• No results found

Novel applications of growth factors in solid tumors - 2: Growth factors in human ovarian cancer

N/A
N/A
Protected

Academic year: 2021

Share "Novel applications of growth factors in solid tumors - 2: Growth factors in human ovarian cancer"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Novel applications of growth factors in solid tumors

Westermann, A.

Publication date

1999

Link to publication

Citation for published version (APA):

Westermann, A. (1999). Novel applications of growth factors in solid tumors.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)

and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

Anneke M. Westermann

1

, Jos H. Beijnen

1

, Wouter H. Moolenaar

2

,

Sjoerd Rodenhuis

1

From

1

the Department of Medical Oncology and

2

the Division of

Cellular Biochemistry, The Netherlands Cancer Institute,

Amsterdam, The Netherlands.

n

• o

CD

Growth factors in human ovarian NJ

cancer

(3)

Abstract

Background Ovarian cancer remains confined to the peritoneal cavity, even in the late stages of the disease. This particular growth pattern might be the result of the activity of growth factors present in malignant ascites, a common feature of advanced disease. However, presently identified growth factors cannot fully account for the mechanics of ovarian cancer.

Methods Papers on growth of ovarian cancer cell lines in vitro and the clonogenic assay of human ovarian cancers, and the factors influencing them are summarized. Presently known peptide growth factors were reviewed for their association with human ovarian cancer, as were cytokines. The information on the growth-promoting properties of ascites was surveyed, and the available evidence for possible novel growth factors was collected.

Results Growth of ovarian cancer cells, both in cell lines and in the clonogenic assay, is stimulated by peptide growth factors, although no combination of peptide growth factors can replace serum or cell free ascites. The growth-promoting characteristics of ascites seem to be completely explained by the presence of lysophosphatidic acid (LPA), a bioactive phospholipid with mitogenic features.

Conclusion The bioactive lipid LPA seems to be an important growth promoter present in ascites of ovarian cancer patients. LPA inhibition may be a novel target for therapy.

(4)

Introduction

Human ovarian cancer is the leading cause of death from gynecologic malignancy. In most cases, the initial diagnosis is established at an advanced stage, when current therapy can only cure a small proportion of patients. Even in the presence of advanced disease, there is rarely evidence of tumor cells outside the peritoneal cavity.1 Local growth factors may play a part in

promoting intraperitoneal spread and growth, and it is conceivable that these could represent unique targets for therapy.

Although many studies suggest that malignant ascites may contain one or more specific growth factors required for the proliferation of ovarian cancer cells, so far no specific ascites-borne growth factors have been isolated and identified that are candidates for such a role. A group of bioactive lipids, the lysophosphatidic acids, are present in both serum and in malignant effusions.2 It is possible that the elusive ascitic growth factor may belong to this novel group of

established mitogens. The present review summarizes current insights into the function of well-characterized growth factors in ovarian cancer growth, and discusses the potential influence of the abovementioned ascites-borne lipid growth promoters.

Growth of ovarian carcinoma cell lines in vitro

A large number of ovarian cancer cell lines have been described since the early seventies.323

Media used to maintain cell lines were routinely supplemented with antibiotics, vitamins and fetal calf serum (FCS) or horse serum. Although insulin, transferrin, gonadotropins, estradiol, dexamethasone, L-thyroxin or epidermal growth factor (EGF) were added in some instances, human ovarian cancer cell lines almost universally required the addition of serum. In Table 1, a selection of human ovarian carcinoma cell lines with their requirements for growth in vitro is listed.

Establishment of ovarian cancer cell lines in serum-free medium is apparently difficult. In the early 1980s, however, investigators succeeded in growing rat ovarian cancer cells in serum-free medium by adding EGF and transferrin.24 The serum requirement to sustain human ovarian

cancer cell lines could be reduced from 20 down to 0.5% when suitable growth factors were employed (table 1). In the complete absence of serum, serum components such as fetuin or bovine serum albumin were usually necessary to maintain the cell lines.10'2022'23 Rare exceptions

to this rule include certain subcultures of established ovarian cancer cell lines, as reported by Jozan ef a/.22 and Golombick etal.25 The latter described a cell line grown in a solution of amino acids, vitamins and simple salts. Berchuck etal. could sustain growth of ovarian cancer cell lines in serum-free medium for 48 hours, but not in permanent culture.26

It has been suggested that cell lines requiring little or no serum for in vitro growth are less well differentated than those that do not grow in chemically defined media,10 but this has not

(5)

Table 1. Reports of permanent human ovarian cancer cell lines in vitro.

Cell line Supplements References

154, 163 20% fetal calf serum (FCS), antibiotics DiSaia 3

#2774 20% FCS, antibiotics Freedman 4

163 20% FCS, antibiotics Kanabus 5

HeW 20% FCS Pattillo 6

A7, A10 10% FCS, antibiotics Abu Sinna 7

COLO 110, 316, 319, 330 10-20% FCS, antibiotics Woods 8

NIH:OVCAR-3 2% FCS, insulin 10 lU/ml, antibiotics Hamilton 9

EFO-3, EFO-21, EFO-27, 10% FCS, fetuin 5 |ig/ml, transferrin 2.5 ng/ml, insulin Simon 10 EFO-47, EFM-63 0.08 lU/ml, human menopausal gonadotropin

0.04 lU/ml, L-thyroxine 10 nM, antibiotics

EFO-21, EFO-47 fetuin 5 u.g/ml, 0 . 1 % bovine serum albumin, transferrin 2.5 ng/ml, insulin 0.08 lU/ml, L-thyroxine 10 nM, antibiotics

OAW42 10% FCS Wilson 11

CAOV-3, SKOV-3, HOC-1, 10-15 % FCS Buick 12

HOC-7, HEY

la 288 10% horse serum, 10% FCS, 1 \i/\ transferrin, T3 6.5 ng/ml, dexamethasone 14 ng/ml, 108M

estradiol, insulin 20 |ig/ml, antibiotics

Clamon 13

IGROV1 10% FCS Benard 14

JA-1.TR175, TR170 10% FCS Hill 15

HTOA 15% FCS Ishiwata 16

PE01, PE04, PE06, PE014, 10% FCS Langdon 17

PE016, PE023, PEA1, PEA2, T014

DO-s 2-15% FCS Briers 18

A1, A69, A90, A121 20% FCS Crickard 19

UWOV1, UWOV2 2-5% FCS, antibiotics Golombick 20

UWOV2 (Sf) insulin 1 mg/l, transferrin 1 mg/l, bovine serum albumin 500 mg/l

OMC-3 10% FCS Yamada 21

OVCCR1/sf insulin, transferrin, epidermal growth factor (EGF) in varying concentrations3

Jozan 22 SCHM-1, RIC-2, MAC-2, 3% FCS, antibiotics, EGF 5 ng/ml, insulin 5 ng/ml,

SIB-1 transferrin 10 u,g/ml

MAC-2, SIB-1 Bovine serum albumin 3 mg/ml, antibiotics, EGF 5 ng/ml, insulin 5 ng/ml, transferrin 10 ng/ml

Hirte 23

UWOV2 (Pf) vitamins Golombick 25

a0.5% serum was required for each passage to allow cell adhesion to the plastic.

been confirmed in later publications.23

The scarcity of serum-free cell lines strongly suggests the presence of as yet unknown growth promoting factors in serum. A large number of potential candidates for this serum-derived growth factor have been proposed and will be discussed below.

(6)

Clonogenic assay of human ovarian carcinoma cells

In the late 1970s, interest arose in the isolation of human tumor cells directly from patients for drug sensitivity testing and other studies. Successful direct cultures of human cancer cells were rarely described27 until Hamburger and Salmon developed a clonogenic assay providing

conditions for cell growth of most tumor types.28 In their original bioassay, tumor cells were

cultured in a two-layer system, with the upper layer containing tumor cells suspended in agar supplemented with horse serum, antibiotics, glutamin, CaCI2, insulin, asparagine, dextran and

2-mercaptoethanol. The lower agar layer contained medium conditioned by adherent spleen cells of mineral oil-primed BALB/c mice. The majority (85 to 100%) of cells obtained from either biopsies or effusions of ovarian cancer patients could form colonies using this method.29 32 Selected reports on conditions for the clonogenic assay of human ovarian cancer cells are

summarized in Table 2.

It later turned out that feeder layers were not the only method to allow clonogenic in vitro growth, although it proved impossible to clone ovarian cancer cells in serum-free conditions with the addition of wellknown growth factors alone.33 For in vitro growth, ovarian cancer cells

may require a specific and individual set of growth factors, as has been suggested for other tumors grown in serum-free conditions, such as small cell lung cancer.34

Peptide growth factors

Peptide growth factors play a pivotal role in the control of proliferation of normal and tumor cells.35 They exert their action through binding of a specific cell surface receptor after secretion

into the extracellular environment. The capacity of tumor cells to produce and react to their own growth factors via functional external receptors is a central principle that has been termed 'autocrine secretion' or 'autocrine loop'. This paradigm includes the notion that growth factors can cause malignant transformation through both overproduction of positive autocrine growth factors and the loss of negative growth control present in normal cell growth.36

The role of peptide growth factors in human ovarian cancer cell lines and in direct cultures is summarized in Table 3.

Epidermal growth factor family

Human epidermal growth factor (EGF) was among the first polypeptide growth factors isolated.37

It stimulates growth of normal human epithelial cells in culture as well as clonogenic growth of human tumor cell lines and freshly isolated tumor material, although inhibition of some cell lines

(7)

EGF-Table 2. , Reports on short term culture of human ovarian carcinoma from bi opsies or effusions.

Mat.a Medium Supplements Growth col/5x10 5 cells Ref.

T/E 78 Eagle's modified essential medium (MEM) E 8 two-layer system (TLS) of

0.3% agar in CMRL 1066, on medium conditioned by adherent spleen cells of mineral oil-primed BALB/c mice T/E 31 -TLS with medium conditioned

by adherent spleen cells of mineral oil-primed BALB/c mice -TLS with type-0 human

red blood cells

-TLS with medium conditioned by CD-1 or DBA/2 or non-oil primed BALB/c mice -TLS with medium conditioned

by MA-184, Wi-38 cell lines -TLS with cell-free autologous effusion 1:4

E 27 standard TLS T 51 enriched Eagle's MEM

E 13

T6 enriched Eagle's MEM in 0.3% agar

15%FCS, 10% human cord serum

15% PCS, 20% horse serum, antibiotics, 2-ME

15% horse serum, antibiotics, +/- 2-ME

10%FCS, 10% human cord serum, antibiotics, irradiated xenogeneic feeder layer of adherent peritoneal cells

10%FCS, 10% human cord serum, antbiotics

10%FCS, 10% human cord serum, antibiotics, irradiated xenogeneic feeder layer of adherent peritoneal cells 10% PCS, 10% human cord serum, antbiotics

69% 100% 10% PCS, 15% horse serum 81 % 4 1 % 3 1 % 100-800 85% 99-2549 320-833 109-320 0 29 20-620 100% 299(26-1464) 100% 507(50-2528) 27 28 29,30 32 31 T/E 16 T35 standard TLS 15% FCS, 20% horse 53% standard TLS, with 0-75% enriched CMRL 1066 in top layer serum 15% FCS, 20% horse serum, substitution of CMRL with 25-100% cell free ascites 71%

standard TLS 2.5-15% horse serumb

EGF 50 ng/ml, insulin 5 p.g/ml, transferrin 5 u,g/ml, selenium 5 ng/ml, 17B-estradiol W M , hydrocortisone lO^M 66% 0 113 33

(8)

Table 3. Overview of growth factors in ovarian cancer

Substance Effect on ovarian

cancer cell growth

References

Epidermal growth factor (EGF)

Amphiregulin (AR)

Transforming growth factor ß (TGF-ß) Platelet derived growth factor (PDGF) Basic fibroblast growth factor (bFGF) Insulin-like growth factors (IGF) Interleukin 1 (IL-1)

Interleukin 6 (IL-6)

Tumor necrosis factor a (TNF-oc)

Macrophage-colony stimulating factor (M-CSF) Cell free ascites (CFA)

Lysophosphatidic acid (LPA)

dual 26,39

stimulating 22,38,40,41 (cell lines),33,43-46 (direct cultures)

dual 65

inhibiting 39,42,70-72 (cell lines),73 (direct cultures) no effect 26 stimulating 19 stimulating 85 stimulating 93 no effect 95 stimulating 93 no effect 97 stimulating 11,38,58,116,117 (celllines),29,30 105, 113,118 (direct cultures) stimulating 128, Jalink 1995 (unpublished

observations)

receptor commonly present on many different cell types, including ovarian cancer cells. This general g r o w t h stimulating effect has been reported in most ovarian cancer cell lines26-3842 and

also in direct clonogenic assays of ovarian carcinoma.33'43-46 Some cell lines, however, showed no

response to EGF,26 or were inhibited by EGF. Varying responses of cell lines to peptide g r o w t h

factors are common. They are not usually dependent on concentration, but they are possibly due to different post-receptor events in the various cell lines.39

EGF was later shown to be part of a family of structurally homologous peptides also including transforming growth factor a (TGF-a)47 and amphiregulin (AR).48 The members of this family bind

to the EGF-receptor (EGFR), causing receptors to dimerize and initiate intracellular signalling. The EGFR has marked homology with the human c-erbB-2 gene (which is identical to HER-2 or the rat neu oncogene) product, a 185 kDa protein (p185e*B2) with tyrosine kinase activity.49 Although

TGF-a and EGF do not bind to p185e*B2, the not yet fully characterized 30 kDa glycoprotein ligand

of the erbB-2 oncogene product has been shown to interact directly with the EGFR.50 Other members

of this receptor family, c-enbB-3 and c-erbB-4, have recently been identified, and the dose similarity in structure of the four receptors allows extensive heterodimerization of these different receptor

a T denotes solid tumor, E effusion, and T/E both as a source of malignant cells, followed by the

number of patients tested.

b The serum requirement could be reduced by the growth factors and hormones specified in the

serum-free experiments.

(9)

subtypes.51-52 Immunohistochemically, increased expression of these receptor proteins has been

demonstrated in malignant ovarian tumors compared to benign ones.53

TGF-a differs from EGF in that it can induce anchorage-independent growth not only in cancer cells but also in untransformed, non-neoplastic, indicator cells.54 TGF-a promotes ovarian cancer

cell line growth in vitro and enhances direct clonogenic growth of ovarian tumor cells, thereby mimicking EGF,3M1 while the amphiregulin effects are similar but less pronounced.48

Evidence to support the autocrine loop concept in ovarian cancer was presented by Bauknecht

et al., who documented elevated levels of EGF and of EGF-like factors in crude cellular extracts

of ovarian tumors compared to non-malignant tissue.55 In contrast, neither Berchuck etal.26 nor Ottensmeier et a I.56 found EGF activity in ovarian cancer cell-conditioned media. Production of EGF and TGF-a by ovarian cancer cell lines was, however, demonstrated by Kurachi etal.40 and Stromberg et al.57 Hanauske et al. showed a higher frequency and level of TGF-a in malignant effusions in a variety of tumors including ovarian cancer, as compared to benign effusions.58

Further support for a TGF-a/EGFR autocrine loop in ovarian cancer was provided by experiments with primary tumor cultures in which TGF-a but not EGF was detected in EGFR-positive ovarian tumors.59 Gordon etal. reported mRNA expression of EGF, TGF-a, AR, EGFR and erbB-2 in three

ovarian cancer cell lines, both in serum-supplemented and serum-reduced conditions.60 Other

authors found that in the presence of exogenous EGF the amount of serum needed to sustain in

vitro proliferation of cell lines42 and direct clonogenic assay33 could be lowered, though it could

not compensate completely for the serum requirement.

EGFR-positive ovarian cancer cells appear indeed to require the presence of a growth stimulating EGFR ligand for growth in vivo, as shown by experiments in nude mice who were EGF depleted by sialoadenectomy. Subcutaneous injection of EGFR-positive ovarian cancer cells led to tumors in animals receiving exogenous EGF, but not in those not receiving the growth factor.61

In ovarian cancer cell lines overexpression of erbB-2 is frequently encountered.64 erbB-2

expression can be detected by immunohistochemistry in 30 to 70% of human ovarian cancers,62'63

but in only 5 to 10% of normal ovarian cells.64

Both normal and ovarian cancer cells express and respond to amphiregulin. At low concentrations (picomolar range), amphiregulin inhibits growth of certain normal and malignant ovarian cells. At higher concentrations (nanomolar range) growth stimulation can be observed. These effects may vary between cell lines,65 as described above for EGF effects.

Transforming growth factor ß

The hallmark of transforming growth factors (TGF) is their ability to confer a transformed phenotype on untransformed indicator cells. In the early 1980s the TGFs were found to include two groups: TGF-a mentioned previously, which is structurally and functionally homologous

(10)

with EGF, and TGF-ß.66 TGF-ß does not bind to the EGFR, but a synergistic effect was observed

with EGF orTGF-a, leading to a potent transforming action on untransformed indicator cells.67

Later research indicated that the predominant effect of TGF-ß on normal cells in vitro is inhibition of proliferation. This dual effect makes TGF-ß a central mediator of cell growth regulation. TGF-ß is considered a principal negative control element in cell growth, and it is present in most body fluids and tissues.68 Loss of responsiveness to the growth inhibitory effects of TGF-ß may be a

general characteristic of malignant transformation. The TGF-ß family can be divided into at least three different subtypes in humans with similar in vitro functions and with multiple receptor subtypes.69

Studies on the effects of TGF-ß on ovarian tissue showed inhibition of growth of normal epithelial ovarian cells,70 ovarian carcinoma cell lines,39'427072 and directly cultured ovarian cancer cells,73

However, some cell lines in these studies showed no response to exogenous TGF-ß at all.26'70 or

only to one or two particular subtypes of TGF-ß.72

Production of TGF-ß was demonstrated in normal ovarian tissues,7074 ovarian cancer cell

lines,4270-7275 and directly cultured ovarian cancer cells.73-74 Though some tumors seemed to

have lost the capacity to produce TGF-ß,73 others were found to have increased expression of

certain TGF-ß isoforms.74 TGF-ß is secreted by most cells in a biologically inactive form and

regulation of bioactivation may be of physiologic importance. As a result, immunohistochemical demonstration of the peptide in tumors or cells may not directly reflect its biological significance.

Platelet derived growth factor

Platelet derived growth factor (PDGF) is a polypeptide growth factor first described in the 1980s as a megakaryocyte-derived mitogen for mesenchymal cells, but later it was also shown to be synthesized by a plethora of other normal and transformed cells. It stimulates both epithelial and endothelial cells in an autocrine fashion.76 The growth factor has three isoforms and binds

to two defined receptors (termed PDGFRa and PDGFRß).77

Normal ovarian tissue or benign ovarian tumors do not show PDGF expression, although PDGF-expression is common (70-100%) in ovarian cancer tissue and in cell lines,78 as detected

by immunohistochemistry. PDGFRa is less frequently expressed than PDGF in human tumors (0-35%).79 PDGF was found to have an autocrine role in a cell line growing in a defined

serum-free medium.75 Berchuck et al. found no stimulation of ovarian cancer cell lines by exogenous

PDGF,26 consistent with the lack of expression of PDGF receptors in other studies. It has therefore

been suggested that PDGF acts in a paracrine fashion in ovarian tumors and mediates supportive connective tissue stroma formation.71

(11)

Basic fibroblast growth factor

Basic fibroblast growth factor (bFGF) is part of a family of heparin-binding growth factors for many mesodermal and ectodermal cells, though originally described as a mitogen for cultured fibroblasts. The family consists of at least seven distinct proteins and four receptors. (For review see 80 ,81)

bFGF as well as the FGFR1 receptor subtype have been detected in ovarian cancer cell lines, while exogenous bFGF stimulated ovarian cancer cell line proliferation.82 An earlier study indicated

growth stimulation by FGF of only one of four ovarian carcinoma cell lines.26

Insulin-like growth factors

The insulin-like growth factors (IGF) IGF-1 and IGF-2 are polypeptides with endocrine, paracrine and autocrine effects on cell proliferation. Their mitogenic effects in vitro are suggestive of a role in growth regulation and even in the oncogenesis of malignant tumors. The IGFs are present in body fluids, bound to specific binding proteins that not only carry IGF, but also regulate access to the IGF receptors, thereby mitigating IGF's mitogenic action, (reviewed in 83) Both IGF-1, its binding proteins and its receptor are expressed in ovarian cancer cell lines and ovarian tumors.84 In a small study, Karasik et al. found that levels of IGF-1 and one of its binding

proteins could discriminate between malignant and benign ovarian tumors.85 IGF-1 stimulated

the growth of two ovarian cancer cell lines, whereas specific IGF-1 receptor RNA anti-sense oligodeoxynucleotides markedly stilted the growth of those cell lines. This hints at a role in the autocrine growth of ovarian cancer cell lines for IGF and its receptor,86 though more extensive

data are required to corroborate these findings.

Cytokines

Cytokines are peptide molecules initially thought to modulate the proliferation and bioactivity of cells of the immune system. It is presently clear that cytokines are peptide factors that mediate a host of different reactions in different cell populations and in the extracellular environment, and the distinction between cytokines and other peptide growth regulatory factors has lost most of its significance. The cytokine family was originally divided into interleukins (thought to be produced by leukocytes), lymphokines (originating from lymphocytes), monokines (released by monocytes), interferons (IFN) and colony-stimulating factors (CSF). These names stem from historical concepts about function and origin of these proteins, that are largely structurally unrelated, (reviewed in 87)

(12)

type, dose and circumstances.88 A large body of evidence has been accumulated that cytokines

are produced by ovarian tumors, but growth modulatory effects of cytokines have not been decisively documented, (for a review of the role of cytokines in ovarian cancer see ref 89).

Tumor necrosis factor

TNF was one of the first cytokines to be identified.90 Although the subtypes TNF-a and TNF-ß

have a similar scale of biological activities, TNF-ß effects are generally weaker, and TNF has come to mean TNF-a unless specified otherwise in most publications. Both TNF and its encoding mRNA have been detected in uncultured ovarian cancer specimens,9193 although Wu eta/.found

no expression in ovarian cancer cell lines in vitro.93 Exogenous TNF as well as interleukin 1 (IL-1) could reinduce TNF expression in a clonogenic assay of ovarian cancer cells that had lost this ability after 7 days in culture.93

Interleukin 1

IL-1 is a cytokine produced primarily by monocytes and macrophages, with both inhibitory and stimulatory effects on human tumor cells. Two subtypes, IL-1 a and IL-1 ß, can be discerned, that bind to the same receptors. Promotion of tumor metastases by IL-1 has been described for some tumors,94 and in a proportion of ovarian tumors IL-1 can be detected.89 IL-1 can induce

TNF expression in a primary culture of ovarian cancer cells, as described above.93

Interleukin 6

II-6 is a pleiotropic cytokine with both growth- and differentiation-inducing activity, that can be produced by T lymphocytes, monocyte/macrophages, and a variety of tumor cells. It is also secreted by ovarian cancer cell lines and by ovarian tumor cells in the clonogenic assay, and it can be detected in malignant ascites.95 IL-6 mRNA expression has furthermore been demonstrated

in normal ovarian epithelial cells.96 However, exogenous IL-6 did not stimulate proliferation of

six different ovarian cancer cell lines.95

Culture supernatants from human monocytes contain significant amounts of IL-6, TNF-a and IL-1, and stimulate clonogenic growth of ovarian cancer cell lines in vitro.97 These growth factors could, however, not completely account for the growth enhancement observed when ovarian cancer cells were grown in peripheral blood monocyte-conditioned medium,97 suggesting that

as yet unidentified monocyte-derived factors also play a part.

Macrophage colony stimulating factor

Macrophage-colony stimulating factor (M-CSF, also called CSF-1) enhances monocytopoiesis, attracts macrophages and can stimulate production of TNF-a. In ovarian cancer cell lines and in

(13)

ovarian cancer in vivo, expression of M-CSF,98100 and its receptor101J02 could be demonstrated,

while normal ovarian epithelial cells do not express M-CSF. Addition of either one of the colony stimulating factors M-CSF, G-CSF or GM-CSF did not influence growth of ovarian tumor cell lines,97 although this has been reported for other CSF-receptor positive cancer cell lines.103

Cytokines in ascites

Increased levels of CSF,104'105 TNF-a,100'106<107 |L-1 ß95-100-106 and |L-6 95-100-l0S-108 have been detected

in effusions of ovarian cancer patients,100 while benign ovarian follicular fluid contained much

lower levels, especially of IL-6 and ll_-1ß.106 It has been suggested that high CSF levels in ascites

correlate with a poor prognosis.105 The cellular source of these cytokines is uncertain. Malignant

effusions are known to contain large numbers of macrophages. An increased number of cytokine-producing macrophages found at ovarian cancer sites may account for some of the biological characteristics of the tumor.

In conclusion, there is strong evidence that the peptide growth factor/cytokine network is inappropriately functioning in human ovarian cancer, and that this imbalance contributes to ovarian cancer cell growth. The growth promoting characteristics of serum can not, however, be fully substituted for or explained by the activities of the known peptide growth factors and cytokines.

Growth promoting properties of malignant effusions

As in many other solid tumors, no single group of growth factors or cytokines has been shown to be unequivocally responsible for ovarian tumor growth. Research into possibly specific growth substances has enhanced interest in novel growth substances present in ascites. In addition, cell lines grew more rapidly during establishment in the presence of cell-free ascites (CFA) compared to serum,17 suggesting the presence of adequate levels of one or more critical

growth promoters in ascites.

Macrophages

For clonogenic growth of ovarian cancer cells obtained from effusions, feeder layers were not required in the clonogenic assay. When tumor cells freshly obtained from patients were kept suspended in their native ascites, colony growth could be observed until 24 hours after collection.32

This was ascribed to the abundant macrophages present in malignant effusions, providing a feeder layer effect. This hypothesis was supported by the fact that removal of macrophages from cell populations in malignant effusions lowered the average number of colonies in the original clonogenic assay by Hamburger ef a/.2930 Buick ef a/, showed that this decrease in

(14)

plating efficiency was reversible in reconstitution experiments that used the removed autologous adherent cells as an underlayer in a two-layer agar system.109 Even xenogeneic peritoneal

macrophages (obtained from mice) were able to improve the cloning efficiency of primary ovarian cancer cells in culture.31

Mantovani etal. found an array of effects of macrophages isolated from human ascitic ovarian tumors on in vitro tumor growth of established murine and human cell lines, with both cytotoxic, cytostatic and stimulatory effects observed in different experiments.110

These data suggest that adherent, phagocytic cells - possibly macrophages - may release or produce one or more factors vital to clonogenic growth of human tumors.111

Cell-free ascites

Vaage and Agarwal have reported growth stimulation of several murine neoplastic cells not only by serum but even more so by cell-free ascites (CFA).112 Uitendaal etal. also demonstrated

the growth promoting properties of cell-free ascites on ovarian tumors in the clonogenic assay.113

Replacement of the medium supplements (including FCS) by CFA from ovarian cancer patients resulted in increased plating efficiency in the clonogenic assay of ovarian tumor specimens, and this effect was maintained if ascites of other ovarian cancer patients was used. In further experiments by Broxterman et al.,38 CFA consistently enhanced clonogenic growth of ovarian cancer cell lines more efficiently than a range of sera.

When Hamburger and Salmon first added CFA to the culture medium in the clonogenic assay, they found decreased plating efficiency as compared to BALB/c spleen cell-conditioned media.30 In

addition to growth-stimulating factors, ascites may thus also contain growth inhibiting molecules. The presence of growth inhibiting factors in ascites that co-purified with TGF-a was shown in experiments with rat ascites fluids that suppressed the growth of human colon carcinoma cell lines CBS and MOSER.114 The growth of other cell lines, HCT 116 and HCT C was not inhibited.

These observations are suggestive of a balance between growth promoting and inhibitory factors present in ascites, with a net effect of growth promotion in malignant ascites consistent with the familiar autocrine concept of the role of growth factors in tumorigenesis. The ascitic growth factors have not yet been fully characterized, and their cellular source remains undefined, with both peritoneal macrophages and tumor cells being possible candidates.

In a study of Wilson etal., a comparison was made between the growth promoting properties of ascitic fluids, cyst fluids and peritoneal washings from patients with benign or malignant gynecological conditions. Effects on a variety of tumor- and normal cell lines were studied. All fluids stimulated colony formation in soft agar, and this activity was not tumor or cancer-related. The 'colony stimulating activity' could be inhibited by the addition of heparin and thrombin.115

(15)

Evidence for a non-peptide growth factor

Mills et al. provided evidence for the presence of a novel growth factor in ascites. They reported that ascitic fluids from ovarian cancer patients but not from patients with other cancers or benign diseases induced proliferation of fresh ovarian cancer cells and of the ovarian cancer cell line HEY. The proliferative response was associated with rapid increases in phospholipid hydrolysis and free cytosolic calcium. None of a number of well characterized growth regulators such as EGF, PDGF, FGF, thrombin, vasopressin, angiotensin, IL-1, IL-2, interferons, TGF-ß, or TNF-oc, increased intracellular calcium concentrations. Pretreatment of the tested cell lines with these factors did not in any way alter the response to CFA, suggesting that CFA contains hitherto unknown growth promoters that stimulate cells through phospholipid hydrolysis. CFA could completely substitute for human plasma or FCS in these experiments. "6 Subsequent experiments

showed that the human ovarian cancer cell line HEY could grow intraperitoneal^ in immunodeficient nude mice in the presence, but not in the absence of ascitic fluid from ovarian cancer patients. Ascitic fluid from patients with benign disease could not support this growth. After the intraperitoneal tumors progressed, CFA was no longer needed for tumor growth. Eventually the xenografted tumors developed ascites with potent growth factor activity, suggesting an autocrine mechanism.117

These data suggest that CFA contains a growth promoting substance that is different from known polypeptide mediators. Xu and Mills termed the putative growth factor present in ascites from ovarian cancer patients OCAF (ovarian cancer ascites factor) and postulated that it might be a lipid, based on the observations that the growth promoting fraction in ascites is resistant to all known proteases, and stable under such drastic conditions as boiling, pH extremes and detergent treatment that should destroy proteins. It was furthermore soluble in 80% acetone and 100% methanol, though it could be purified by techniques used in protein chemistry such as reverse phase chromatography and isoelectric focusing, probably due to a tight association with albumin or other serum proteins.118 In their analysis of extracted purified OCAF, OCAF

appeared to be a phospholipid, with biochemical characteristics identical to lysophosphatidic acid (LPA), an established bioactive phospholipid with growth promoting properties.

Phospholipid growth factors - lysophosphatidic acid (LPA)

Since the 1980s, it has become increasingly clear that phospholipids do not only have a structural function in biological systems, but are also active in intracellular signalling and can act as growth factors. The best known of these is platelet-activating factor (PAF), a phospholipid that induces a rapid increase in intracellular calcium levels after binding to a high affinity

(16)

OH receptor.119120 It has a role in hypersensitivity and inflammatory

Q _ p Q u responses such as asthma and septic shock, but has so far not been ! implicated in cancer growth nor in the control of normal cell y proliferation.121

Q Q Q Lysophosphatidic acid (LPA; monoacylglycerol-3-phosphate) (See

I | fig. 1 for structure) is the simplest naturally occurring glycero-ls phospholipid, that has long been known to be a critical intermediate 0 = C in de novo phospholipid biosynthesis.122

Recently, the broad spectrum of biological activities of LPA has p

been found to include platelet activation, induction of neuronal shape

LPA

changes, smooth muscle contraction and stimulation of cell Figure 1. Structure of proliferation when added to fibroblasts and epithelial cells in lysophosphatidic acid (LPA).

culture.123 LPA acts on its cognate G-protein coupled cell surface

receptor, which is apparently present in many cell types.124

LPA is a normal constituent of serum, where it is present in albumin-bound form, but it is absent in platelet-poor plasma. Serum contains a factor that co-purifies with albumin and that causes neurite retraction in PC12 cells, inhibits the proliferation of certain tumor cells in vitro, and activates the phophatidylinositol/Ca2+ second messenger system in a variety of cells. There

is evidence that albumin-bound LPA is responsible for at least part of these activities. Synthetically prepared lysophosphatidates reproduce the biological activities of the natural serum factor.125

Kreps etal. demonstrated that serum effects on cyclic AMP accumulation in nine different cell lines were completely mimicked by LPA.126 Jalink etal. reported identical neuronal shape changes

in neuroblastoma cell lines after exposure to LPA or serum.127

LPA is formed and released by activated platelets, but this is probably not the only source of LPA, as there is evidence that LPA is also produced by growth factor stimulated fibroblasts and injured cells.128 It is of interest that earlier studies have demonstrated that platelet lysates can

enhance the growth of some, but not all epithelial and mesodermally-derived tumors in soft agar,129'130 and that a platelet-derived TGF-like compound present in serum, distinct from PDGF,

EGF or insulin can stimulate fibroblast growth in vitro.'13'

The mitogenic action of LPA on fibroblasts in culture is well-documented,132 and occurs in

the absence of synergizing peptide growth factors. The concentration of LPA required for the full mitogenic effect is micromolar as opposed to the nanomolar concentration necessary for neurite retraction and for the other responses mentioned previously. The mitogenic activity critically depends on the length of the fatty acid chain, with 1-oleoyl LPA being most potent and 1-decanoyl LPA showing hardly any mitogenic activity.133

(17)

LPA promotes invasion of hepatoma and carcinoma cells into monolayers of mesothelial cells. Serum produced a similar effect, but peptide growth factors could not substitute for serum or LPA in this study.134 Xu et al. found that LPA could stimulate proliferation of the human Jurkat T

cell line in the absence of serum.135 LPA activates breast and ovarian cancer cell lines SKOV-3

and OCC1 in completely serum-free medium,136 as well as OVCAR-3 and Lucy (Jalink K et al.

Unpublished observations.), suggesting that LPA might be the serum-derived growth factor that makes it so difficult to culture cells in serum-free media.

Since Xu ef a/, suggested that LPA might be the potent growth enhancing factor present in ascites,118 both their group and ours have shown the presence of an LPA-like mitogenic activity

in ascites of ovarian cancer patients, and found its levels to be substantially higher than those in

Conclusion

Known peptide growth factors and cytokines have a role in ovarian tumor growth, but they cannot fully account for the growth promoting properties of both serum and malignant effusions of ovarian cancer patients. We postulate that the so far elusive growth promoter might be the bioactive phospholipid LPA, based on the known properties of LPA and on the substitution experiments described above. More data are required to confirm the connection between LPA and ovarian cancer. Further studies should include quantitation of LPA-levels in malignant effusions of ovarian cancer patients compared to other patients, as well as identification of the cellular source of ascitic LPA.

Ovarian cancer remains confined to the peritoneal cavity until late in its clinical course. If LPA in ascites is indeed an important growth factor in this disorder, therapy might be targeted towards inhibiting its activity. The multifunctional growth factor inhibiting agent suramin currently is the only drug available to block LPA activity at the receptor level, but the development of more potent and more specific LPA antagonists may be a viable enterprise.

(18)

References

1. Ozols, R.F., Rubin, S.C, Dembo, A.J. & Robboy, S.J. (1992) Epithelial ovarian cancer. In: Hoskins, W.J., Perez, C.A. & Young, R.C. eds. Principles and practice of gynecologic oncology, 1 st edition. Philadelphia, PA: Lippincott, pp.732-781.

2. Moolenaar, W.H., Van der Bend, R.L., Van Corven, E.J., Jalink, K., Eichholtz, T. & Van Blitterswijk, W.J. (1992) Lysophosphatidic acid: a novel phospholipid with hormone- and growth factor-like activities. Cold Spring Harb. Symp. Quant. Biol. 17:163-167.

3. DiSaia, P.J., Morrow, M., Kanabus, J., Piechal, W. STownsend D.E. (1975) Two new tissue cultures lines from ovarian cancer. Gyn. Oncol. 3:215-219.

4. Freedman, R.S., Pihl, E., Kusyk, C, Gallager, H.S. & Rutledge, F. (1978) Characterization of an ovarian carcinoma cell line. Cancer 42:2352-2359.

5. Kanabus, J., Braunstein, G.D., Emry, P.K., DiSaia, P.J. & Wade, M.E. (1978) Kinetics of growth and ectopic production of human chorionic gonadotropin by an ovarian cystadenocarcinoma cell line maintained in vitro. Cancer Res. 38:765-770.

6. Pattillo, R.A , Story, M.T. & Ruckert, A.C.F. (1979) Expression of cell-mediated immunity and blocking factor using a new line of ovarian cancer cells in vitro. Cancer Res. 39:1185-1191.

7. Abu Sinna, G., Beekman, G., Lundgren, E., Nordenson, I. & Roos, G. (1979) Characterization of two new human ovarian carcinoma cell lines. Gyn. Oncol. 7:267-280.

8. Woods, L.K., Morgan, R.T., Quinn, L.A., Moore, G.E., Semple, T.U. & Stedman, K.E. (1979) Comparison of four new cell lines from patients with adenocarcinoma of the ovary. Cancer Res. 39:4449-4459.

9. Hamilton, T.C., Young, R.C, McKoy, W.M. et al. (1983) Characterization of a human ovarian carcinoma cell line (NIH:OVCAR-3) with androgen and estrogen receptors. Cancer Res. 43:5379-5389.

10. Simon, W.E., Albrecht, M., Haensel, M., Dietel, M. & Hoelzel, F. (1983) Cell lines derived from human ovarian carcinomas: growth stimulation by gonadotrophic and steroid hormones. J. Natl. Cancer Inst. 70:839-845.

11. Wilson, A.P. (1984) Characterization of a cell line derived from the ascites of a patient with papillary serous cystadenocarcinoma of the ovary. J. Natl. Cancer Inst. 72:513-521.

12. Buick, R.N., Pullano, R. & Trent, J.M. (1985) Comparative properties of five human ovarian adenocarcinoma cell lines. Cancer Res. 45:3668-3676.

13. Clamon, G., Loh, P-M., White, M. & Robinson, R. (1985) Development and characterization of a human ovarian cancer cell line which clones without agar in vitro. Gynecol. Oncol. 20:92-99. 14. Benard, J., Da Silva, J., de Blois, M-C. et al. (1985) Characterization of a human ovarian

adenocarcinoma line, IGROV1, in tissue culture and in nude mice. Cancer Res. 45:4970-4979. 15. Hill, B.T., Whelan, R.D.H., Gibby, E.M. et al. (1987) Establishment and characterization of three

new human ovarian carcinoma cell lines and initial evaluation of their potential in experimental chemotherapy studies. Int. J. Cancer 39:219-225.

16. Ishiwata, I., Ishiwata, C, Soma, M., Nozawa, S. & Ishikawa H. (1987) Characterization of newly established human ovarian carcinoma cell line: special reference of the effects of cis-platinum on cellular proliferation and release of CA 125. Gynecol. Oncol. 26:340-354.

17. Langdon, S.P., Lawrie, S.S., Hay, F.G. et al. (1988) Characterization and properties of nine human ovarian adenocarcinoma cell lines. Cancer Res. 48:6166-6172.

18. Briers, T.W., Stroobants, P., Vandeputte, T.M. et al. (1989) Establishment and characterization of a human ovarian neoplastic cell line DO-s. Cancer Res. 49:5153-5161.

19. Crickard, K„ Niedbala, M.J., Crickard, U. et al. (1989) Characterization of human ovarian and endometrial carcinoma cell lines established on extracellular matrix. Gynecol. Oncol. 32:163-173. 20. Golombick, T., Dansey, R., Bezwoda, W.R. & Rosendorff, J. (1990) Establishment and

characterization of two new human ovarian cancer cell lines UWOV1 and UWOV2 and a subline UWOV2 (Sf) growing in serum-free conditions: growth characteristics, biochemical and cytogenetic studies. In Vitro Cell. Dev. Biol. 26:447-454.

(19)

21. Yamada, T., Ueda, M., Otsuki, Y., Ueki, M. & Sugimoto, O. (1991) Establishment and characterization of a cell line (OMC-3) originating from a human mucinous cystadenocarcinoma of the ovary. Gynecol. Oncol. 40:118-128.

22. Jozan, S., Roche, H., Cheutin, F., Carton, M. & Salles, B. (1992) New human ovarian cancer cell line OVCCR1/sf in serum-free medium. In Vitro Cell. Dev. Biol. 28A:687-689.

23. Hirte, H.W., Kaiser, J.S. & Bacchetti, S. (1994) Establishment and characterization of four human epithelial ovarian carcinoma cell lines. Cancer 74:900-906.

24. Turner, J.T. SWyche, J.H. (1980) Regulation of metastatic ovarian cells in serum-free medium by epidermal growth factor and transferrin. J. Cell. Physiol. 104:233-240.

25. Golombick, T., Dajee, D.R. & Bezwoda, W.R. (1995) Extracellular matrix interactions 1: production of extracellular matrix with attachment and growth-sustaining functions by UWOV2 ovarian cancer cells growing in protein-free conditions. In Vitro Cell. Dev. Biol. 31:387-395.

26. Berchuck, A., Olt, G.J., Everitt, L, Soisson, A.P., Bast, R.C. & Boyer, C M . (1990) The role of peptide growth factors in epithelial ovarian cancer. Obstet. Gynecol. 75:255-262.

27. loachim, H.L., Sabbath, M., Andersson, B. & Barber, H.R.K. (1974) Tissue cultures of ovarian carcinomas. Lab. Invest. 31:381-390.

28. Hamburger, A.W. & Salmon, S.E. (1977) Primary bioassay of human tumor stem cells. Science 197:461-463.

29. Hamburger, A.W., Salmon, S.E., Kim, M.B. etal. (1978) Direct cloning of human ovarian carcinoma cells in agar. Cancer Res. 38:3438-3443.

30. Hamburger, A.W., Salmon, S.E. & Alberts, D.S. (1980) Development of a bioassay for ovarian carcinoma colony-forming cells. Progress in Clin. Biol. Res. 48:63-73.

31. Welander, CE., Natale, R.B. & Lewis, J.L. (1982) In vitro growth of human ovarian cancer cells by xenogeneic peritoneal macrophages. J. Natl. Cancer Inst. 69:1039-1042.

32. Ozols, R.F., Willson, J.K.V., Grotzinger, K.R. & Young, R.C. (1980) Cloning of human ovarian cancer cells in soft agar from malignant effusions and peritoneal washings. Cancer Res. 40:2743-2747.

33. Hamburger, A.W., White, C.P., Dunn, F.E., Citron, M.L. & Hummel, S. (1983) Modulation of human tumor colony growth in soft agar by serum. Int. J. Cell Cloning 1:216-229.

34. Carney, D.N., Bunn, P.A., Gazdar, A.F., Pagan, J.A. & Minna, J.D. (1981) Selective growth in serum-free hormone-supplemented medium of tumor cells obtained by biopsy from patients with small cell carcinoma of the lung. Proc. Natl. Acad. Sei. USA 78:3185-3189.

35. Sporn, M.B. & Roberts, A.B. (1988) Peptide growth factors are multifunctional. Nature 332:217-218.

36. Sporn, M.B. & Roberts, A.B. (1985) Autocrine growth factors and cancer. Nature 313:745-747. 37. Cohen, S. & Carpenter, G. (1975) Human epidermal growth factor: isolation and chemical and

biological properties. Proc. Nat. Acad. Sei. USA 72:1317-1321.

38. Broxterman, H.J., Sprenkels-Schotte, C , Engelen, Ph., Levy a, A. & Pinedo, H.M. (1987) Analysis of human ascites effect on clonogenic growth of human tumor cell lines and NRK-49F cells in soft agar. Int. J. Cell Cloning 5:158-169.

39. Zhou, L. & Leung, B.S. (1992) Growth regulation of ovarian cancer cells by epidermal growth factor and transforming growth factor a and b1. Biochim. Biophys. Acta 1080:130-136. 40. Kurachi, H., Morishige, K., Amemiya, K. et al. (1991) Importance of transforming growth factor

a/epidermal growth factor receptor autocrine growth mechanism in an ovarian cancer cell line in Vivo. Cancer Res. 51:5956-5959.

41. Crew, J.A., Langdon, S.P., Miller, E.P. & Miller, W.R. (1992) Mitogenic effects of epidermal growth factor andtransforming growth factor-a on EGF-receptor positive human ovarian carcinoma cell lines. Eur. J. Cancer 28:337-341.

42. Jozan, S., Guerrin, M., Mazars, P. et al. (1992) Transforming growth factor ß1 (TGF-ß1) inhibits growth of a human ovarian carcinoma cell line (OVCCR1) and is expressed in human ovarian tumors. Int. J. Cancer 52:766-770.

(20)

43. Hamburger, A.W., White, C.P. & Brown, R.W. (1981) Effect of epidermal growth factor on proliferation of human tumor cells in soft agar. J. Natl. Cancer Inst. 67:825-829.

44. Pathak, M.A., Matrisian, L.M., Magun, B.E. & Salmon, S.E. (1982) Effect of epidermal growth factor on clonogenic growth of primary human tumor cells. Int. J. Cancer 30:745-750.

45. Singletary, S.E., Baker, F.L., Spitzer, G. et al. (1987) Biological effect of epidermal growth factor on the in vitro growth of human tumors. Cancer Res. 47:403-406.

46. Wimalasena, J., Dostal, R. & Meehan, D. (1992) Gonadotropins, Estradiol, and growth factors regulate epithelial ovarian cancer cell growth. Gynecol. Oncol. 46:345-350.

47. Todaro, G.J., Fryling, C. & De Larco, J.E. (1980) Transforming growth factors produced by certain human tumor cells: polypeptides that interact with epidermal growth factor receptors. Proc. Natl. Acad. Sei. USA 77:5258-5262.

48. Shoyab, M., Plowman, G.D., McDonald, V.L., Bradley, J.G. & Todaro, G.J. (1988) Structure and function of human amphiregulin: a member of the epidermal growth factor family. Science 243:1074-1076.

49. Yamamoto, T., Ikawa, S., Akiyama, T. et al. (1986) Similarity of protein encoded by human c-erbB2 gene to epidermal growth factor receptor. Nature 319:230-234.

50. Lupu, R., Colomer, R., Zugmaier, G. et al. (1990) Direct interaction of a ligand for the erbB2 oncogene product with the EGF receptor and p185e/±>B2. Science 249:1552-1555.

51. Kraus, M.H., Issing, W., Miki, T., Popescu, N.C. & Aaronson, S.A. (1989) Isolation and characterization of ERBB3, a third member of the ERBB/epidermal growth factor receptor family: evidence for overexpression in a subset of human mammary tumors. Proc. Natl. Acad. Sei. USA 86:9193-9197.

52. Plowman, G.D., Culouscou, J.-M., Whitney, G.S. et al. (1993) Ligand-specific activation of HER4/ p180er6B4, a fourth member of the epidermal growth factor receptor family. Proc. Natl. Acad. Sei. USA 90:473-475.

53. Simpson, B.J.B., Phillips, H.A., Lessells, A.M. et al. (1995) c-erbB growth-factor-receptor proteins in ovarian tumours. Int. J. Cancer 64:202-206.

54. Roberts, A.B., Anzano, M.A., Lamb, L.C., Smith, J.M. & Sporn, M.B. (1981) New class of transforming growth factors potentiated by epidermal growth factor: isolation from non-neoplastic tissues. Proc. Natl. Acad. Sei. USA 78:5339-5343.

55. Bauknecht, T., Kiechle, M., Bauer, G. & Siebers, J.W. (1986) Characterization of growth factors in human ovarian carcinomas. Cancer Res. 46:2614-2618.

56. Ottensmeier, C, Swanson, L., Strobel, T., Druker, B., Niloff, J. & Cannistra, S.A. (1996) Absence of constitutive EGF receptor activation in ovarian cancer cell lines Br. J. Cancer 74(3):446-452. 57. Stromberg, K., Collins, T.J., Gordon, A.W., Jackson, C.L. & Johnson, G.R. (1992) Transforming

growth factor b acts as an autocrine growth factor in ovarian carcinoma cell lines. Cancer Res. 52:341-347.

58. Hanauske, A-R., Arteaga, C.L., Clark, G.M. et al. (1988) Determination of transforming growth factor activity in effusions from cancer patients. Cancer 61:1832-1837.

59. Morishige, K., Kurachi, H., Amemiya, K. etal. (1991) Evidence for the involvement of transforming growth factor a and epidermal growth factor receptor autocrine growth mechanism in primary human ovarian cancers in vitro. Cancer Res. 51:5322-5328.

60. Gordon, A.W., Pegues, J.C., Johnson, G.R., Kannan, B., Auersperg, N. & Stromberg, K. (1995) mRNA phenotyping of the major ligands and receptors of the EGF supergene family in human ovarian epithelial cells. Cancer Lett. 89:63-71.

61. Kurachi, H., Morishige, K., Adachi, H. etal. (1994) Implantation and growth of epidermal growth factor (EGF) receptor expressing human ovarian cancer xenografts in nude mice is dependent on EGF. Cancer 74:2984-2990.

62. Slamon, D.J., Godolphin, W., Jones, L.A. etal. (1989) Studies of the HER-2/neu protooncogene in human breast and ovarian cancer. Science 244:707-712.

(21)

63. Haldane, J.S., Hird, V., Hughes, C M . & Gullick, W.J. (1990) c-erfaB-2 oncogene expression in ovarian cancer. J. Pathol. 162:231-237.

64. Hung, M-C, Zhang, X., Yan, D-H. et al. (1992) Aberrant expression of the c-erbB-2/neu protooncogene in ovarian cancer. Cancer Lett. 61:95-103.

65. Johnson, G.R., Saeki.T., Auersperg, N. et al. (1991) Response to and expression of amphiregulin by ovarian carcinoma and normal ovarian surface epithelial cells: nuclear localization of endogenous amphiregulin. Biochem. Biophys. Res. Com. 180:481-488.

66. Roberts, A.B., Anzano, M.A., Lamb, L.C. etal. (1982) Isolation from murine sarcoma cells of novel transforming growth factors potentiated by EGF. Nature 295:417-419.

67. Anzano, M.A., Roberts, A.B., Meyers, C.A. et al. (1982) Synergistic interaction of two classes of transforming growth factors from murine sarcoma cells. Cancer Res. 42:4776-4778.

68. Moses, H.L., Yang, Y.Y. & Pietenpol, J.A. (1990) TGF-b stimulation and inhibition of proliferation: new mechanistic insights. Cell 63:245-247.

69. Massague, J. (1992) Receptors for the TGF-ß family. Cell 69:1067-1070.

70. Berchuck, A., Rodriguez, G., Olt, G. etal. (1992) Regulation of growth of normal ovarian epithelial cells and ovarian cancer cell lines by transforming growth factor-b. Am. J. Obstet. Gynecol. 166:676-684.

71. Marth, C, Lang, T., Koza, A., Mayer, I. & Daxenbichler, G. (1990) Transforming growth factor-beta and ovarian carcinoma cells: regulation of proliferation and surface antigen expression. Cancer Lett. 51:221-225.

72. Bartlett, J.M.S., Rabiasz, G.J., Scott, W.N., Langdon, S.P., Smyth, J.F. & Miller, W.R. (1992) Transforming growth factor-b mRNA expression and growth control of human ovarian carcinoma cells. Br. J. Cancer 65:655-660.

73. Hurteau, J., Rodriguez, G.C., Whitaker, R.S. et al. (1994) Transforming growth factor-beta inhibits proliferation of human ovarian cancer cells obtained from ascites. Cancer 74:93-99.

74. Henriksen, R., Gobi, A., Wilander, E., Oberg, K., Miyazono, K. & Funa, K. (1995) Expression and prognostic significance of TGF-b isotypes, latent TGF-b1 binding protein, TGF-b type I and type II receptors, and endoglin in normal ovary and ovarian neoplasms. Lab. Invest. 73:213-220. 75. Golombick, T., Dajee, D. & Bezwoda, W.R. (1995) Extracellular matrix interactions 2: extracellular

matrix structure is important for growth factor localization and function. In Vitro Cell. Dev. Biol. 31:396-403.

76. Ross, R., Raines, E.W. & Bowen-Pope, D.F. (1986) The biology of platelet-derived growth factor. Cell 1986;46:155-69.

77. Westermark, B. & Heldin, C-H. (1991) Platelet-derived growth factor in autocrine transformation. Cancer Res. 51:5087-5092.

78. Versnel, M.A., Haarbrink, M., Langerak, A.W., De Laat, P.A.J.M., Hagemeijer, A. & Van der Kwast, T.H. (1994) Human ovarian tumors of epithelial origin express PDGF in vitro and in vivo. Cancer Genet. Cytogenet. 73:60-64.

79. Henriksen, R., Funa, K., Wilander, E., Backstrom, T., Ridderheim, M. & Oberg, K. (1993) Expression and prognostic significance of platelet-derived growth factor and its receptors in epithelial ovarian neoplasms. Cancer Res. 53:4550-4554.

80. Burgess, W.H. & Maciag.T. (1989) The heparin-binding (fibroblast) growth factor family of proteins. Annu. Rev. Biochem. 58:575-606.

81. Goldfarb M. (1990) The fibroblast growth factor family. Cell Growth Diff. 1:439-445.

82. Crickard, K., Gross, J.L., Crickard, U. et al. (1994) Basic Fibroblast growth factor and receptor expression in human ovarian cancer. Gynecol. Oncol. 55:277-284.

83. Macaulay, V.M. (1992) Insulin-like growth factors and cancer. Br. J. Cancer 65:311-320. 84. Yee, D., Morales, F.R., Hamilton, T.C. & Von Hoff, D.D. (1991) Expression of insulin-like growth

(22)

85. Karasik, A., Menczer, J., Pariente, C. & Kanety, H. (1994) Insulin-like growth factor-l (IGF-I) and IGF-binding protein-2 are increased in cyst fluids of epithelial ovarian cancer. J. Clin. Endocrinol. Metab. 78(2):271-276.

86. Resnicoff, M., Ambrose, D., Coppola, D. & Rubin, R. (1993) Insulin-like growth factor-1 and its receptor mediate the autocrine proliferation of human ovarian carcinoma cell lines. Lab. Invest. 69(6):756-760.

87. Aggarwal, B.B.&Pocsik, E. (1992) Cytokines: from clone to clinic. Arch. Biochem. Biophys. 292:335-359.

88. Iversen, O.H. (1991 ) The hunt for endogenous growth-inhibitory and/or tumor suppression factors: Their role in physiological and pathological growth regulation. Adv. Cancer Res. 57:413-453. 89. Malik, S. & Balkwill, F. (1991) Epithelial ovarian cancer: a cytokine propelled disease? Br. J. Cancer

64:617-620.

90. Pennica, D., Nedwin, G.E., Hayflick, J.S. et al. (1984) Human tumor necrosis factor: precursor structure, expression and homology to lymphotoxin. Nature 312:724-729.

91. Naylor, M.S., Malik, S.T.A., Stamp, G.W.H., Jobling, T. & Balkwill, F.R. (1990) In situ detection of tumour necrosis factor in human ovarian cancer specimens. Eur. J. Cancer 26:1027-1030. 92. Naylor, M.S., Burke, F. & Balkwill, F.R. (1995) Cytokines and ovarian cancer. In: Sharp, F., Mason,

P., Blackett, T. & Berek, J. eds. Ovarian Cancer 3, 1st edition. London: Chapman & Hall Medical, pp.89-97.

93. Wu, S., Boyer, C M . , Whitaker, R.S. et al. (1993) Tumor necrosis factor a as an autocrine and paracrine growth factor for ovarian cancer monokine induction of tumor cell proliferation and tumor necrosis factor a expression. Cancer Res. 53:1939-1944.

94. Giavazzi, R., Garofalo, A., Bani, M.R. et al. (1990) lnterleukin-1-induced augmentation of experimental metastases from a human melanoma in nude mice. Cancer Res. 50:4771-4775. 95. Watson, J., Sensintaffar, J.L., Berek, J.S. & Martinez-Maza, O. (1990) Constitutive production of

interleukin 6 by ovarian cancer cell lines and by primary ovarian tumor cultures. Cancer Res. 50:6959-6965.

96. Lidor, Y.J.,Xu, F.I., Martinez-Maza, O. etal. (1993) Constitutive production of macrophage colony stimulating factor and IL-6 by human ovarian surface epithelial cells. Exp. Cell. Res. 207:332-339. 97. Wu, S., Rodabaugh, K., Martinez-Maza, O. et al. (1992) Stimulation of ovarian tumor cell

proliferation with monocyte products including interleukin-1, interleukin-6, and tumor necrosis factor-alpha. Am. J. Obstet. Gynecol. 166:997-1007.

98. Ramakrishan, S., Xu, F.J., Brandt, S.J., Niedel, J.E., Bast, R.C. & Brown, E.L. (1989) Constitutive production of macrophage colony-stimulating factor by human ovarian and breast cancer cell lines. J. Clin. Invest. 83:921-926.

99. Kacinski, B.M., Carter, D., Mittal, K. et al. (1990) Ovarian adenocarcinomas express fms-complementary transcripts and fms antigen, often with coexpression of CSF-1. Am. J. Pathol.

137:135-147.

100. Berek, S. & Martinez-Maza, O. (1994) Molecular and biologic factors in the pathogenesis of ovarian cancer. J. Reprod. Med. 39:241-248.

101. Kommoss, F., Wolfle, J., Bauknecht, T. et al. (1994) Co-expression of M-CSF transcripts and protein, FMS (M-CSF receptor) transcripts and protein, and steroid receptor content in adenocarcinomas of the ovary. J. Pathol. 174:111-119.

102.Bauknecht, T., Kiechle-Schwarz, M., Du Bois, A., Wolfle, J. & Kacinski, B. (1994) Expression of transcripts for CSF-1 and for the "macrophage" and "epithelial" isoforms of the CSF-1 R transcripts in human ovarian carcinomas. Cancer Detect. Prev. 18:231-239.

103.Filderman, A.E., Bruckner, A., Kacinski, B.M., Deng, N. & Remold, H.G. (1992) Macrophage colony-stimulating factor (CSF-1) enhances invasiveness in CSF-1 receptor-positive carcinoma cell lines. Cancer Res. 52:3661-3666.

104.Price, F.V., Chambers, S.K., Chambers, J.T. et al. (1993) Colony-stimulating factor-1 in primary ascites of ovarian cancer is a significant predictor of survival. Am. J. Obstet. Gynecol. 168:520-527.

(23)

105.Price, F.V., Chambers, S.K., Chambers, J.T. et al. (1993) Colony-stimulating factor-1 in primary ascites of ovarian cancer is a significant predictor of survival. Am. J. Obstet. Gynecol. 168:520-527. 106. Kutteh, W.H. & Kutteh, C.C. (1992) Quantitation of tumor necrosis factor-a, interleukin-1 b, and interleukin-6 in the effusions of ovarian epithelial neoplasms. Am. J. Obstet. Gynecol. 167:1864-1869.

107.Moradi, M.M., Carson, L.F., Weinberg, B., Haney, A.F., Twiggs, LB. & Ramakrishnan, S. (1993) Serum and ascitic fluid levels of interleukin-1, interleukin-6, and tumor necrosis factor-alpha in patients with ovarian epithelial cancer. Cancer 72:2433-2440.

108.Gastl, G., Plante, M., Finstad, C L et al. (1993) High IL-6 levels in ascitic fluid correlate with reactive thrombocytosis in patients with epithelial ovarian cancer. Br. J. Haematol. 83:433-441. 109.Buick, R.N., Fry, S.E. & Salmon, S.E. (1980) Effect of host-cell interactions on clonogenic carcinoma

cells in human malignant effusions. Br. J. Cancer 41:695-704.

110.Mantovani, A., Peri, G., Polentarutti, N., Bolis, G., Mangioni, C. & Spreafico, F. (1979) Effects on in vitro tumor growth of macrophages isolated from human ascitic ovarian tumors. Int. J. Cancer 23:157-164.

111. Salmon, S.E. & Hamburger, A.W. (1978) Immunoproliferation and cancer: a common macrophage-derived promoter substance. Lancet 1(8077):1289-90.

112.Vaage, J. &Agarwal, S. (1979) Endogenous tumor growth factor indicated by increased ornithine decarboxylase activityin malignant cells treated with host serum or ascites fluid. Cancer Res. 39:1511-1517.

113. Uitendaal, M.P., Hubers, H.A.J.M., McVie, J.G. & Pinedo, H.M. (1983) Human tumour clonogenicity in agar is improved by cell-free ascites. Br. J. Cancer 48:55-59.

114.Levine, A.E., Hamilton, D.A., Yeoman, L.C., Busch, H. & Brattain, M.G. (1984) Identification of a tumor inhibitory factor in rat ascites fluid. Biochem. Biophys. Res. Commun. 119:76-82. 115. Wilson, A.P., Fox, H., Scott, I.V., Lee, H., Dent, M. & Golding, P.R. (1991) A comparison of the

growth promoting properties of ascitic fluids, cyst fluids and peritoneal fluids from patients with ovarian tumours. Br. J. Cancer 63:102-108.

116. Mills, G.B., May, C, McGill, M., Roifman, C M . & Mellors, A. (1988) A putative new growth factor in ascitic fluid from ovarian cancer patients: identification, characterization, and mechanism of action. Cancer Res. 48:1066-1071.

117. Mills, G.B., May, C, Hill, M., Campbell, S., Shaw, P. & Marks, A. (1990) Ascitic fluid from human ovarian cancer patients contains growth factors necessary for intraperitoneal growth of human ovarian adenocarcinoma cells. J. Clin. Invest. 86:851-855.

118.Xu, Y. & Mills, G.B. (1995) Activation of human ovarian cancer cells: role of lipid factors in ascitic fluid. In: Sharp, F., Mason, P., Blackett, T. & Berek, J. eds. Ovarian Cancer 3, 1st edition. London, Chapman & Hall Medical, pp.121-135.

119.Hanahan D.J. (1986) Platelet activating factor: a biologically active phosphoglyceride. Annu. Rev. Biochem. 55:438-509.

120.Chao,W.& Olson, M.S. (1993) Platelet-activating factor:receptors and signal transduction. Biochem. J. 292:617-629.

121.Bussolino, F. SCanussi, G. (1995) Platelet-activating factor produced by endothelial cells. A molecule with autocrine and paracrine properties. Eur. J. Biochem. 229:327-337.

122. Bishop, W.R. & Bell, R.M. (1988) Assembly of phospholipids into cellular membranes: biosynthesis, transmembrane movement and intracellular translocation. Annu. Rev. Cell. Biol. 4:579-610. 123. Jalink, K., Hordijk, P.L. & Moolenaar, W.H. (1994) Growth factor-like effects of lysophosphatidic

acid, a novel lipid mediator. Biochim. Biophys. Acta 1198:185-196.

124. Van der Bend, R.L., Brunner, J., Jalink, K., Van Corven, E.J., Moolenaar, W.H. & Van Blitterswijk, W.J. (1992) Identification of a putative membrane receptor for the bioactive phospholipid, lysophosphatidic acid. EMBO 11:2495-2501.

(24)

125.Tigyi, G. & Miledi, R. (1992) Lysophosphatidates bound to serum albumin activate membrane currents in Xenopus oocytes and neurite retraction in PC12 pheochromocytoma cells. J. Biol. Chem. 267:21360-21367.

126.Kreps, D.M., Whittle, S.M., Hoffman, J.M. & Toews, M.L. (1993) Lysophosphatidic acid mimics serum-induced sensitization of cyclic AMP accumulation. FASEB J. 7:1376-1380.

127.Jalink, K. & Moolenaar, W.H. (1992) Thrombin receptor activation causes rapid neural cell rounding and neurite retraction independent of classic second messengers. J. Cell. Biol. 118:411-419. 128. Moolenaar, W.H. (1994) LPA: a novel lipid mediator with diverse biological action. Trends Cell.

Biol. 4:213-219.

129.Cowan, D.H. & Graham, J. (1983) Stimulation of human tumor colony formation by platelet lysate. J. Lab. Clin. Med. 102:973-986.

130.Cowan, D.H., Graham, J., Paskevich, M.C. & Quinn, P.G. (1983) Influence of platelet lysate on colony formation of human breast cancer cells. Breast Cancer Res. Treatm. 3:171-178.

131.Childs, C.B., Proper, J.A., Tucker, R.F. & Moses, H.L. (1982) Serum contains a platelet-derived transforming growth factor. Proc. Natl. Acad. Sei. USA 79:5312-5316.

132. Van Corven, E.J., Groenink, A., Jalink, K., Eichholtz, T. & Moolenaar, W.H. (1989) Lysophosphatidate-induced cell proliferation: identification and dissection of signalling pathways mediated by G proteins. Cell 59:45-54.

133. Van Corven, E.J., Van Rijswijk, A., Jalink, K., Van der Bend, R.L., Van Blitterswijk, W.J. & Moolenaar, W.H. (1992) Mitogenic action of lysophosphatidic acid and phosphatidic acid on fibroblasts. Biochem. J. 281:163-169.

134.lmamura, F., Horai, T., Mukai, M., Shinkai, K., Sawada, M. & Akedo, H. (1993) Induction of in vitro tumor cell invasion of cellular monolayers by lysophosphatidic acid or phospholipase D. Biochem. Biophys. Res. Commun. 193:497-503.

135.Xu, Y., Casey, G. & Mills, G.B. (1995) Effect of lysophospholipids on signaling in the human Jurkat Tcell line. J. Cell. Physiol. 163:441-450.

136.Xu, Y., Fang, X-J., Casey, G. & Mills, G.B. (1995) Lysophospholipids activate ovarian and breast cancer cells. Biochem. J. 309:933-940.

137. Moolenaar, W.H. (1995) Lysophosphatidic acid signalling. Curr. Opin. Cell. Biol. 7:203-210.

(25)

Referenties

GERELATEERDE DOCUMENTEN

Micrographs of the immunoreaction for TrkC in the normal dura mater (E) and in the meningotheliomatous- psammomatous meningioma (F)/ surrounding dural tissue; the

Wat zijn de zwakke plekken in de verschillende typen verzorgingsstaten en wat betekent dit voor de sociaal-economische posities van vrouwen binnen de Europese

(De vertegenwoordiging in de Belgische KMO’s is al enige tijd een wederkerend discussiepunt in contact tussen bonden en werkgevers.) In België is die

Met inachtneming van a) heeft de EOR het recht te worden geïnformeerd en geraadpleegd door het centrale manage­ ment over alle voorstellen die waarschijnlijk ernstige

economisch onevenwichtig proces, waarin men­ selijke arbeid in bepaalde beroepen, bedrijfstak­ ken, regio’s en landen verdrongen wordt, terwijl tegelijkertijd als gevolg van de

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly

Hypothesis 3 (H3): Right and left eurosceptic parties in the European Parliament will vote as independent groups more similar on issues which form the core of

Om te onderzoeken of er significante verschillen tussen de patiënten in de groepen hoge en lage gevoeligheid voor sociale vergelijking en de groepen goede en slechte