throbber
ENDOCRINOLOGY
`
`OE BREAST CANCER
`
`Edited by
`
`ANDREA MANNI, MD
`The Penny/[0472121 State University
`College ofMedicine, Hers/794 PA
`
`InnoPharma Exhibit 1069.0001
`
`HUMANA PRESS
`TOTOWA, NEW JERSEY
`
`l \
`
`\w
`/
`
`
`
`

`

`
`
`© 1999 Humana Press Inc.
`999 Riverview Drive, Suite 208
`Totowa, New Jersey 07512
`
`For additional copies, pricing for bulk purchases, and/or information about other Humana titles,
`contact Humana at the above address or at any ofthe following numbers: Tel: 973-256-1699;
`Fax: 973-256-8341; E—mail: humana@humanapr.c0m or visit our website at http://hutnanapress_com
`
`All rights reserved. No part ofthis book may be reproduced, stored in a retrieval system, or transmitted in any form or
`by any means, electronic, mechanical, photocopying, microfilming, recording, or otherwise without written permission
`from the Publisher.
`
`All articles, comments, opinions, conclusions, or recommendations are those of the author(s), and do not necessarily reflect
`the views ofthe publisher.
`
`This publication is printed on acid-free paper. ®
`ANSI 239.48-1984 (American National Standards lnstitute)
`Permanence ofl’aper for Printed Library Materials.
`
`Photocopy Authorization Policy:
`Authorization to photocopy items for internal or personal use, or the internal or personal use ofspecifie clients, is granted
`by Humana Press lnc., provided that the base fee of US $10.00 per copy, plus US $00.25 per page, is paid directly to
`the Copyright Clearance Center at 222 Rosewood Drive, Danvers, MA 01923. For those organizations that have been
`granted a photocopy license from the CCC, a separate system ofpayment has been arranged and is acceptable to Humana
`Press Inc. The fee code for users ofthe Transactional Reporting Service is: [0-89603-591-3/99 $10.00 + $00.25}
`
`Printed in the United StatesofAmerica.
`
`10
`
`9
`
`8
`
`7
`
`6
`
`5
`
`4
`
`3
`
`7
`
`1
`
`Library of Congress Cataloging-in-Publication Data
`
`Endocrinology of breast cancer/ edited by Andrea Manni
`p.
`cm.
`-(Contemporary endocrinology; 1 1)
`Includes index.
`ISBN 0-89603-591-3
`
`1. Manni, Andrea.
`l. Breast~Cancer—Endocrinc aspects.
`Contemporary endocrinology (Totwa, N. 1.):
`l l.
`
`11. Series:
`
`[DNLMz 1. Breast Neoplasms—physiopathology. 2. Breast Neoplasms—etiology.
`3. Breast Neoplasms—therapy. 4. Hormones. WP 87013565 1999]
`RC280.B8E54
`19999
`616.99‘449—dc21
`
`DNLM/DLC
`for Library of Congress
`
`98-33989
`ClP
`
`InnoPharma Exhibit 1069.0002
`
`

`

`This material may be protected by Copyright law (Title 17 US. Code)
`
`
`
`12
`
`Role of Angiogenesis in the
`
`Transition to Hormone
`
`Independence and Acquisition
`of the Metastatic Phenotype
`
`
`Francis G. Kern, PHD
`
`INTRODUCTION
`
`Breast cancer becomes a life-threatening illness when the malignant tumor cells have
`acquired the capability of invading through the basement membrane and into the circu—
`lation, where they are then disseminated to distant sites in the body. Therefore it may
`seem intuitive that tumors that have acquired a capability of stimulating the growth of
`new blood vessels would possess a greater threat to the patient, and indeed a good amount
`of correlative clinical data now link tumors that have a higher number of microvessels
`with poor prognosis (1—5). However, while neoangiogenesis is likely to be important in
`the dissemination of tumor cells, it is becoming more apparent that additional factors are
`required to allow proliferation of disseminated tumor cells at the distant sites.
`When breast cancer presents as an invasive disease, it has been useful to use estrogen
`receptor (ER) content as a basis for choice oftherapy. This relates to the fact that estrogen
`itself can stimulate the growth of breast cancer cells that contain ER through still
`unknown mechanism that involves binding of estrogen to nuclear ER and subsequent
`stimulation of its transcriptional activation functions. Consequently, a highly useful
`treatment for most patients with ER-positive tumors is hormonal therapy with
`antiestrogens, the most common being tamoxifen (6-8). Although other ER-independent
`mechanisms may also contribute to the therapeutic effect of this agent (9—11), it is
`commonly assumed that the benefit of this drug derives from its ability to bind to ER
`without stimulating the same type oftranscriptional activity as an estrogen-bound receptor.
`An invariable and unfortunate outcome of treatment of patients with tumors that
`initially respond to tamoxifen is the outgrowth of populations of cells that no longer
`respond to this treatment. In addition, some ER-positive tumors fail to Show any initial
`response to this relatively benign form of treatment. What may not seem so intuitive is
`how expression of angiogenic growth factors might also be involved in either acquired
`or de novo antiestrogen resistance, yet recent studies with transfected breast cancer cell
`lines (discussed in greater detail below) raise this as a possibility. If this turns out to be
`
`From: Contemporary Endocrinology: Endocrinology ofBreast Cancer
`Edited by: A. Manni © Humana Press Inc., Totowa, NJ
`
`169
`
`This material was {copied
`at the NLM and man: be
`
`InnoPharma Exhibit 1069.0003
`
`

`

`170
`
`Part III / Tumor Progression
`
`substantiated by clinical studies, then in the appropriate context, treatment with agents
`that inhibit angiogenesis may afford a means of restoring antiestrogen sensitivity to
`patients with acquired resistance or a means ofconferring sensitivity to patients with de
`novo resistance.
`
`CLINICAL DILEMMAS
`
`ASSOCIATED \WITH TAMOXIFEN TREATMENT
`
`A major dilemma for the clinician currently confronted with a patient with an ER—
`positive breast tumor is trying to make the best guess as to whether a patient will respond
`to this type of antiestrogen therapy in either an adjuvant or advanced breast cancer
`treatment setting. Since approximately 60% of patients with invasive ductal carcinoma
`present with ER-positive tumors, this particular question is relevant for a significant
`portion of women with breast cancer. It is also known that around 65—70% of patients
`with ER-positive tumors with advanced stage disease will show at least some response
`to tamoxifen (12,13). Among those that eventually fail on this therapy, 50—70% of these
`patients still have tumors that contain ER when these sites are biopsied (12—15). There—
`fore loss ofER is not the primary mechanism involved in the acquisition of antiestrogen
`resistance.
`
`Two different types of second-line antihormonal therapies are currently in the final stages
`of clinical trials for use in patients who have failed on tamoxifen. One approach involves the
`use of inhibitors of the enzyme aromatase, which is involved in the biosynthesis of estradiol
`produced at extraovarian sites (16—23). A second form of second-line therapy is the use of
`what have been called pure antiestrogens (24). These are compounds that bind to the ER,
`result in no transcriptional activation, and may increase the degradation of the receptor (25).
`They also do not possess any of the partial agonist properties of tamoxifen.
`Various recent studies have shown that 20~40% of patients who have failed on tam-
`oxifen therapy show some response to these second-line therapies and that an additional
`30% show no immediate disease progression when switched to these forms of therapy
`(19—23,26——28). Therefore these second-line approaches are likely to be of some value,
`but they also raise another dilemma for the oncologist. In addition to needing help from
`molecular oncologists and epidemiologists in determining who is most likely to respond
`initially to tamoxifen, the clinician will soon also need assistance in determining which
`patients who do not respond are likely to benefit from these alternative hormonal therapies.
`
`MECHANISMS OF TAMOXIFEN RESISTANCE
`
`A number of different mechanisms have been proposed for tamoxifen resistance.
`Based on what is known about some of these mechanisms, some educated guesses can
`be made as to whether the patient with a tamoxifen-resistant tumor that is due to one of
`these mechanisms is likely to respond to a second-line therapy. However, for many of
`these mechanisms for which a molecular basis for tamoxifen resistance has been demon-
`strated with cell lines or with in vitro assays, subsequent examination of patient samples
`has revealed that these types of alterations are unlikely to be responsible for most
`tamoxifen—resistant tumors. This includes point mutations in the hormone binding domain
`of the estrogen receptor, alterations in splicing of the ER mRNA that can result in con-
`stitutiver active forms of the receptor, or metabolism of tamoxifen to metabolites with
`estrogenic activity (8,29—35).
`
`This material was {a pied
`at the NLM and mav bie-
`
`InnoPharma Exhibit 1069.0004
`
`

`

`
`Chapter 12 / Angiogenic Growth Factors and Breast Cancer Progression
`171
`
`There is now recent evidence that the balance in the amounts of recently discovered
`steroid receptor coactivators and corepressors can influence whether tamoxifen will act
`as an antagonist or agonist of the estrogen receptor. At this point, work with in vitro
`systems suggests that the pure antiestrogens will still act as antagonists when tamoxifen
`is changed to an agonist as a result ofalterations in the ratio ofcoactivators to corepressors
`(36). Therefore other mechanisms are also probably responsible for those ER-positive
`tamoxifen-resistant breast tumors that fail to respond to second—line therapies.
`Two potential mechanisms involve the downstream signaling events associated with
`activation of growth factor receptors. Work from a number of laboratories has demon-
`strated what has been termed cross talk between steroid receptors and either growth factor
`tyrosine kinase receptors or G-protein-coupled receptors (3 7—44). In examples relevant
`to the question oftamoxifen resistance, phosphorylation ofthe estrogen receptor by MAP
`kinases, which are downstream effectors of a number of different signaling pathways
`including those mediated by growth factor receptors, or activation of G-protein-coupled
`receptors can result in tamoxifen acquiring agonistic properties (3 7,38, 43). However, the
`same studies show that the ER is not activated by these mechanisms when pure
`antiestrogens are present (3 7—43).
`Therefore at least one additional mechanism probably remains underlying the de novo
`or acquired tamoxifen resistance of ER-positivc tumors that do not subsequently respond
`to a second—line treatment with the pure antiestrogens. Recent data suggest that one such
`mechanism may also involve growth factor signaling, which provides the breast tumor
`cell with an alternative pathway to the growth stimulation mediated by ER (45). This
`would bypass the requirement for activation ofthe ER by an agonist for growth promotion
`. to occur and would account for the failure of second-line antihormonal therapies.
`
`ROLE OF GROWTH FACTORS IN THE
`
`ACQUISITION OF TAMOXIFEN RESISTANCE
`
`It can now be seen that growth factor signaling can affect the resistance of a breast
`tumor cell to tamoxifen in a number of different ways, with two of these mechanisms
`potentially leading to tamoxifen acting as an agonist. In one scenario, growth factor
`signaling can affect the level of coactivators or corepressors. Although no experimental
`data currently exist indicating that this type of regulation is indeed occurring in breast
`cancer cells, the field is very new, the number of coactivators and corepressors being
`identified is continually expanding, and the experiments remain to be performed (46). In
`a second scenario, growth factor signaling could result in activation of ER via phospho—
`rylation, leading to increased agonistic activity for tamoxifen. In the third scenario,
`growth factor signaling could bypass the requirement for ER activation for growth. These
`three scenarios are not mutually exclusive, and all three mechanisms may be operating
`simultaneously within a cell to provide additive or perhaps synergistic growth-stimula—
`tory effects. As such, these mechanisms may account for the clinical response observed
`in some patients when tamoxifen is withdrawn (4 7,48). However the available evidence
`to date suggests that a better understanding is required of what particular growth factor
`signaling pathways can bypass the need for ER since these pathways may also be respon-
`sible for resistance to second—line therapies.
`The initial indication of the potential importance growth factors might have in the
`process of the development of hormone independence came from work by Dickson and
`
`This material was tooled
`at the NLM and man: be
`
`InnoPharma Exhibit 1069.0005
`
`

`

`172
`
`Part III / Tumor Progression
`
`Lippman (49). These two investigators formulated the ideas that the genes for growth
`factors were part of the set of genes that were transcriptionally activated in ER-positive
`cells in response to estrogen treatment and that the subsequent growth stimulation of
`ER-positive breast cancer cell line xenografts in nude mice was the result of either the
`autocrine or paracrine effects ofthese factors (49). This line ofreasoning was supported
`by findings that continuous administration of epidermal growth factor (EGF) could
`facilitate the hormone-independent growth of an estrogen-dependent cell line as xe-
`nografts in ovariectomized athymic nude mice (50) and that transforming growth fac—
`tor—0t (TGF-oc), another ligand for the EGF receptor, was induced by estrogen in the
`same cell line (51—54).
`Subsequently a number of laboratories have used a transfection approach to express
`constitutively growth factors or growth factor receptors whose overexpression was impli-
`cated by various clinical studies to be associated with estrogen-independent tumors or
`induced by estrogen in vitro (39, 55—66). Molecules such as protein kinase c (PKC)-OL,
`ras“, and raf-l, which are downstream effectors of many growth factor signaling path-
`ways, have also been overexpressed in estrogen-dependent cell lines (6 7—72). The trans-
`fected cell lines were then tested to determine whether the overexpression had any effect
`on the estrogen responsiveness or antiestrogen sensitivity in vitro or in vivo.
`Many of these studies utilized the MCF-7 breast cancer cell line as a recipient since
`it is estrogen responsive in vitro and sensitive to treatment with antiestrogens (73). In
`vivo, there is a strict estrogen dependence for tumor formation, and tumors will not
`develop in ovariectomized nude mice unless they are supplemented with estrogen (74, 75).
`Even though this cell line was derived from the pleural effusion of a patient with breast
`cancer (76), the tumors that do develop with this cell line and most other breast cancer
`cell lines capable of in vivo growth are noninvasive and nonmetastatic (69, 77). This in
`vivo phenotype seen with MCF-7 cells is in line with the poor invasive ability seen in in
`vitro Matrigel basement membrane invasion assays (78). Consequently, this cell also
`represents a useful model to determine the effects ofoverexpression on the metastatic and
`invasive phenotypes. To facilitate this process, we developed a clonal line of MCF-7
`cells, designated as the ML-20 cell line, which stably expresses a transfected bacterial
`lacZ gene (77, 79). In many of our studies we have used this cell line as a recipient for
`retransfection since this allows a chromogenic substrate for B-galactosidase to be used
`to stain metastatic deposits of cells blue.
`
`EFFECTS OF ANGIOGENIC GROWTH FACTOR
`
`OVEREXPRESSION ON THE METASTATIC PHENOTYPE
`
`Varying degrees of increased estrogen-independent in vitro or in vivo growth can be
`conferred to ER-positive breast cancer cell lines by transfection with vectors for EGFR
`(56,5 7, 64) HER-2/neu (62, 63), heregulin (39,66), activated rasH (67—70), constitutively
`activated raf—l (71), PKC-(x (72), TGF-B (61), insulin-like growth factor (IGF)-II (59, 60),
`fibroblast growth factor (EGF)-l (65) and FGF-4 (55). In those transfection studies in
`which estrogen-independent in vivo growth was a consequence ofoverexpression, it was
`only with the PKC-a and FGF-1 and FGF-4 transfectants that an increased metastatic
`phenotype was also consistently observed (55, 65, 72). F GF-overexpressing cells acquire
`the ability to form tumors in ovariectomized athymic nude mice or beige mice without
`estrogen supplementation. They can also form tumors in either nude or beige mice treated
`
`ThiS material was rapied
`at the NLM arm max: ma-
`
`InnoPharma Exhibit 1069.0006
`
`

`

`Chapter 12 / Angiogenic Growth Factors and Breast Cancer Progression
`
`173
`
`with tamoxifen pellets. In either type ofmice, and in either untreated or tamoxifen-treated
`mice, X-gal-stained cells can be readily detected in either the lungs or lymph nodes of
`tumor-bearing mice injected with FGF-l- or FGF-4 overexpressing cells (45, 65, 77). By
`contrast, no cells are detected in lungs or lymph nodes of mice bearing control vector-
`transfected ML-20 cell tumors in estrogen-supplemented mice, even when these tumors
`are the same size as the tumors that form in mice injected with FGF-overexpressing cells
`(65). However, if the same ML-20 cells are coinjected with FGF-4-overexpressing cells
`that do not themselves express B—galactosidase, X—gal-stained cells can be detected in the
`tumors of either untreated or tamoxifen~treated ovariectomized nude mice and in the
`
`lungs and lymph nodes of mice bearing tumors composed of the mixture (55).
`The underlying mechanism responsible for the increased metastatic phenotype asso-
`ciated with PKC-oc overexpression remains to be elucidated. Both FGF-1 and FGF-4 are
`angiogenic growth factors and both can therefore stimulate the growth of new blood
`vessels in a tumor producing either factor (80,81). Thus the mechanism likely to be
`responsible for the increased diSSemination of tumor cells is likely to be related to the
`increased neoangiogenesis that is occurring in these tumors.
`Although deposits of X-gal-stained cells can be seen in essentially 100% ofthe mice
`bearing tumors of either FGF-4- or FGF- 1 ~0verexpressing cells, we never observed the
`development of tumor nodules at either of these sites (65, 77). Since the disseminated
`tumor cells express the cDNAs for both hygromycin and G418, it is a relatively simple
`task to establish cell lines from the tumor cells present in the lungs or lymph nodes. These
`cells continue to express FGF and remain capable of forming tumors in either untreated
`or ovariectomized mice. However, these cells also do not appear to be able to develop into
`macrometastases. When inoculated via a tail vein route, these tumor cells disappear from
`the lungs with approximately the same kinetics as FGF—overexpressing cells that had not
`been established from lungs of tumor-bearing mice. With both cell types, X-gal-stained
`cells can no longer be detected by 96 h after injection (82).
`Long-lived angiogenic inhibitors produced by the primary tumor cells have been
`shown to be capable of inhibiting the development of macrometastases from cells that
`have been disseminated from the primary site. In these model systems, macrometastases
`are found when the primary tumor is resected (83—86). However this was not the case
`when the primary tumors of FGF-l-overexpressing cells were resected. Tumors were
`allowed to develop for 3 weeks, at which point the tumors were of sufficient size that
`100% of the animals sacrificed at this point had large numbers of disseminated cells
`present in both the lungs and lymph nodes. Tumors were resected from the remaining
`animals and after 3 weeks, this group of animals was sacrificed. Examination of the lungs
`and lymph nodes did not reveal any X-gal-stained cells in these organs (82).
`Taken together, these results indicate that the increased neoangiogenesis brought
`about by the production of an angiogenic growth factor facilitates the constant shedding
`of tumor cells into the circulation. However, the cells that accumulate within the lungs
`and lymph nodes of tumor-bearing mice do not possess any selective advantage at these
`distant sites compared with the cells remaining in the primary tumor. These cells are
`lacking in the ability to either attach, extravasate, or proliferate at these distant sites; as
`a consequence, they are cleared from these organs within a relatively short period. The
`mechanism of clearance remains to be elucidated but may involve an increased rate of
`apoptosis relative to tumor cells at the primary tumor site or attack by cells of the remain-
`ing immune system present in the athymic nude mice.
`
`This material was {copied
`at the NLM and man: be
`
`InnoPharma Exhibit 1069.000? i
`
`

`

`174
`
`Part III / Tumor Progression
`
`In addition to facilitating the spread of tumor cells, the production of an angiogenic
`growth factor would presumably confer some growth advantage over parental MCF-7
`cells at these distant sites if the other missing components of the metastatic process were
`otherwise present. Consequently, the FGF-transfected cell lines are likely to be a useful
`model in determining the ability of the various proteases, attachment factors, apoptosis
`modulators, and other putative regulators ofthe metastatic process to facilitate the devel-
`opment of macrometastases in this system.
`
`MECHANISMS MEDIATING ESTROGEN—INDEPENDENT
`
`GROWTH IN CELLS OVEREXPRESSING GROWTH FACTORS
`
`A critical question is whether the estrogen-independent growth conferred by FGF
`overexpression is the result of a mechanism that activates ER or bypasses the need for
`an ER-regulated growth pathway. Heregulin overexpression can also confer on MCF-7
`cells the ability to form tumors in ovariectomized nude mice (39,66). Heregulin is a
`ligand for homodimers of erbB-3/HER-3 and erbB-4/HER-4 and heterodimers of
`erbB-Z/HER-Z/neu with either of these two transmembrane tyrosine kinase receptors
`(87,88). Recent clinical evidence shows that patients with tumors overexpressing
`HER-2/neu respond poorly to tamoxifen therapy (89), implying that activation of the
`erbB family of receptors is involved in resistance to this antiestrogen. In heregulin-
`overexpressing MCF-7 cells, the ER becomes tyrosine phosphorylated, and the num-
`bers ofER are downregulated (39, 66). There is also evidence from transient expression
`assays with estrogen response element (ERE)-containing reporter constructs that the
`ER is constitutively activated in heregulin-overexpressing cells and that this effect can
`be reversed by treatment with the pure antiestrogen ICI 182,780 (39). This raises the
`possibility that cells that have acquired resistance to tamoxifen through the activation of
`the erbB family of transmembrane tyrosine kinase receptors may be able to respond to
`this second-line antihorrnonal therapy.
`By contrast, the evidence available to date suggests that signaling mediated by FGF
`receptors may instead confer on ER-positive breast cancer cells a mechanism that can
`bypass the need for ER activation. FGF-overexpressing cells have obtained the ability to
`form soft agar colonies with high efficiency in media depleted ofestrogens. In media that
`contain estrogen, the effect of added 4-hydr0xytamoxifen is diminished compared with
`control transfected cells. The same holds true in anchorage-dependent growth assays in
`which FGF-overexpressing cells have faster doubling times in estrogen-depleted media
`or in estrogen-containing media with added 4-hydroxytamoxifen (65). The enhanced
`anchorage-dependent or anchorage-independent growth in estrogen-depleted media is
`not abrogated by treatment with the pure antiestrogen ICI 182,780, as might be expected
`if the effect were due to ligand-independent activation of ER mediated by FGF signaling
`pathways. Similarly, the FGF-transfected cells continue to show enhanced growth in
`either anchorage-dependent or anchorage-independent growth assays when ICI 182,780
`is added to estrogen-containing media. There is no downregulation of ER in FGF-trans-
`fected cells, and there is no evidence from analysis ofthe basal levels ofestrogen-induced
`genes or from transient assays with ERE-containing reporter constructs that FGF-
`overexpressing cells contain a constitutively activated ER. Furthermore, FGF-over-
`expressing cells continue to be able to form tumors in mice that have been treated with
`ICI 182,780 (45).
`
`This material was {sawed
`at the NLM and max Ea
`
`InnoPharma Exhibit 1069.0008
`
`

`

`Chapter 12 / Angiogenic Growth Factors and Breast Cancer Progression
`
`175
`
`ROLE OF PARACRINE AND AUTOCRINE EFFECTS
`
`OF FGF IN MEDIATING TAMOXIFEN RESISTANCE
`
`Although FGF—4 is not expressed in human breast tumor tissues, there is considerable
`evidence that both FGF-1 and all four of the FGF transmembrane tyrosine kinase receptors
`are expressed in many breast tumor tissues (65, 90—1 00). The transfection studies discussed
`above certainly suggest a potential role for FGF expression in the acquisition of hormone
`independence or antiestrogen resistance in ER-positive breast tumors. Since FGF-1 can
`have both paracrine and autocrine or intracrine effects, it is important to dissect the relative
`contributions of these effects to the phenotypes observed in FGF-l-overexpressing cells.
`This process can be performed by retransfecting the FGF-l-overexpressing cells with a
`dominant—negative FGF receptor. Receptor dimerization is required for transmission ofthe
`signal resulting from FGF binding (101,102). Therefore, a construct directing the expres-
`sion ofa transmembrane-anchored truncated FGF receptor lacking the cytoplasmic tyrosine
`kinase domain will effectively block the transmission of the growth signal through the
`formation of nonfunctional homo- and heterodimers. In such a pairing, the mutant lacking
`the tyrosine kinase domain will not be capable of phosphorylating the wild-type receptor
`on tyrosine residues in the cytoplasmic regulatory region of the receptor and thus will
`prevent the binding of downstream signaling molecules (103—105).
`FGF-l—overexpressing cells retransfected with the dominant negative FGF receptor no
`longer contained elevated levels ofphosphorylated MAPKs in cell extracts, indicating that the
`dominant negative receptor was blocking FGF signaling. As expected, these cells were also
`no longer capable of forming soft agar colonies in either estrogen-depleted or antiestrogen-
`containing media, confirming that autocrine or intracrine effects of FGF were important in
`conferring this phenotype. In line with these in vitro results, when they are injected into
`ovariectomized nude mice, the retransfected cells are no longer capable of forming tumors
`without estrogen supplementation. However, the tumors that form in estrogen-supplemented
`animals are typically much larger than those formed by MCF—7 cells, suggesting that the
`paracrine effects of FGF-1 production can act in an additive or synergistic manner with the
`mitogenic effect ofestrogen to stimulate tumor growth. An interesting finding from this study
`was the observation that in tamoxifen-treated animals, the FGF—overexpressing cell lines
`formed tumors at the same rate regardless of whether or not they were also expressing the
`dominant negative receptor. Thus the paracrine effects alone ofFGF were capable ofallowing
`antiestrogen-resistant in vivo tumor growth (106). These results also indicate that tamoxifen
`resistance can be dissociated from estrogen independence.
`It has been suggested that the partial agonistic properties of tamoxifen may be greater in the
`mouse than they are in the human (107). When tamoxifen-treated mice are injected with MCF—7
`cells that do not overexpress FGF, small tumors sometimes develop, but they usually remain
`static or regress (75, I 08). Thus it is possible that the paracrine effects ofFGF may cooperate with
`the weak mitogenic activity of tamoxifen to allow continued and progressive tumor develop-
`ment to occur. While this an intriguing possibility, it remains to be determined if these results
`seen in nude mice have any relevance to the tamoxifen resistance that occurs in human patients.
`
`EFFECTS OF VEGF OVEREXPRESSION ON ANTIESTROGEN
`
`SENSITIVITY AND THE METASTATIC PHENOTYPE
`
`As previously stated, FGF-1 is a strong angiogenic growth factor (80). Therefore, one
`of the paracrine effects that is likely to be a strong contributor to the observed tumor
`
`This material was {retried
`at the NLM and man: be
`
`InnoPharma Exhibit 1069.0009
`
`

`

`176
`
`Part III / Tumor Progression
`
`growth of the dominant-negative receptor-transfected cell lines in tamoxifen-treated
`animals is the neoangiogenesis resulting from the continued production of this growth
`factor. To test the effect of neoangiogenesis directly, B-galactosidase-expressing MCF-7
`cells were transfected with a vector directing the overexpression of the l65-amino acid
`form of vascular endothelial growth factor (VEGF). There is a considerable amount of
`evidence that expression of this particular isoform of this growth factor is an important
`determinant in the establishment ofa neoangiogenic phenotype in many types ofcancers.
`VEGF has been shown to be expressed in breast tumors (93,109—113), and the extent of
`expression correlates with the number of microvessels present in the tumor (109). This
`is a growth factor that is relatively specific for endothelial cells. There are two transmem-
`brane tyrosine kinase receptors for VEGF. One is known as Flkl in the mouse and as KDR
`in the human; the second is called Flt-1 (114—11 7). Expression of both receptors is
`restricted to endothelial cells, hematopoietic stem cells, monocytes, megakaryocytes,
`and platelets (1 16—120). mRNA for these receptors is strongly expressed in the endothe—
`lial cells of small vessels adjacent to infiltrating ductal carcinoma or metastatic ductal
`carcinoma cells as well as in the endothelial cells adjacent to comedo-type ductal carci-
`noma in situ. However, there is no evidence ofexpression ofeither receptor in endothelial
`cells in the normal areas of the breast (113).
`There are a few reports of ectopic expression of VEGF receptors in some types of
`tumor cells (120—123), but an examination of the recipient ML-20 cell line using a
`sensitive reverse transcription polymerase chain reaction assay has failed to show any
`evidence of expression of either receptor in this MCF-7-derived breast cancer cell line.
`In line with this finding, cells overexpressing either the 121- or 165-amino acid forms of
`VEGF do not show any altered in vitro growth properties with regard to growth in either
`estrogen-depleted media or in antiestrogen-containing media (124). Both types ofVEGF
`transfectants are unable to form tumors in ovariectomized mice without estrogen supple—
`mentation. Overexpression ofthe 121-amino acid form ofVEGF was reported to increase
`the in vivo growth rate in estrogen-supplemented animals (125), but no statistical differ-
`ence was observed between control transfectants and transfectants overexpressing the
`l65-amino acid form of VEGF in this treatment group (124). This may relate to the fact
`that different sublines of MCF-7 cells have different in vivo growth rates in estrogen-
`supplemented mice and that the controls for the 121-amino acid transfection study grew
`poorly whereas the controls for the 165—amino acid study grew well in estrogen-supple—
`mented mice, making any further stimulation due to VEGF overexpression difficult to
`observe.
`
`No effect of overexpression of the 121-amino acid form of VEGF on either tamoxifen
`sensitivity or metastatic potential was seen in the one study. However, overexpression of
`the 165-amino acid form was found to increase the tumorigenicity of MCF-7 cells in
`tamoxifen-treated animals and to facilitate tumor cell dissemination. For the effect of
`
`tamoxifen to be observed, coinjection with Matri gel was required (124). This is a mixture
`of basement membrane components that has been demonstrated to increase the tumori-
`genicity of a number of human cancer cell lines, including MCF~7 cells (126,127). The
`12 1 -amino acid form ofVEGF does not bind to heparin, whereas the 165-amino acid form
`is a heparin-binding growth factor (128). It is difficult to compare studies with two
`different growth factor isoforms performed in two different laboratories. The difference
`in effects observed may simply relate to differences in expression levels. However, it is
`also possible that this difference in heparin binding capability may also explain the
`Th is material was {opiad
`at the NLM arm mav Ea-
`
`InnoPharma Exhibit 1069.0010
`
`

`

`
`
`Chapter 12 / Angiogenic Growth Factors and Breast Cancer Progression 177
`
`apparent difference between the 121- and l65-amino acid forms with regard to tamoxifen,
`since heparan-sulfate—containing proteoglycans present in the Matrigel mixture may
`protect the VEGF from inactivation due to binding to (x2—macroglobulin (129). Small
`amounts of other growth factors are present in the Matrigel mixture, and a cooperat

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket