br />

myoglobin and cytochrome P-450, in the liver is conjugated<br />

with glucuronic acid and than excreted in bile via ATPdependent<br />

transporter [5]. Currently, injuries <strong>of</strong> molecular<br />

bases <strong>of</strong> this metabolic pathway are known [5]. Although, this<br />

knowledge allow for precision diagnosis <strong>of</strong> non-hemolytic<br />

hyperbilirubinemias in most cases, but still ultrastructural investigation<br />

<strong>of</strong> biopsy samples have diagnostic application.<br />

Clinically, conjugated and unconjugated hyperbilirubinemia<br />

can be distinguished on the basis <strong>of</strong> blood bilirubin level, normal<br />

liver tests and exclusion <strong>of</strong> hemolytic disease. In conjugated<br />

hyperbilirubinemias i.e Gilbert, Crigler-Najjar syndrome,<br />

transport <strong>of</strong> bilirubin from plasma into liver cells is<br />

impaired, while in conjugated hyperbilirubinemias, i.e. Dubin-<br />

Johnson, Rotor syndrome, microsomal enzymes transferase,<br />

bilirubin transport from hepatic cells into bile caniculi system<br />

are disturbed [2, 5]. In non-hemolytic hyperbilirubinemias,<br />

liver histopathology may present normal feature or cholestasis.<br />

These investigations were aimed to identify ultrastructural<br />

pattern <strong>of</strong> characteristic features for different nonhemolytic<br />

hyperbirubinemias among group <strong>of</strong> patients with<br />

clinical presentation <strong>of</strong> hyperbilirubinemia and normal histopathology.<br />

Material and methods<br />

Material<br />

62 biopsy samples from patients (age 6-17 years, mean 10,2<br />

± 4,6) with marked liver disfunction and clinical symptoms<br />

<strong>of</strong> non-hemolytic hyperbilirubinemia were selected for electron<br />

microscopic studies among samples collected in the registry<br />

<strong>of</strong> <strong>Pathology</strong> Department <strong>of</strong> CMHI. Tissue samples were<br />

routinely preserved for histological, histochemical and ultrastructural<br />

examination. All <strong>of</strong> the examinated patients had total<br />

bilirubin in the range <strong>of</strong> 1,3-5,1 mg% (Table 1).<br />

Address for correspondence<br />

Elzbieta Czarnowska, PhD<br />

Department <strong>of</strong> <strong>Pathology</strong><br />

The Children's Memory Health Institute<br />

Al. Dzieci Polskich 20, 04-730 Warsaw, Poland<br />

Phone: +48 22 815 48 15<br />

E-mail: czar@czd.waw.pl


116<br />

Table 1<br />

Total and direct bilirubin level in serum in distinguished groups <strong>of</strong> primary hyperbilirubinemia and with non-specific changes<br />

Disease<br />

Gilbert<br />

Compl.<br />

Incompl.<br />

Number <strong>of</strong> patients Age (years)<br />

10<br />

11<br />

10-17<br />

6-7<br />

Bilirubin (mg%)<br />

Total Average Direct Ave<br />

1,5-4,6 2,56 0,5-0,9 0,62<br />

1,2-5,1 2,48 0,3-1,1 0,95<br />

Dubin-Johnson 5 9-17 1,3-4,7 2,54 0,6-2,0 1,05<br />

Rotor 3 16-17 1,6-4,5 2,10 0,4-0,8 0,55<br />

Non specific 33 7-17 1,2-5,7 2,62 0,2-2,8 1,45<br />

Histology<br />

Five mm thick paraffin sections were stained in a routine manner<br />

with hematoxylin-eosin, azan, silver, PAS, PAS with<br />

diastaze digestion and than reviewed to exclude other disease<br />

than hyperbilirubinemia.<br />

Electron microscopy<br />

Samples were fixed in 2.5% glutaraldehyde in cacodylate<br />

buffer at pH 7.3, postfixed in 2% OsO 4,<br />

dehydrated in alcohols<br />

and embedded in Epon 812. Thick sections stained with toluidine<br />

blue were analysed under light microscope. Ultrathin<br />

sections stained with uranyl acetate and lead citrate were examined<br />

by electron microscopy.<br />

A<br />

B<br />

Results<br />

Examinations <strong>of</strong> paraffin and semi-thin sections showed in all<br />

cases an apparently normal liver, except for Dubin-Johnson<br />

that shown the existence <strong>of</strong> dark pigment predominantly in<br />

centrolobular areas.<br />

On the basis <strong>of</strong> EM analysis morphological equivalents<br />

<strong>of</strong> primary congenital hyperbilirubinemias were confirmed<br />

in 29 cases and secondary reactive symptomatic<br />

hyperbilirubinemias were recognized in 33 cases.<br />

Gilbert syndrome<br />

Gilbert syndrome was diagnosed in 21 cases. There were observed<br />

shortening and mal-developed vascular pole microvilli<br />

or loss <strong>of</strong> microvilli, and increase <strong>of</strong> collagen in the Disse<br />

space in various intensity. In all patients an increase <strong>of</strong> the<br />

endoplasmic reticulum pr<strong>of</strong>iles with coexisting dilatation <strong>of</strong><br />

rough endoplasmic reticulum with degranulation and pigment<br />

granules variable in size containing both electron dense material<br />

which alternate with less dense material and lipid droplets<br />

was observed (Fig. 1A). In 10 biopsy samples enlarged or<br />

giant, oval or polymorphic mitochondria with paracrystalline<br />

inclusions (Fig. 1B) were present. In all cases the biliary pole<br />

<strong>of</strong> hepatocytes shown normal caniculi and microvilli, but with<br />

numerous pigment granules around. Lip<strong>of</strong>uscin granules were<br />

Fig. 1 Gilbert syndrome: (A) vascular pole <strong>of</strong> hepatocyte with single microvilli.<br />

Mag. x 40 000, (B) mitochondrium containing paracrystallines, mag. x<br />

22 000<br />

common finding. The remaining group <strong>of</strong> 11 patients showed<br />

uncompleted ultrastructural features <strong>of</strong> Gilbert disease, i.e. lack<br />

<strong>of</strong> giant mitochondria and no paracristalline in mitochondrial<br />

matrix.<br />

Dubin-Johnson syndrome<br />

Dubin-Johnson syndrome was defined in 5 cases. There<br />

were not seen alterations in the vascular pole <strong>of</strong> hepatocytes,<br />

which exhibit normal microvilli. Slightly increased number<br />

<strong>of</strong> smooth endoplasmic reticulum pr<strong>of</strong>iles that formed small<br />

vesicles, normal mitochondria with some variation in size,<br />

well developed Golgi pr<strong>of</strong>iles located around bile canicules<br />

were common features. Dilation <strong>of</strong> bile caniculi with loss <strong>of</strong><br />

microvilli and characteristic pigment granules around were<br />

seen (Fig. 2). Pigment granules were composed mainly <strong>of</strong><br />

granular material, however vesicle-like or lipid components<br />

were also seen. They were polygonal in shape and delineated<br />

by a single membrane. The lack <strong>of</strong> lip<strong>of</strong>uscin in hepatocytes<br />

was common feature.


117<br />

Fig. 2 Dubin-Johnson syndrome: a bile pole <strong>of</strong> hepatocytes with pigment<br />

granules around the bile canicule. Mag.x 30 000<br />

Rotor Syndrome<br />

Rotor syndrome was recognised in 3 cases. Only slight changes<br />

in hepatocytes were observed: some <strong>of</strong> the bile canicules were<br />

dilated and exhibited reduced number <strong>of</strong> microvilli, the<br />

interhepatocytic junctional interspaces were dilatated and cell<br />

membranes along the lateral cell surface formed microvilli<br />

(Fig. 3a), cell membranes facing the Disse space showed reduced<br />

number <strong>of</strong> microvilli. In side <strong>of</strong> hepatocytes a few pleomorphic<br />

mitochondria or megamitochondria with<br />

paracristalline were seen. The most characteristic feature was<br />

lack <strong>of</strong> lipid component in pigment granule that composed <strong>of</strong><br />

electron dense and less dense granular material. They were<br />

distributed mainly around bile canicules (Fig. 3b).<br />

Non characteristic lesions<br />

Non characteristic ultrastructural fine lesions accompanying<br />

secondary hyperbilirubinemia were found in 33 cases. Reactive<br />

changes were recognized in 9 cases. There were increased<br />

number and elongation <strong>of</strong> microvilli on the vascular pool (Fig.<br />

4), polymorphic mitochondria (sometimes with paracrystalline<br />

inclusions), increased number <strong>of</strong> smooth endoplasmic reticulum<br />

pr<strong>of</strong>iles, the presence <strong>of</strong> single lipid droplets and pigment<br />

or lip<strong>of</strong>uscin granules in cytoplasm, and collagen fibrils<br />

in Disse spaces.<br />

Fig. 4 Non specific changes. Vascular pole <strong>of</strong> hepatocyte with numerous,<br />

elongated microvilli. Mag.x 40 000<br />

Discussion<br />

In about 50% <strong>of</strong> the patients with clinical symptoms <strong>of</strong> hyperbilirubinemia<br />

electron microscopy confirmed primary congenital<br />

hyperbilirubinemia. Among <strong>of</strong> them, complete ultrastructural<br />

features <strong>of</strong> Gilbert syndrome were presented in 10 cases,<br />

uncomplete Gilbert in 11 cases, Dubin-Johnson in 5 cases<br />

and Rotor in 3 cases.<br />

In seventies the ultrastructural observations were helpful<br />

to establish the morphological equivalent <strong>of</strong> primary hyperbilirubinemia<br />

and distinguish combination <strong>of</strong> defects impairing<br />

primary hyperbilirubinemias, particularly in Gilbert<br />

syndrome when complete and incomplete features coexisted<br />

[1, 4, 6]. Our patients with Gilbert disease clinically were characterized<br />

by relatively low-grade direct bilirubin and they<br />

reached a higher proportion <strong>of</strong> unconjugated bilirubin. These<br />

abnormalities were correlated with decrease or the lack <strong>of</strong> microvilli<br />

at sinusoidal pole.<br />

It is known that the liver in Dubin-Johnson syndrome<br />

might be characterized by slight and inconsistant changes. In<br />

our patients, the canicular pole exhibited normal microvilli or<br />

disappearing microvilli coexisting with a slightly dilated lumen.<br />

These results suggest that disturbances <strong>of</strong> bile flow were<br />

caused by different disfunctions <strong>of</strong> the canicular area. These<br />

are in agreement <strong>of</strong> with distinct mechanism <strong>of</strong> bilirubin glucuronide<br />

transport involving ATP-dependent and ATP-independent<br />

system that have been proposed [7].<br />

Fig. 3 Rotor syndrome: (A) lateral hepatocytes<br />

surface with number <strong>of</strong> microvilli. Mag. x<br />

50 000; (B) pigment granules composed <strong>of</strong><br />

dense granular and lipid material, mag.x20 000<br />

A<br />

B


Patients with Rotor syndrome ultrastructurally presented fairly<br />

similar features to Gilbert and to Dubin-Johnson simultaneously:<br />

some injury <strong>of</strong> vascular and canicular pole concomitant<br />

with an increase <strong>of</strong> smooth endoplasmic reticulum and<br />

pleomorphic mitochondria in hepatocytes. These similarities<br />

between some ultrastructural features <strong>of</strong> Gilbert and Dubin-<br />

Johnson syndrome were signaled in literature before [8]. The<br />

presence <strong>of</strong> microvilli along the lateral surface <strong>of</strong> hepatocytes<br />

was a particular feature, which distinguished patients with<br />

Rotor syndrome from other patients. Patients with Rotor syndrome<br />

exhibited lower level <strong>of</strong> direct bilirubin than<br />

unconjugated, and this observation does not agree with main<br />

idea <strong>of</strong> liver defect [5]. However, similar data on a direct bilirubin<br />

in serum have been mentioned in literature [3].<br />

Electron microscopic methods permit on recognition<br />

<strong>of</strong> non-specific changes in the ultrastructure <strong>of</strong> the liver tissue<br />

among samples obtained from patients with non-hemolytic<br />

hyperbilirubinemias and can be still usefull in diagnosis <strong>of</strong><br />

jaundince.<br />

References<br />

1. Bethelot P, Dhumeaux D (1978) New<br />

signts in the classification and mechanism<br />

<strong>of</strong> hereditiary, chronic, nonhaemolytic<br />

hyperbilirubinemias. Gut<br />

19:474-480<br />

2. Chowdhury JR, Wolk<strong>of</strong>f AW, Wolk<strong>of</strong>f<br />

N, Chodhury R, Arias JM (1995)<br />

Hereditiary jaundice and disorders <strong>of</strong><br />

bilirubin metabolism. In: Scriver ChR,<br />

Beaudet AL, Sly WS, Walle D (eds) The<br />

metabolic basis <strong>of</strong> inherited diseases,<br />

New York, Vol 1, pp 2161-2208<br />

3. Czarnecki J, Cichosz H, Slomke M,<br />

Siezieniewska G, Czechowska G (1988)<br />

Clinical, biochemical and morphologic<br />

analysis <strong>of</strong> congenital non-hemolytic<br />

hyperbilirubinemia. Wiad Lek<br />

11(17):1151-1156 (in Polish)<br />

4. Dawson J, Carr-Loske DL, Talbot JC,<br />

Rossenthal FD (1978) Hepatic ultrastructure<br />

in Gilbert syndrome: evidence<br />

two populations. Gut 19A:995-999<br />

5. Iyanagi T, Emi Y, Ikushiro S (1998) Biochemical<br />

and molecular aspects <strong>of</strong> genetic<br />

disorders <strong>of</strong> bilirubin metabolism.<br />

Biochem Biophys 1407:173-184<br />

6. McGee JOD, Allan JG, Russel RJ,<br />

Patrick RS (19750 Liver ultrastructure<br />

in Gilbert syndrom. Gut 16: 220-224<br />

7. Nishida T, Gatmaitan Z, Roy Chodhry<br />

R, Arias IM (1992) Two distinct mechanisms<br />

for bilirubin glucuronide transport<br />

by rat ble membrane vesicles. Demonstration<br />

<strong>of</strong> defective ATP-dependent<br />

transport in rats (TR-) with inherited<br />

conjugated hyperbilirubinemia. J Clin<br />

Invest 90:21-2135<br />

8. Okolicsanyi L, Torrado A, Nussle D,<br />

Gardid D, Gautier A (1969) Etude<br />

clinique et ultrastructurale d’un cas de<br />

syndrome de Rotor. Revue Int Hepat<br />

19:393-398


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Congenital Heart Diseases Database: analysis <strong>of</strong> occurrence,<br />

diagnostics and coexistence with anomalies <strong>of</strong> other systems.<br />

Single institution experience<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

Anna Rybak, Aneta Wójcik, Bogdan WoŸniewicz<br />

Department <strong>of</strong> <strong>Pathology</strong><br />

The Children’s Memorial Health Institute<br />

Warsaw, Poland<br />

Abstract<br />

The Congenital Heart Diseases Database was created in the Children’s Memorial Health Institute during<br />

the period <strong>of</strong> 2001 – 2003. It contains over 1000 preparations <strong>of</strong> congenital heart defects <strong>of</strong> children aged<br />

from 0 till 14 years. According to our data, the most common defect was ventricular septal defect. The<br />

compatibility <strong>of</strong> the clinic and pathological diagnoses was high (about 80% <strong>of</strong> the diagnoses were confirmed),<br />

however significantly higher after 1990. Although in our Database plenty <strong>of</strong> coexisting anomalies<br />

and diseases <strong>of</strong> the other systems or organs occurred, we found no significant relations between them and<br />

congenital heart defects.<br />

Key words: congenital heart disease, database, heart defects<br />

Introduction<br />

Since 1980 until now, the Children’s Memorial Health Institute<br />

<strong>of</strong> Warsaw has assembled over 1200 congenital heart defects<br />

preparations from patients aged 0-14 years. Between<br />

September 2001 and March 2003 we have made the repeated<br />

sections <strong>of</strong> those preparations and created “The Congenital<br />

Heart Diseases Database” (CHDDb) that includes 1037 records<br />

with full descriptions <strong>of</strong> the hearts’ and defects’ anatomy, clinical<br />

and sectional diagnoses, anomalies <strong>of</strong> other systems and<br />

performed operations. This is to present the direct conclusions<br />

that have arisen from this Database, including the frequency<br />

<strong>of</strong> occurrence <strong>of</strong> the particular heart defects, compatibility <strong>of</strong><br />

the clinical and autopsy diagnoses and finally, regularity in<br />

coexistence <strong>of</strong> the congenital heart defects with the anomalies<br />

<strong>of</strong> the other systems.<br />

Material and methods<br />

The Congenital Heart Diseases Database is composed <strong>of</strong> 1037<br />

preparations <strong>of</strong> the hearts with the congenital defects. They<br />

are fully described in four files which contain personal patients’<br />

data, clinical diagnoses with the descriptions <strong>of</strong> defects<br />

Address for correspondence<br />

Bogdan Wozniewicz<br />

Department <strong>of</strong> <strong>Pathology</strong><br />

The Children's Memorial Health Institute<br />

Aleja Dzieci Polskich 20, 04-736 Warsaw, Poland<br />

<strong>of</strong> the other systems and types <strong>of</strong> performed operations, pathologic<br />

diagnoses <strong>of</strong> the heart defect and coexisting diseases.<br />

The last file shows complete measurement <strong>of</strong> all the hearts<br />

including external dimensions as well as internal measurements<br />

<strong>of</strong> the valves, arteries, veins or myocardium.<br />

The patients’ age was between 1 day and 14 years.<br />

There were 437 female and 600 male patients described and<br />

they were all admitted to Children’s Memorial Health Institute<br />

during the period 1980 – 2003 and almost half <strong>of</strong> them<br />

were operated because <strong>of</strong> the heart defect.<br />

A nomenclature for the congenital heart disease was<br />

based on the International Nomenclature for Congenital Heart<br />

Surgery that was <strong>of</strong>ficially adopted at the Annual Meeting <strong>of</strong><br />

the EACTS in Glasgow on September 6, 1999 [3].<br />

Results and discussion<br />

Occurrence <strong>of</strong> the congenital heart defects<br />

The most common anomaly that appeared in our material was<br />

ventricular septal defect (VSD) with the frequency just under<br />

30% (Fig.1). In details, there were 31 cases <strong>of</strong> VSD in muscular<br />

part <strong>of</strong> the septum (VSD single–29; VSD multiple-2 cases)<br />

and 259 cases <strong>of</strong> this defect in other parts <strong>of</strong> the interventricular<br />

septum (VSD; NOS).<br />

Phone: +4822-815-1960<br />

E-mail: b.wozniewicz@czd.waw.pl


%<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

VSD PDA TGA ASD TOF AVC CoA HLHS PA HRV PS DORVTAPVC<br />

Fig. 1 Occurrence <strong>of</strong> the congenital heart diseases<br />

The frequency <strong>of</strong> patent arterial duct (PDA) was about 19%.<br />

Similarly, other congenital heart defects like transposition <strong>of</strong><br />

the great arteries (TGA) and atrial septal defect (ASD) are<br />

about 18%. Although there were over 160 ASD diagnosed,<br />

the largest group was due to persisted foramen ovale (PFO )<br />

type (50%). ASD secundum type appeared in 69 cases that<br />

was just over 40% <strong>of</strong> all atrial defects. Other atrial defects,<br />

like ASD type sinus venosus, coronary sinus type or single<br />

atrium, appeared rarely (14 cases).<br />

Tetralogy <strong>of</strong> Fallot (TOF) was diagnosed in about 12%<br />

<strong>of</strong> all cases. Atriovertricular canal (AV canal) as well as coarctation<br />

<strong>of</strong> aorta (CoA) concerned 9% <strong>of</strong> all heart defects. Hypoplastic<br />

left heart syndrome (HLHS) likewise pulmonary atresia<br />

(PA) were visible in 7% <strong>of</strong> the examined preparations. Similarly<br />

hypoplastic right ventricle (HRV) or pulmonary stenosis<br />

(6,5%). Double outlet from the right ventricle (DORV) and total<br />

anomalous pulmonary venous connection (TAPVC) appeared<br />

in less than 5% <strong>of</strong> all cases. The rarest were aortic atresia (3<br />

patients), aortic valve hypoplasia (1 patient), pseudotruncus (3<br />

patients) and inversion <strong>of</strong> the ventricles (2 patients).<br />

There is also some regularity with reference to the gender<br />

<strong>of</strong> the patients. The difference is most significant in the<br />

group <strong>of</strong> diseases like partial anomalous pulmonary venous<br />

connection or right ventricle outlet obstruction, which occurred<br />

two to five times more <strong>of</strong>ten in the female group than in the<br />

male group. However aortic atresia, Bland-White-Garland syndrome<br />

and cor triatriatum occurred in our material only in<br />

reference to the male group. Such defects like truncus arteriosus,<br />

aortic stenosis, HLHS, TGA or mitral atresia occurred<br />

over two times more <strong>of</strong>ten in the group <strong>of</strong> boys than in the<br />

group <strong>of</strong> girls.<br />

Clinical and autopsy diagnosis<br />

There are 1037 congenital heart diseases in the Database, however<br />

clinical diagnostics and diagnoses were established due<br />

to 867 patients. In this group, autopsy diagnoses differed from<br />

clinical diagnoses in about 19% (166 cases).<br />

Clinical diagnostics was based on physical examination, ECG,<br />

blood pressure measurement, echocardiography and invasive<br />

cardiology, which was used from the beginning 1990. Because<br />

<strong>of</strong> that fact, we decided to divide our data into two groups: in<br />

the first group there were patients that had been admitted to<br />

the Children’s Memorial Health Institute till the end <strong>of</strong> 1989<br />

(533 patients); the second group included rest <strong>of</strong> the patients<br />

(334 patients). The results <strong>of</strong> a comparison between clinical<br />

and pathologic diagnoses were as follows: 166 children (22%)<br />

from the first group were misdiagnosed during their hospitalization.<br />

In the second group, where the patients were additionally<br />

diagnosed by means <strong>of</strong> the invasive cardiology, the<br />

number <strong>of</strong> misdiagnoses has fallen to 15% (Table 1). Among<br />

166 incorrectly diagnosed congenital heart defects, the most<br />

<strong>of</strong>ten was HLHS (about 13%), than TGA and AV canal (just<br />

under 9%). In contrast, the greatest conformity between clinic<br />

and autopsy was due to interrupted aortic arch (over 90%)<br />

and TAPVC or PDA (about 80%).<br />

Table 1<br />

Compatibility between clinical and autopsy diagnoses<br />

Period<br />

1980-1989 78%<br />

1990-2003 85%<br />

Compatibility<br />

Coexistence <strong>of</strong> the congenital heart defects with the<br />

anomalies <strong>of</strong> other systems<br />

In reference to the CHDDb there were no significant relations<br />

between heart defects and other anomalies. The most common<br />

were malformations <strong>of</strong> the alimentary tract, polysplenia,<br />

asplenia, urinary tract defects and additional superior caval<br />

vein which occurred mainly in reference to heart defects like<br />

AV canal, coarctation <strong>of</strong> aorta, TAPVC and TOF. There were<br />

eight patients with situs inversus, although with different heart<br />

defects.<br />

In the group <strong>of</strong> patients with chromosome aberrations<br />

there were 24 patients with Down syndrome. Over 62% <strong>of</strong><br />

these patients suffered from atrioventricular canal and about<br />

34% from VSD. There was one patient with Patau syndrome<br />

that had coarctation <strong>of</strong> aorta and one patient with Edward’s<br />

syndrome who suffered from VSD.<br />

In our Database occurred six patients with developmental<br />

multiply malformations. Three patients with Pierre-<br />

Robin sequence suffered from ASD, VSD and TOF. Two children<br />

with DiGeorge syndrome had TOF and tricuspid valve<br />

atresia, while in literature, the most common are the anomalies<br />

<strong>of</strong> the aortic arch [1, 2]. The congenital heart disease <strong>of</strong><br />

one patient with Noonan syndrome was AV canal (in literature,<br />

over 50% patients with Noonan syndrome suffer from<br />

pulmonary stenosis) [2].<br />

References<br />

1. Becker AE, Anderson RH (1985) Pathologie des Herzens. Georg<br />

Thieme Verlag<br />

2. Connor M, Ferguson-Smith M (1998) Podstawy genetyki<br />

medycznej, PZWL (in Polish)<br />

3. Lacour-Gayet F, Maruszewski B, Mavroudis C, Jacobs JP, Elliott<br />

MJ (2000) Presentation <strong>of</strong> the International Nomenclature for<br />

Congenital Heart Surgery. Eur Journ Card-Thor Surg 18:128-<br />

135


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

Solid pseudopapillary tumor <strong>of</strong> pancreas.<br />

Case report and the review <strong>of</strong> literature<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

Andrzej Chilarski 1 , Anna Piaseczna-Piotrowska 1 , Jan Strzelczyk 2 , Stanis³aw £ukaszek 3 , Andrzej Kulig 3<br />

1<br />

Depart. <strong>of</strong> Pediatric Surgery<br />

3<br />

Depart. <strong>of</strong> <strong>Pathology</strong>,<br />

Polish Mother’s Health Institute,<br />

Lodz, Poland<br />

2<br />

Depart. <strong>of</strong> General Surgery and Transplantology,<br />

Medical University,<br />

Lodz, Poland<br />

Abstract<br />

Authors describe the case <strong>of</strong> 14 year-old girl operated on because <strong>of</strong> the neoplasm <strong>of</strong> tail <strong>of</strong> pancreas proved<br />

to be solid pseudopapillary pancreatic tumor. Radical resection was curative. After surgery she made uneventful<br />

recovery being symptoms’ free for 8 months. The review <strong>of</strong> literature on this rare, relatively low,<br />

malignant tumor is presented.<br />

Key words: pancreas, pseudopapillary tumor, solid tumors<br />

Introduction<br />

Solid pseudopapillary tumors <strong>of</strong> pancreas (SPTP) first described<br />

by Frantz in 1959 are rare findings among other pancreatic<br />

tumors [5, 6, 9, 10, 11, 13]. To date a bit more then 500<br />

cases have been described in the literature [16]. SPTP occurs<br />

predominantly in teenage girls and young women although<br />

all age groups may be affected and also males are not disease<br />

– free [12, 15, 17, 18].<br />

The tumors are reckoned as a low-grade malignant papillary<br />

– cystic neoplasms <strong>of</strong> the pancreas <strong>of</strong> unclear histogenetic<br />

origin, but with highly characteristic, distinct histology and<br />

usually quite benign biologic behaviour [3, 9, 10, 13, 15, 18].<br />

Their clinical presentation is vague and non characteristic. The<br />

most common symptoms are abdominal pains, nausea, emesis,<br />

sometimes palpable abdominal mass and jaundice – if the<br />

head <strong>of</strong> pancreas is involved [1]. Asymptomatic course is not<br />

uncommon and diagnosis can be established incidentally following<br />

blunt abdominal trauma [3, 9, 19]. Complete resection<br />

is usually curative and so the radical surgery is the treatment<br />

<strong>of</strong> choice. No adjuvant therapy is recommended.<br />

Case report<br />

Patient<br />

Girl 14-year-old girl (J. P.; files 054774B) was transferred to<br />

Department <strong>of</strong> Pediatric Surgery, Polish Mother’s Health Institute<br />

from local hospital because <strong>of</strong> suspected pancreatic tumor.<br />

She gave the history <strong>of</strong> year-long, moderate, occasional<br />

abdominal pain and nausea and single incident <strong>of</strong> syncope<br />

just before admission to the hospital. Her general condition<br />

was quite good. Physical examination revealed palpable mass<br />

in upper abdomen on left side. No other abnormalities have<br />

been detected. Laboratory investigations were all within normal<br />

range. In abdominal ultrasonography a solid tumor in left<br />

upper quadrant, originating probably from pancreating tail and<br />

reaching spleen, was found. CT scan confirmed this finding<br />

(Fig. 1) showing the tumor <strong>of</strong> pancreas tail about 6 cm in<br />

diameter in the vicinity <strong>of</strong> spleen and its vessels. The indications<br />

for surgical treatment were established. Exposure <strong>of</strong> the<br />

pancreas revealed the presence <strong>of</strong> the well vascularized mass<br />

arising from the tail <strong>of</strong> pancreas and reaching to the hilum <strong>of</strong><br />

the spleen. Main splenic vessels were surrounded by the tumor<br />

and could not be liberated. The resection <strong>of</strong> the tumor<br />

Address for correspondence<br />

Andrzej Chilarski,<br />

Depart. <strong>of</strong> Pediatric Surgery,<br />

Polish Mother's Health Institute, Lodz<br />

Rzegowska St. 281/289<br />

Lodz, Poland<br />

Phone: + 48 42 2712136<br />

E-mail: klinikachirdziec@poczta.onet.pl


122<br />

In the cystic part <strong>of</strong> the tumor there were foci <strong>of</strong> degeneration<br />

and haemorrhage resulting in creation <strong>of</strong> pseudocystic and<br />

pseudopapillary pattern, particularly around small vessels (Fig.<br />

3). No vascular invasion was found. On the borderline <strong>of</strong> the<br />

tumor and pancreatic parenchyma a few otherwise normal<br />

glands were found with the lining partly consisting <strong>of</strong> tumor<br />

cells (Fig. 4). The tumor cells showed: positive immunostaining<br />

for á 1<br />

antitrypsin (Fig. 5), chromogranin, vimentin, neuron –<br />

specific enolase (NSE) and synaptophysin. The final histopathologic<br />

diagnosis was - solid pseudopapillary tumor <strong>of</strong> pancreas<br />

(SPTP).<br />

Fig. 1 CT scan: tumor <strong>of</strong> the tail <strong>of</strong> pancreas<br />

was performed “en bloc” with the spleen and distal part <strong>of</strong> the<br />

pancreas 1 cm from the borderline – within healthy tissue.<br />

The hemostasis was achieved and the line <strong>of</strong> resection <strong>of</strong> pancreatic<br />

tissue protected with continuous 4.0 nonabsorbable<br />

suture. No other abnormalities were detected in abdominal<br />

cavity and parietes were closed in layers.<br />

Because <strong>of</strong> splenectomy that has been performed together<br />

with the tumor’s resection - vaccinations against Streptococcus<br />

pneumoniae, Haemophilus influenzae and Neisseria<br />

meningitidis were carried out. Now, eight months postoperatively<br />

girl is well, free <strong>of</strong> symptoms. The patient has made an<br />

uneventful recovery and was discharged home on 12 th postoperative<br />

day.<br />

Histopathology<br />

The specimen consisted <strong>of</strong> tumor – 6 cm in diameter with margin<br />

<strong>of</strong> normal pancreatic tissue attached to the hilum <strong>of</strong> the spleen,<br />

without its infiltration, encompassing splenic vessels. The tumor<br />

was well demarcated, its architecture was partly solid and<br />

partly cystic. The solid part was built <strong>of</strong> unifrom, medium-size<br />

cells with faintly eosinophilic cytoplasm and round nuclei. Mitoses<br />

were not numerous, but foci <strong>of</strong> increased mitotic activity<br />

were present as well. The tumor’s cells formed solid structures,<br />

cords and trabeculae (Fig. 2).<br />

Fig. 3 Pseudopapillary pattern, particularly around small vessels. (HE 100x)<br />

Fig. 4 Tumor's cells within the lining <strong>of</strong> some glands. (HE 200x)<br />

Fig. 2 General architecture <strong>of</strong> the tumor - the solid part. Two mitoses are<br />

present. (HE 100x)<br />

Fig. 5 Positive immunostaining for alfa1 antitrypsin. (HE 100x)


123<br />

Discussion<br />

Preoperative diagnosis <strong>of</strong> pancreatic tumor, putting aside uncharacteristic<br />

clinical symptoms as far as its size, localization,<br />

detailed anatomy and topography is concerned, is based<br />

upon ultrasonography, computed tomoghraphy, magnetic resonance<br />

image [4, 7, 15, 17]. Still diagnostic failures, such as<br />

pseudocyst or ruptured hepatic mass are described in literature<br />

[17, 19]. Some authors advocate preoperative fine needle<br />

biopsy to establish exact histopatology <strong>of</strong> neoplasm [3, 17].<br />

However such a policy is controversial as needle biopsy may<br />

not allow an undisputed diagnosis and the indications for operation<br />

exist, this way or another. This was a case in our patient,<br />

too. Aggressive surgery and complete resection produces<br />

the specimen to be meticulously examined [1, 5, 13, 15, 16].<br />

In 1996 solid pseudopapillary tumor was introduced in<br />

WHO classification <strong>of</strong> tumors <strong>of</strong> the exocrine pancreas [8, 11].<br />

It is usually included into cystic group <strong>of</strong> exocrine neoplasm <strong>of</strong><br />

pancreas that encompasses [8, 20]:<br />

- serous cystis neoplasms,<br />

- mucinous cystic neoplasms,<br />

- solid papillary neoplasms,<br />

- intraductal pancreatic mucinous neoplasm.<br />

Prognosis for the patients with these tumors is much<br />

better than that <strong>of</strong> patients with adenocarcinoma, although in<br />

about 5% <strong>of</strong> patients with SPTP malignant course with metastases<br />

and peritoneal spread can develop [2, 6]. It is estimated<br />

that after surgical resection the outcome is excellent<br />

resulting in long – term survival in about 90% <strong>of</strong> patients [5,<br />

9, 15, 16, 18]. Complete radical resection may need extension<br />

to neighbouring organs e. g. transverse colon or spleen – as<br />

was a case in our patient. Recurrence has been described in<br />

approximately 10% <strong>of</strong> cases [2, 5, 10, 15]. Solid<br />

pseudopapillary tumor <strong>of</strong> pancreas should be always taken<br />

into consideration in the differential diagnosis <strong>of</strong> pancreatic<br />

neoplasm.<br />

Acknowledgements<br />

Authors greatly appreciate the assistance <strong>of</strong> Tomasz Krawczyk,<br />

MD in preparing microscopic pictures.<br />

References<br />

1. Akiyama H, Ono K, Takano M, Sumida<br />

K, Ikuta K, Miyamoto O (2002) Solidpsudopapillary<br />

tumor <strong>of</strong> the pancreatic<br />

head causing marked distal atrophy: a<br />

tumor originated posterior to the main<br />

pancreatic duct. Int Gastrointest Cancer<br />

32(1):29-31<br />

2. Andronikou S, Moon A, Usser R (2003)<br />

Peritoneal metastatic disease in a child<br />

after excision <strong>of</strong> a solid pseudopapillary<br />

tumor <strong>of</strong> the pancreas: a unique case.<br />

Pediatr Radiol 33(4):269-271<br />

3. Asthon J, Sutherland F, Nixon J, Nayak<br />

V (2003) A case <strong>of</strong> solid-pseudopapillary<br />

tumor <strong>of</strong> the pancreas: preoperative cyst<br />

fluid analysis and treatment by<br />

enueclation. Hepatogastroenterology<br />

50(54):2239-2241<br />

4. Cantisani V, Mortele KJ, Levy A,<br />

Glickman JN, Ricci P, Passariello R, Ros<br />

PR, Silverman SG (2003) MR imaging<br />

features <strong>of</strong> solid pseudopapillary tumor<br />

<strong>of</strong> the pancreas in adult and pediatric<br />

patients. AJR Am J Roentgenol<br />

181(2):395-401<br />

5. Cervantes-Montiel F, Florez-Zorrilla C,<br />

Alvarez-Martinez I (2002) Solid-cystic<br />

pseudopapillary tumor <strong>of</strong> the pancreas:<br />

acute post-traumatic presentation. Case<br />

report review <strong>of</strong> the literature. Rev<br />

Gastroenterol Mex 67(2):93-96<br />

6. Illert B, Luhrs H, Timmermann W,<br />

Muller JG (2003) Solid-pseudopapillary<br />

tumor <strong>of</strong> the pancreas as differential diagnosis<br />

in pancreatic tumors <strong>of</strong> the<br />

young. Zentralbl Chir 128(5):438-442<br />

7. Kato T, Egawa N, Kamisawa T, Tu Y,<br />

Sanaka M, Sakaki N, Okamoto A, Bando<br />

N, Funata N, Isoyama T (2002) A case<br />

<strong>of</strong> solid psudopapillary neoplasm <strong>of</strong> the<br />

pancreas and tumor doubling time.<br />

Pancreatology 2(5):495-498<br />

8. Kloppel G, Bogomoletz WV (2003)<br />

<strong>Diagnostic</strong> histopathology <strong>of</strong> tumors.<br />

Ch. Fletcher (Ed), Churchill,<br />

Livingstone, p. 470<br />

9. Mancini GJ, Dudrick PS, Grindstaff<br />

AD, Bell JL (2004) Solidpseudopapillary<br />

tumor <strong>of</strong> the pancreas:<br />

two cases in male patients. Am Surg<br />

70(1):29-31<br />

10. Martin RC, Klimstra DS., Brennan MF,<br />

Conlon KC (2002) Solid<br />

pseudopapillary tumor <strong>of</strong> the pancreas:<br />

a surgical enigma Ann Surg Oncol<br />

9(1):35-40<br />

11. Meyer S, Aliani S, Graf N, Bonkh<strong>of</strong>f<br />

H, Schneideer G, Reinhard H (2002)<br />

Solid-pseudopapillary tumor <strong>of</strong> the pancreas<br />

in a 9-year-old girl. Klin Pediatr<br />

214(5):316-318<br />

12. Molino D, Perriotti P, Napoli V,<br />

Antropoli C, Gargano E, Antropoli M,<br />

D’Antonio D, Vicenzo L, Boscaino A,<br />

D’Anna L, Guidi G (2003) Solid<br />

pseudopapillary tumour <strong>of</strong> the pancreas.<br />

Report <strong>of</strong> a case. Minerva Chir<br />

58(6):815-821<br />

13. Nachulewicz P, Kalicinski P, Polnik D,<br />

Chy¿yñska A, Broniszczak, D (2002)<br />

Solid and papillary epithalial tumor <strong>of</strong><br />

the pancreas. A case report <strong>of</strong> surgically<br />

curable tumor. Surg Childh Intern 3:148-<br />

150<br />

14. Nadler EP, Novikov A, Landzberg BR,<br />

Pochapin MB, Centeno B, Fahey TJ,<br />

Spigland N (2002) The use <strong>of</strong> endoscopic<br />

ultrasund in the diagnosis <strong>of</strong> solid<br />

pseudopapillary tumors <strong>of</strong> the pancreas<br />

children. J Pediatr Surg 37(9):1370-1373<br />

15. Patel VG, Fortson JK, Weaver WL,<br />

Hammami A (2002) Solidpseudopapillary<br />

tumor <strong>of</strong> the pancreas<br />

masquerading as a pancreatic<br />

pseudocyst. Am Surg 68(7):631-632<br />

16. Pettinato G, Di Vizi D, Manivel JC,<br />

Pambuccian SE, Somma P, Insabato L<br />

(2002) Solid-pseudopapillary tumor <strong>of</strong><br />

the pancreas: a neoplasm with distinct<br />

and highly characteristic cytological<br />

features. Diagn Cytopathol 27(6):325-<br />

334<br />

17. Pezzolla F, Lorusso D, Caruso ML,<br />

Demma I (2002) Solid pseudopapillary<br />

tumor <strong>of</strong> the pancreas. Consideration <strong>of</strong><br />

two cases. Anticancer Res 22(3):1807-<br />

1812<br />

18. Pedevin J, Triau S, Mirallie E, Le<br />

Borgne J (2003) Solid-pseudopapillary<br />

tumor <strong>of</strong> the pancreas: a study <strong>of</strong> five<br />

cases, and review <strong>of</strong> the literature. Ann<br />

Chir 128(8):543-548<br />

19. Potrc S, Kavalar R, Horvat M, Gadzijev<br />

EM (2003) Urgent Whipple resection for<br />

solid pseudopapillary tumor <strong>of</strong> the pancreas.<br />

J Hepatobiliary Pancreat Surg<br />

10(5):386-389<br />

20. Sheehan MK, Beck K, Pickleman J,<br />

Aranha GV (2003) Spectrum <strong>of</strong> cystic<br />

neoplasms <strong>of</strong> the pancreas and their surgical<br />

management. Arch Surg<br />

138(6):657-660


<strong>Annals</strong> <strong>of</strong> <strong>Diagnostic</strong> <strong>Paediatric</strong> <strong>Pathology</strong> 2004, 8(3-4):<br />

© Copyright by Polish <strong>Paediatric</strong> <strong>Pathology</strong> Society<br />

The invited lecture<br />

Liver tumors <strong>of</strong> childhood - pathology. Report <strong>of</strong> the Kiel Pediatric<br />

Tumor Registry*<br />

Dieter Harms<br />

<strong>Annals</strong> <strong>of</strong><br />

<strong>Diagnostic</strong><br />

<strong>Paediatric</strong><br />

<strong>Pathology</strong><br />

Department <strong>of</strong> Pediatric <strong>Pathology</strong>,<br />

University <strong>of</strong> Kiel,<br />

Kiel, Germany<br />

Dear Colleagues,<br />

Over the years I have collected a series <strong>of</strong> 522 primary liver<br />

tumors. Hepatoblastomas, accounting for 51.3% <strong>of</strong> the cases,<br />

are by far the most frequent liver tumors in the files <strong>of</strong> the<br />

Registry, followed by infantile hemangioendothelioma and<br />

hepatocellular carcinoma (15.5%, each). All other liver tumors,<br />

including mesenchymal hamartoma, focal nodular hyperplasia,<br />

embryonal rhabdomyosarcoma and undifferentiated<br />

sarcoma, are rare.<br />

Hepatoblastoma has the peak incidence in the first two years<br />

<strong>of</strong> life, and up to 90% <strong>of</strong> the cases occur before the age <strong>of</strong> five<br />

years. As it is well known, the wide majority <strong>of</strong><br />

hepatoblastomas present as a single mass involving in decreasing<br />

frequency the right lobe, both lobes and the left lobe. Approximately<br />

20% <strong>of</strong> the cases are multifocal. Microscopically,<br />

the more frequent epithelial hepatoblastomas (56%) and the<br />

less frequent (44%) mixed epithelial and mesenchymal<br />

hepatoblastomas have to be distinguished. With regard to the<br />

subtypes mixed epithelial/mesenchymal tumors without teratoid<br />

features are most frequent (34%), followed by pure fetal<br />

(31%), fetal/embryonal (19%) and mixed teratoid<br />

hepatoblastomas (10%). The macrotrabecular and the small<br />

cell/anaplastic hepatoblastoma variants are very rare (3%,<br />

each). Notably, a pure embryonal variant <strong>of</strong> epithelial<br />

hepatoblastoma does virtually not exist.<br />

Nevertheless, data from the Liver Tumor Study HB 94<br />

show that the histologic subtype does not influence the prognosis<br />

significantly. By contrast, significant prognostic factors<br />

are the tumor growth pattern in the liver, vascular invasion,<br />

distant metastases, surgical radicality, initial levels <strong>of</strong> AFP,<br />

and the response to chemotherapy. The overall survival rate<br />

<strong>of</strong> patients with hepatoblastoma in different multicenter studies<br />

is approximately 75%.The prognostically much more unfavorable<br />

hepatocellular carcinoma (HCC) is significantly less<br />

frequent than hepatoblastoma except in countries with high<br />

hepatitis B virus infection rates. In the files <strong>of</strong> the Kiel Pediatric<br />

Tumor Registry 81 cases <strong>of</strong> HCC including 14 cases <strong>of</strong> the<br />

fibrolamellar variant are collected. The proportion <strong>of</strong> HCC to<br />

hepatoblastoma is 0,3:1. Most cases <strong>of</strong> HCC did occur clini-<br />

cally in the second decade <strong>of</strong> life, this in contrast to the wide<br />

majority <strong>of</strong> hepatoblastomas. Microscopically, pediatric HCC<br />

are similar to HCC <strong>of</strong> the adults. Usually the tumor cells are<br />

larger than the surrounding normal hepatocytes. The tumor<br />

cells can be arranged in a pseudoglandular, trabecular, or<br />

pseudoacinar pattern. In most cases the cytoplasm shows an<br />

intensive eosinophilia. Rarely HCC displays tumor cells with<br />

clear cytoplasm similar to clear-cell renal carcinoma and to<br />

some fetal-epithelial hepatoblastomas. Mitotic activity and<br />

nuclear atypia are variable from case to case and may be variable<br />

even in the same tumor. In this case at least moderate<br />

atypia and some mitotic figures can be seen.<br />

Immunohistochemically, tumor cells express AFP, and vessel<br />

invasions are very common. Since many HCC are hepatitis B<br />

virus (HBV)-associated, it is important to look for features <strong>of</strong><br />

preceding HBV infections.The fibrolamellar variant <strong>of</strong> HCC<br />

is not associated with virus infections and, moreover, is not<br />

associated with other conditions like metabolic diseases or<br />

biliary atresia, which are well-known risk factors for the development<br />

<strong>of</strong> classic liver cell carcinoma. In our files 17% <strong>of</strong><br />

the liver cell carcinomas belong to the fibrolamellar variant.<br />

Fibrolamellar carcinoma occurs predominantly in older children,<br />

adolescents and young adults and usually presents as a<br />

solitary, well-circumscribed tumor mass with a predilection<br />

to the left lobe. Microscopically, cords <strong>of</strong> relatively large tumor<br />

cells are separated by paucicellular collagen bands. The<br />

cytoplasm is intensively eosinophilic, and frequently contains<br />

lumina. The proliferation activity is comparably low indicating<br />

slow tumor growth. Late tumor relapses can occur, and in<br />

so far the final prognosis <strong>of</strong> fibrolamellar carcinoma is not so<br />

favorable as was supposed initially.<br />

The most important mesenchymal tumors <strong>of</strong> the liver<br />

are infantile hemangioendothelioma, mesenchymal hamartoma,<br />

embryonal rhabdomyosarcoma, and undifferentiated<br />

(embryonal) sarcoma with 15.5%, 4.8%, 5.6%, and 2.9%, respectively,<br />

<strong>of</strong> the cases collected in our Registry. With regard<br />

to the age at clinical manifestation, significant differences<br />

between these tumors become overt. Thus, most cases <strong>of</strong> infantile<br />

hemangioendothelioma are diagnosed in very young<br />

children. Referring to our publication on tumors <strong>of</strong> the newborn<br />

and the very young infant [3], 86% <strong>of</strong> the<br />

* The lecture was presented during the Meeting <strong>of</strong> the Polish Pediatric Group for the Solid Tumors’ Treatment (Liver Tumors),<br />

22-24.04.2004, Gdansk, Poland


126<br />

hemangioendotheliomas occured in children below the age <strong>of</strong><br />

6 months. For comparison, 9 hepatoblastomas have manifested<br />

in this age class, and the proportion <strong>of</strong> hemangioendothelioma<br />

to hepatoblastoma is 2.1 to 1 in early infancy. Mesenchymal<br />

hamartoma can be present at birth, like hemangioendothelioma.<br />

The wide majority <strong>of</strong> mesenchymal hamartomas occurs<br />

in patients under the age <strong>of</strong> 2 years. Embryonal rhabdomyosarcoma<br />

<strong>of</strong> the liver has a broad age distribution, but<br />

the majority <strong>of</strong> rhabdomyosarcomas develop within the first<br />

5 years <strong>of</strong> life, this by contrast to the probably rhabdomyosarcoma-related<br />

undifferentiated (embryonal) sarcoma (malignant<br />

mesenchymoma <strong>of</strong> the liver in the old nomenclature),<br />

for which the peak incidence is between five and ten years <strong>of</strong><br />

age. Infantile hemangioendothelioma occurs as a solitary mass<br />

or consists <strong>of</strong> multiple tumor nodes involving large parts <strong>of</strong><br />

the liver. Under the microscope infantile hemangioendotheliomas<br />

are composed <strong>of</strong> multiple, sometimes dilated vascular<br />

channels lined by plump endothelial cells. Large or multicentric<br />

lesions can be complicated clinically by consumption<br />

coagulopathy or disseminated intravascular coagulation<br />

(Kasabach-Merritt-syndrome) or, due to intralesional vascular<br />

anastomoses, by severe congestive heart failure. Most infantile<br />

hemangioendotheliomas have to be classified according<br />

to DEHNER’s proposal [2] as type I-hemangioendotheliomas.The<br />

rare type II-hemangioendothelioma shows a more<br />

distinct papillary pattern and endothelial cells with more atypical<br />

nuclei than in type I -tumors. Type II-infantile<br />

hemangioendotheliomas are now considered to be pediatric<br />

angiosarcomas, but malignant behavior <strong>of</strong> infantile<br />

hemangioendotheliomas is generally rare. The majority <strong>of</strong> the<br />

tumors are principally benign lesions. The prognosis depends<br />

on size and distribution <strong>of</strong> the tumor throughout the liver, on<br />

the severity <strong>of</strong> complications, and the resectability, which may<br />

be impossible in multifocal disease. Mesenchymal hamartomas<br />

are generally solitary, no encapsulated and can be very<br />

large. Up to 30% <strong>of</strong> the tumors are pedunculated.Typically,<br />

mesenchymal hamartomas show cysts containing a clear fluid.<br />

Microscopically, in typical cases a so-called ductal-plate pattern<br />

with tortous bile ducts, surrounded by loose connective<br />

tissue, and at the periphery by a more dense fibrous tissue,<br />

can be seen, so that ductal-plates are reminiscent on fibrocystic<br />

disease <strong>of</strong> the female breast. Some bile ducts may show slight<br />

nuclear atypia. Fluid accumulations in the tumor tissue my<br />

cause lymphangioma-like spaces and cysts, which are lined<br />

by flattened non-endothelial, fibroblastic cells. Taking together<br />

these macroscopic and microscopic findings and with regard<br />

to the very young age <strong>of</strong> the patients the diagnosis <strong>of</strong> mesenchymal<br />

hamartoma is easy, except in small biopsies, in which<br />

it may be impossible to exclude undifferentiated sarcoma <strong>of</strong><br />

the liver. The prognosis <strong>of</strong> mesenchymal hamartomas is very<br />

favorable. Tumor relapses are rare.<br />

Embryonal rhabdomyosarcoma <strong>of</strong> the liver developes<br />

either in the extrahepatic bile ducts or intrahepatic. Occlusion<br />

<strong>of</strong> larger bile ducts induces obstruction jaundice. Microscopically,<br />

rhabdomyosarcomas <strong>of</strong> the liver are in most cases embryonal<br />

rhabdomyosarcomas. Particularly, they present as the<br />

botryoid variant <strong>of</strong> embryonal RMS exhibiting the characteristic<br />

subepithelial cambium layer. This layer consists <strong>of</strong> parallel<br />

to the surface arranged, at least moderately differentiated,<br />

intensivly desmin-positive rhabdomyoblasts. This diagnostic<br />

cambium layer the botryoid RMS usually shows small<br />

stellate-like cells arranged in a loose pattern. By contrast to<br />

botryoid rhabdomyosarcomas <strong>of</strong> other locations, the surface<br />

<strong>of</strong> the tumor proliferations can be covered by cylindrical or<br />

cuboidal epithelial cells and not by metaplastic squamous<br />

epithelial cells. Treatment should be done according to the<br />

well-established s<strong>of</strong>t-tissue sarcoma therapy protocols. The<br />

same is true for undifferentiated sarcoma <strong>of</strong> the liver.<br />

Undifferentiated (embryonal) sarcoma <strong>of</strong> the liver usually<br />

develops in children between five to ten years <strong>of</strong> life. The<br />

median age <strong>of</strong> 17 patients with undifferentiated sarcoma treated<br />

in a cooperative Italian/German study [1] was seven years.<br />

Macroscopically, this undifferentiated sarcoma (this specimen<br />

stems exceptionally from an adult patient) shows a grayishwhite<br />

to red and brown cut surface with intensive necrosis.<br />

Under the microscope the tumor consists <strong>of</strong> spindle and seellate<br />

cells arranged within a myxoid stroma. In addition to these<br />

comparably small cells some larger and pleomorphic tumor<br />

cells can be seen. Typically, the periphery <strong>of</strong> the tumor shows<br />

entrapped bile ducts with slight nuclear atypia. These bile ducts<br />

can appear with moderately dilated lumina.<br />

Immunohistochemically, the tumor cells express vimentin intermediate<br />

filaments. In addition, some tumor cells are positive<br />

for desmin too, but true rhabdomyoblasts, particularly<br />

more differentiated rhabdomyoblasts with cross striations do<br />

not occur, this in contrast to embryonal rhabdomyosarcoma.<br />

Nevertheless, the histologic features <strong>of</strong> undifferentiated sarcoma<br />

and embryonal rhabdomyosarcoma may overlap. However,<br />

basically, the undifferentiated sarcoma is a more primitive<br />

sarcoma than embryonal rhabdomyosarcoma. The prognosis<br />

<strong>of</strong> undifferentiated sarcoma <strong>of</strong> the liver was very poor in<br />

the past (mortality about 80 % - STOCKER and ISHAK 1978).<br />

Introduction <strong>of</strong> multimodal s<strong>of</strong>t-tissue sarcoma therapy strategies<br />

has improved the prognosis dramatically. According to<br />

data <strong>of</strong> the forementioned Italin/German study 12 from 17<br />

patients (approximately 70 % <strong>of</strong> the cases) have survived with<br />

follow-up ranging from 2,4 to 20 years [1].<br />

Liver tumor pathology covers a wide variety <strong>of</strong> malignant<br />

and benign, epithelial and mesenchymal tumor entities.<br />

The wide majority <strong>of</strong> principally epithelial tumors<br />

(hepatoblastomas and liver cell carcinomas) are malignant,<br />

whereas the majority <strong>of</strong> mesenchymal tumors (except undifferentiated<br />

sarcoma and embryonal rhabdomyosarcoma) are<br />

morphologically benign. The prognosis <strong>of</strong> malignant liver tumors<br />

has improved dramatically during the recent few decades.<br />

Improvement <strong>of</strong> prognosis is due to interdisciplinary cooperation<br />

in multimodal therapy studies. Another milestone was<br />

the development <strong>of</strong> new pediatric surgery strategies and techniques,<br />

which have contributed significantly to the prognostical<br />

success in malignant and in surgically problematic benign tumors<br />

<strong>of</strong> the liver too.


1. Bisogno G, Pilz T, Perilongo G (2002) Undifferentiated<br />

sarcoma <strong>of</strong> the liver in childhood: a curable disease. Cancer<br />

94(1):252-257<br />

2. Dehner LP (1978) Hepatic tumors in the pediatric age group:<br />

a distinctive clinicopathologic spectrum. Perspect Pediatr<br />

Pathol 4:217-268<br />

3. Harms D, Schmidt D, Leuschner I (1989) Abdominal, retroperitoneal<br />

and sacrococcygeal tumours <strong>of</strong> the newborn and<br />

the very young infant. Report from the Kiel <strong>Paediatric</strong> Tumour<br />

Registry. Eur J Pediatr 148(8):720-728<br />

127

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

Saved successfully!

Ooh no, something went wrong!