throbber
Vol. 8, 221 232, January 2002
`
`Clinical Cancer Research 221
`
`Differences in Therapeutic Indexes of Combination Metronomic
`
`Chemotherapy and an Anti-VEGFR—Z Antibody in Multidrug-
`resistant Human Breast Cancer Xenografts1
`
`Giannoula Klement, Ping Huang, Barbara Mayer,
`Shane K. Green, Sban Man, Peter Bohlen,
`Daniel Hicklin, and Robert s. Kerbelz
`Sunnybrook and Women’s College Health Sciences Centre, Molecular
`and Cellular Biology, Toronto, Ontario, M4N 3 M5 Canada [G. K.,
`P. H., B. M., S. K. G., S. M., R. S. K.], and ImClone Systems, Inc.,
`New York, New York 10014 [P. B., D. H.] and Department of
`Medical Biophysics, University of Toronto [R. S. K.]
`
`ABSTRACT
`
`One of the greatest barriers to the treatment of cancer
`with chemotherapeutic drugs is acquisition of drug resist-
`ance. This includes multidrug resistance mediated by P-
`glycoprotein (ng) to multiple lipophilic natural compounds
`such as taxanes, doxorubicin (Adriamycin), and vinblastine.
`The considerable efforts made thus far to reverse this and
`
`other types of drug resistance have had very limited success.
`We report here that a variety of orthotopic human breast
`cancer xenografts selected for high levels of ng and multi-
`drug resistance respond in a significant and durable manner
`to different continuous low-dose (e.g., one-tenth the maxi-
`mum tolerated dose of chemotherapy) chemotherapy regi-
`mens, when used in combination with an antivascular endo-.
`thelial cell growth factor (anti-VEGF) receptor-2 (ilk-1)-
`neutralizing
`antibody
`(DC101). The ng substrates
`paclitaxel (Taxol), Adriamycin, and vinblastine were all ef-
`fective using this type of combination treatment, although
`the chemotherapy protocols showed little or no effect as
`monotherapies. Similar results were also obtained using
`cisplatinum (a non-ng substrate drug) against cisplatinum-
`resistant tumors. Evidence of significant tumor cell death by
`the combination treatment was detected within 3 weeks of
`
`in the
`initiation of therapy by histopathological analysis,
`absence of shrinkage of tumor mass. There were, however,
`marked differences in the cumulative toxicity of long-term
`
`Received 7/26/01; revised 9/18/01; accepted 9/18/01.
`The costs of publication of this article were defrayed in part by the
`payment of page charges. This article must therefore be hereby marked
`advertisement in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`' Supported by grants to R. S. K. by the National Institutes of Health
`(Grant CA-41233) and the Canadian Institutes of Health Research, and
`by contract fimds from ImClone Systems, New York. G. K. was funded
`for most of this work by the Terry Fox Fellowship of the National
`Cancer Institute of Canada.
`2 To whom requests for reprints should be addressed, at Molecular &
`Cellular Biology Research, S-218 Research Building, Sunnybrook and
`Women’s College Health Sciences Centre, 2075 Bayview Avenue,
`Toronto, Ontario, Canada M4N 3 M5. Phone: (416) 480-5711; Fax:
`(416) 480-5703; E-mail: robert.kerbel@swchsc.on.ca.
`
`regimens of Adriamycin and cisplatinum, where toxicity
`was observed, when compared with the tubulin inhibitors,
`vinblastine and Taxol, where it was not. We conclude that
`vascular-targeting protocols involving frequent adminis-
`tration of very low doses of certain chemotherapeutic
`drugs can provide a stable and safe way to circumvent
`multidrug resistance in established orthotopically grow-
`ing tumors, as long as these are used in combination with
`a second antiangiogenic drug, in this case, anti-VEGFR—Z
`blocking antibodies.
`
`INTRODUCTION
`
`One of the major factors that account for the limited
`advances made in cancer treatment is acquired drug resistance.
`Cancers that respond to chemotherapeutic drugs such as ovarian,
`breast, colon, and non-small cell lung cancer, or to antihormonal
`therapies. e.g., breast and prostate cancer, almost always relapse
`in a resistant form some time later. Information is also emerging
`that even the newest generation of anticancer drugs such as the
`signal
`transduction inhibitors (e.g.,
`the bcr-abl antagonist.
`ST1571) may be compromised by acquired drug resistance (1—
`3). The numerous and diverse genetic instabilities of cancer cells
`are thought to be a major factor in explaining the propensity of
`cancer cells to give rise to such drug-resistant mutant or variant
`subpopulations (4, 5).
`Various strategies to circumvent or reverse acquired resist-
`ance to chemotherapeutic drugs have had little clinically signif-
`icant success thus far, with respect to the treatment of the
`common adult solid malignancies. Examples of such strategies
`include the use of combination chemotherapy, multimodality
`therapies, and the use of drugs such as ng3 antagonists to block
`the function of this particular mediator of multidrug resistance
`to natural
`lipophilic compounds (6). A new and seemingly
`counterintuitive preclinical strategy to combat drug resistance in
`cancer was developed recently (7). which exploits “chemother-
`apeutics as antiangiogenic agents,” i.e.,
`the property of such
`drugs to damage or kill the genetically stable, host endothelial
`cells of a tumor’s newly, formed neovasculaturc (8, 9). It in-
`volves the use of various chemotherapeutic drugs, e.g., cyclo-
`phosphamide (8) or vinblastine (9), given frequently and in a
`chronic manner (with no significant rest periods), at doses lower
`than the MTD. The administration of chemotherapeutic drugs in
`
`3 The abbreviations used are: ng, P-glycoprotein; MTD, maximum
`tolerated dose of chemotherapy; EC, endothelial cell; VEGF, vascular
`endothelial cell growth factor; VEGFR-Z, type 2 receptor for vascular
`endothelial cell growth factor; SCID, severe combined immunodefi-
`cient; HUVEC, human umbilical vein endothelial cell; MDR, multidrug
`resistance.
`Genentech 2083
`
`Hospira v. Genentech
`|PR2017—00737
`
`Downloaded from clincancerres.aacrjournals.org on December 6, 2017. © 2002 American Association for Cancer
`
`Genentech 2083
`Hospira v. Genentech
`IPR2017-00737
`
`1
`
`

`

`222 Chemotherapy and Anti-VEGFR-Z Antibody in Breast Cancer
`
`this manner has been termed “antiangiogenic” or “metronomic”
`chemotherapy (8, 10).
`A rationale for this type of therapeutic approach is that the
`dividing ECs of newly forming tumor vessels (1]) should be
`sensitive to chemotherapeutic drugs, similar to other types of
`normal dividing cells such as hair follicle, bone marrow, or gut
`mucosa] cells (4). However, because such cells lack the genetic
`instabilities of tumor cells, their ability to mutate and acquire
`resistance properties would be expected to be much more lim-
`ited (4, 5, 12). Indeed, Browder et a1.
`(8) have shown that
`chemotherapeutic drugs given at the MTD can cause EC apo-
`ptosis of tumor-associated vessels in ectopically growing mouse
`tumors, but this damage can be repaired rapidly during the
`prolonged recovery periods necessary for myeloid recovery
`following MTD chemotherapy. Hence, by giving chemotherapy
`more frequently, e.g., weekly or twice weekly, the EC repair
`process can be compromised and the potential antiangiogenic
`effects of chemotherapy enhanced (8).
`These preclinical results may provide an explanation of the
`clinical cases in which patients not responsive to standard MTD
`chemotherapy respond to the same drug at a lower dose, but
`administered more frequently (13—15). In the past such sched-
`ules were used primarily for palliation, because of the less
`severe side effects (14, 15). The availability of oral chemother-
`apeutic drugs (16) makes chemotherapy administered in this
`manner a more practical possibility.
`The long-term antitumor efficacy of antiangiogenic/metro-
`nomic chemotherapy protocols in ectopic syngeneic mouse or
`human tumor xenograft models can be increased, sometimes
`substantially so, by combination with a second,,antiangiogenic
`drug such as D(chloroacetyl-carbamoyl) Fumagillo]
`(8) or
`blocking monoclonal antibodies to VEGFR-2 (9). The rationale
`for this combination is that anti-VEGFR-Z- or anti-VEGF-
`
`targeting drugs (17). agents capable of specific blockade of
`activated EC cell survival mechanisms (18), will selectively
`enhance the damaging or cytotoxic effects of continuous low-
`dose chemotherapy on newly formed blood vessels (9).
`The purpose of the present paper was to address several
`important questions that the previous preclinical studies on
`antiangiogenic/metronomic chemotherapy have raised. First,
`do different classes of chemotherapeutic agents have similar
`potentials in terms of both efficacy and toxicity, when used
`alone or in combination with a second drug such as anti-
`VEGFR-2 antibodies? Would this type of therapeutic ap-
`proach work as well on orthotopically transplanted tumors?
`Finally. would the approach have efficacy on multidrug-
`resistant tumors with high levels of resistance attributable to
`mechanisms such as overexpression of ng? Our results show
`that, indeed, various low-dose antiangiogenic chemotherapy
`regimens are highly effective against orthotopically grown
`multidrug-resistant human breast cancers in SCID mice, but
`, usually only when the drugs are used in combination with the
`antiangiogenic, VEGFR-2-inhibiting antibody. Furthermore,
`at
`least in our study, some drugs [for example, paclitaxel
`(Taxol) and vinblastine] exhibit better therapeutic profiles
`than others
`[cisplatinum or doxorubicin (Adriamycin)]
`mainly because of their lack of cumulative toxicity.
`
`MATERIALS AND METHODS
`Cells and Culture Conditions. Two human breast can-
`
`cer cell lines, MDA-MB—231 and MDA-MB-435, and a number
`of multidrug-resistant, ng-positive variants selected from these
`lines were used in the studies: MVB9, MD22, MPAHS, and
`T01. MVB9 was selected for resistance in vitro to vinblastine
`
`sulfate and maintained in 5 ng/ml vinblastine in culture, MD22
`was selected for resistance to doxorubicin and maintained in 12
`
`mg of doxorubicin/ng, and MPAHS was isolated by transfection
`of MDA-MB-231with mdr—I gene and maintained in 10 ng/ml
`vincristine sulfate and 400 ug of G418. All three were originally
`obtained from Dr. Jeff Lemontt (Genzyme Corporation, Fram-
`ingham, MA). The derivation of these lines is similar to that
`described by Lemontt et al. (19), and expressed stable and high
`levels of cross-resistance to drugs such as colchicine, vinblas-
`tine, vincristine, and Adriamycin“. T0,], a ng-positive multi-
`drug—resistant variant of MDA-MB-435 breast carcinoma, was
`obtained from Dr. Dalia Cohen (Novartis, East Hanover, NJ)
`and was derived by progressive serial in vitro exposure of the
`cell line to increasing concentrations of Taxol; these cells ex-
`press 100-fold resistance to Taxol relative to the parental line
`(20). Finally. a cisplatinum-resistant variant called MDA-
`CDDP—S4 was selected from MDA-MB-231 by serial in viva
`drug exposure, in our laboratory. This variant does not express
`ng. To select this variant, 4 X 106 MDA-MB-23l cells were
`implanted into the mammary fat pads of 8-week-old female
`athymic nude mice, tumors were grown to ~150 mm3, and the
`mice were treated with 5 mg/kg cisplatin every second day three
`times. The tumors were then removed, adapted to culture, and
`grown in vitro for 3 weeks, reimplanted into the mammary fat
`pad of a new group of athymic nude mice, and treated in the
`same manner; the selection process was repeated three more
`times.
`
`lines were expanded as monolayer cultures by
`All cell
`serial passage on tissue culture plates (Nalge Nunc Inter-
`national, Naperville, IL) in DMEM, 5% fetal bovine serum
`(Invitrogen, Carlsbad, CA) with the addition of 22 nM (12
`ng/ml) Adriamycin (Pharmacia Upjohn, Mississauga, Canada)
`for MD22, 12.2 nM (10 ng/ml) vincristine sulfate (Sigma-
`Aldrich Chemical C0,. Canada, Oakville, Ontario, Canada) for
`MPAHS, 6 W (5 ng/ml) vinblastine sulfate (Calbiochem, La
`Jolla, Ca) for MVB9, and 0.1 |.LM Taxol (Abbott Laboratories,
`North Chicago, IL) for TO.1. HUVECs (Clonetics, San Diego,
`CA) were expanded on 1% gelatin-coated tissue culture plates in
`MCDB131 culture medium (JRH Biosciences, Lenexa, KS)
`supplemented with 5 ng/ml bFGF (R & D Systems, Minneap-
`olis, MN), 10 units/ml heparin (Wyeth-Ayerst Laboratories,
`Philadelphia, PA), 10 ng/ml epidermal growth factor (Upstate
`Biotechnology, Lake Placid, NY), and 10% fetal bovine serum.
`In Vitro Determination of Drug Sensitivity. Analysis
`of in vitro drug sensitivity was assessed on cells grown in
`monolayer as well as three-dimensional multicellular spheroids.
`For monolayer analysis, 3,000 cells in 200 H1 of growth medium
`were plated per well
`in 96-well flat-bottomed tissue culture
`plates (Nunc) and incubated at 37°C, 5% CO2 for 24 h prior to
`
`4 J. Lemontt, personal communication.
`
`Downloaded from clincancerres.aacrjoumals.org on December 6, 2017. © 2002 American Association for Cancer
`
`2
`
`

`

`Clinical Cancer Research 223
`
`Table 1 Differences in sensitivity of cancer cell lines and l-lUVECs to chemotherapeutic agents
`
`IC50 (nM)
`
`Monolayer
`
`Spheroid
`
`
`Cell population tested
`Agent
`HUVECs
`Vinblastine
`HUVECs
`Adriamycin
`HUVECs
`Cisplatinum
`HUVECs
`Taxol
`MDA-MB-231 parental tumor
`Taxol
`MD22 variant”
`Adriamycin
`MPAHS variant”
`Vinblastine
`MVB9 variant”
`Taxol
`CDDP-S4 variant”
`Cisplatinum
`
`at 24 h
`0.55
`1.65
`4
`0.4
`2
`79.4
`15.7
`27.4
`2581
`
`at 72 h
`at 24 h
`N/A
`N/A
`N/A
`N/A
`N/A
`N/A
`N/A
`N/A
`848
`3,434
`260
`1,610
`>123,301
`1,349
`1,059
`10,084
`
`27,773 2,3 80
`" All of the variants are drug-resistant sublines of MDA-MB-231 human breast carcinoma cell line.
`
`at 72 h
`N/A
`N/A
`N/A
`N/A
`4.5
`41.5
`7.1
`19
`2083
`
`initiation of treatment. For analysis in three-dimensional culture.
`multicellular tumor spheroids were formed using the liquid overlay
`technique, as described previously (21). Ninety-six-wel] round-
`bottomed tissue culture plates (Nunc) were .covered with 2% Poly-
`Hema in 100% ethanol (Aldrich Chemical C0,, Milwaukee, WI) to
`prevent attachment to the tumor cells to the plastic of the dish. The
`formation of spheroids was initiated by a gentle 10-min spin (1,000
`rpm/min) of the freshly detached cells in suspension, followed by
`24—48 h incubation at 37°C, 5% C02. To assess for the inhibitory
`activity of chemotherapeutic agents, tumor cells, grown either as
`spheroids or as a monolayer, were exposed to 0—12 uM (0—10,000
`ng/ml) of either vinblastine sulfate (Calbiochem, San Diego, CA),
`Adriamycin (Pharmacia & Upjohn Inc. Mississauga, Ontario,
`Canada), cisplatinum (Faulding Canada Inc., Vaudreuil, Que—
`bec, Canada), or Taxol (Abbot) for 24, 48, and 72 h. The cells
`were then pulsed with 2 uCi/well of [methyl-3H]thymidine
`(Amersham Life Science, Buckinghamshire, United Kingdom),
`incubated at 37°C for 6 h to allow for incorporation of [3H]
`thymidine into their DNA, frozen, and thawed; then. the DNA
`was harvested onto a filterrnat using a Titertek cell harvester.
`Radioactivity was measured on a Wallac 1205 BetaPlate scin-
`tillation counter (Wallac Oy, Turku, Finland), and proliferation
`was expressed as absolute counts of [3H]thymidine per minute
`or as percentage of untreated control. Each dose concentration
`was done in sextuplicate and repeated twice. Because the most
`significant effect was observed at 24 h for the monolayer culture
`and at 72 h for the Spheroid culture, only these points are
`presented in Table l and Fig. 1.
`In Vitro Confirmation of Drug Resistance. MDA-MB-
`231 and its drug-resistant variants, grown and expanded in
`DMEM with 5% FCS, were washed with PBS twice and incu-
`bated in fresh medium with 5 uM rhodamine chloride (Sigma-
`Aldrich Canada) with or without either 5 uM cyclosporin
`A (Novartis Pharmaceuticals, Canada Inc., Dorval, Quebec,
`Canada) or 30 uM verapamil hydrochloride (Sigma-Aldrich,
`Canada) for 30 min at 37°C. Cells were then trypsinized at room
`temperature, washed, and resuspended in PBS and analyzed for
`intracellular fluorescence by flow cytometry (excitation 488 nm,
`emission 550 nm) with an EPICS Elite V flow cytometer
`(Coulter Electronics, Ltd., Luton. United Kingdom).
`Effect of Antitubulin Agents in Combination with Anti-
`VEGFR—Z Antibody on HUVECs. Passage two of HUVECs
`was seeded on 1% gelatin-coated sterile microscope slides and
`
`allowed to grow to approximately 80% confluence before stain-
`ing. HUVECs treated with medium containing 0.5 or 1 ng/ml
`vinblastine, 25 ug/ml 1C1] (monoclonal antibody against the
`VEGFR-Z/KDR; Ref. 22), or the combination of the two for 4 h
`were stained for B-tubulin as follows. After a thorough wash
`with PBS, the monolayer was covered with ice-cold methanol
`for 10 min at —20°C, rinsed with ice cold acetone twice for 10 s,
`washed twice with PBS and rehydrated in fresh PBS for at least
`30 min prior to labeling with antibody. The anti-B-tubulin/Cy-3
`conjugate (Sigma-Aldrich Chemical Co., St. Louis, MO) was
`then diluted to 1:100 in PBS, 1% BSA and added to the slides
`for 60 min at room temperature. After rinsing the excess anti-
`body off with three 5-min washes with PBS, the tissues were
`mounted, eoverslipped, and evaluated using a Zeiss confocal
`microscope. The same procedure was done using Taxol at 0.6
`nM (data not shown).
`In Vivo Tumor Growth Assessment. Each of the cell
`
`lines was harvested on the day of injection using 1% trypsin-
`EDTA (Invitrogen), and a single-cell suspension of 2 X 106
`cells in 0.05 ml of serum-free growth medium was injected into
`the mammary fat pad of 4—6-week-old CB-17 SCID mice
`(Charles River, St.-Constant, Quebec, Canada). Approximately
`3 weeks later, when most of the tumors had grown to 300 mm3,
`the mice were randomized into groups of five animals. Two
`independent experiments were done for each xenograft, each
`totaling 30 animals in six groups. The treatment groups were as
`follows: group I (Control): 0.2 m1 of PBS (DC101 vehicle) i.p.
`every 3 days and 0.2 m1 injectable saline (vinblastine vehicle)
`i.p. every 3 days; group II: 0.4 ml of 2 mg/ml DC101 antibody
`(800 [Lg/11101156) i.p. every 3 days and 0.2 m1 of injectable saline
`i.p. every 3 days: group III: 0.5 mg/kg (1.5 mg/mz) vinblastine
`sulfate i.p. every 3 days in the case of MPAHS and MVB9,
`1
`mg/kg (3 mg/mz) Adriamycin i.p. every 3 days in the case of
`MD22.
`1 or 2 mg/kg (3 and 6 mg/mz) cisplatinum i.p. every 3
`days in the case of CDDP-S4, and 0.4 ml of PBS i.p. every 3
`days; group IV (ng inhibitor control): cyclosporin A, 10 mg/kg,
`i.p. every 3 days in the case of MPAHS, MDA-MB-231, and
`MD22, or verapamil, 20 mg/kg,
`i.p. every 3 days in case of
`MVB9; group V: chemotherapy as in group 11 combined with
`ng inhibitor in group IV; group VI: chemotherapy as in group
`11 combined with DC101. Body weight and tumor size were
`assessed weekly, and general clinical status of the animals was
`assessed every day. Perpendicular tumor diameters were meas-
`
`Downloaded from clincancerresaacrjournals.org on December 6, 2017. © 2002 American Association for Cancer
`
`3
`
`

`

`224 Chemotherapy and Anti-VEGFR-Z Antibody in Breast Cancer
`
`160m .
`150.120
`140.00
`130.00
`moo
`110.0)
`100.00
`90.00
`will)
`70.00
`600)
`50.00 '
`0.0)
`30.00
`20.00 '
`10.0)
`
`0.00
`
`01l225459 [35.97191I32815140507JNU99999EQ
`
`HUVEC’S
`
`~<
`
`~ ~
`
`'
`
`lTaxol
`IAdnann'cin
`,
`.
`IVmblastme
`ElCtspLatmum
`
`
`
`Jli‘l‘liymltllneIV.ofControl]
`
`35000]
`30000 -
`5000 ’
`20000
`
`I
`
`3
`
`E 5
`
`1m]
`
`.
`,ui-
`«F mm
`
`m I
`{I
`.
`
`“
`fl
`. L_M
`1 4
`2.9
`5.7
`".4
`229
`45.8
`91.5
`183
`
`.
`
`0
`
`0.7
`
`Drug Concentration [nM]
`
`Adfinmyc'n [nM]
`
`CDDP-S4
`
`MVB9
`..
`
`I
`
`I
`
`120—
`110
`100
`
`= w
`‘s’
`so
`O
`3 70
`.\"
`60
`
`5 50
`3; ‘°
`E 30
`n m
`10
`
`,
`
`.7
`
`_
`
`c _
`
`_
`
`——~ -
`
`-
`
`- - -
`
`'
`
`I
`
`I
`
`I
`
`01112545 9
`
`s
`
`359119 10281515 13023004999993)”
`
`['/oofControll
`
`120.00 c
`110.00 4-
`100.00 .
`9000
`80.00 I
`70.00 .7
`
`6000 I
`5000
`40.00 -
`30,00 -
`20.00
`10.00 -
`0.00
`
`jHThymidine
`
`0
`
`11
`
`215
`
`45
`
`9
`
`I ”9 71.9 M 287 579150230045999D9m
`
`VlnblutheIan
`
`Clsplafinuln [an
`
`Fig. I Chemotherapy sensitivity comparison for HUVECs, MVB9 (vinblastine-resistant variant), MD22 (Adriamycin-resistant variant), and
`CDDP-S4 (cisplatinum-resistant variant). HUVECs were grown in monolayer, whereas cancer cells were grown as both spheroids (blue bars) and
`monolayer (red bars). Comparison of the effect of different chemotherapeutic agents on the proliferative capacity of each was made. In HUVECs
`Taxol and vinblastine were inhibitory at the lowest dose concentrations, followed by Adriamycin and cisplatinum. Tumor cell lines were consistently
`more resistant in spheroid culture, but even in monolayer the drug concentrations necessary for inhibition of proliferation were much higher than those
`for endothelial cells. The results represent two independent experiments, done in sextuplicate on two different experimental days (mean : S.E.).
`
`ured using a vemier scale caliper and tumor volume estimated
`using the formula for ellipsoid: (width2 X length)/2. Growth
`curves were analyzed statistically using repeated measures
`ANOVA.
`
`For histological comparison, a separate group of animals
`was treated and sacrificed at 3 weeks of therapy, at which time
`tumors were excised and fixed in 10% (v/v) formalin or cryo-
`preserved in Tissue-Tek O.C.T. compound (Bayer Corp.,
`Elkhart, IN) until processed for histochemical analysis. Animal
`care was in accordance with institutional guidelines.
`Formalin—
`Immunohistochemistry of Tumor Tissues.
`fixed paraffin-embedded sections were cut to S-um sections and
`stained with H&E according to standard protocols.
`
`RESULTS
`
`In Vitro Drug Sensitivity Determination. Prior to un-
`dertaking any in vivo experiments, drug levels at which signif-
`icant toxicity against endothelial, but not tumor cells, might be
`
`achieved were established. A difference in relative chemother-
`
`apeutic drug sensitivities between human neuroblastoma cells
`and activated endothelial cells has been reported previously for
`vinblastine (9), and we asked whether similar differential sen-
`sitivities could be detected with other chemotherapeutic drugs
`and human breast cancer cells. To address this question, we
`subjected monolayer and spheroid cultures to increasing dose
`concentrations of one of four different drugs: Adriamycin, vin-
`blastine, cisplatinum, or Taxol. To optimize growth conditions
`and achieve comparable baseline growth levels in tumor cells
`and HUVECs, tumor cell lines were grown in DMEM with 10%
`bovine serum, but HUVECs were grown on gelatinized plates,
`and in the presence of growth factors. The untreated controls
`showed similar levels of [3H]thymidine incorporation for all
`three cell lines at the baseline drug level, eliminating the con-
`cern that the differences in proliferation may be attributable to
`inherent factors (Fig. l). The chemotherapeutic drug sensitivi-
`ties were found to be remarkably different between HUVECs
`
`Downloaded from clincancerresaacrjournalscrg on December 6, 2017. © 2002 American Association for Cancer
`
`4
`
`

`

`Clinical Cancer Research 225
`
`and the tumor cells tested. Whereas the proliferation of HU-
`VECs was inhibited by dose concentrations <0.5 nM (0.45
`ng/ml) vinblastine, <0.4 nM (0.33 ng/ml) Taxol, <1.7 nM (0.9
`ng/ml) Adriamycin, and <4 nM (1.2 ng/ml) cisplatinum, there
`was no significant growth inhibition of the parental (“drug-
`sensitive”) mammary carcinoma cell line MDA-MB-23l until
`dose concentrations greater than 3000 nM (3,000 ng/ml) were
`reached. This was further enhanced by culturing cancer cells as
`three-dimensional spheroids, which mirrored the tumor growth
`in vivo. The chemotherapeutic resistance of cancer cells grown
`in three-dimensional spheroid-like structures is,
`interestingly,
`further enhanced and enabled the cancer cell to withstand con-
`
`agent 800—>l0,000-fold
`centrations of chemotherapeutic
`higher than those of ECs. In fact, all of the selected drug-
`resistant cell lines were resistant to a degree that would preclude
`clinical relevance, because these dose concentrations are un-
`achievable in patients. For example,
`the growth rate of the
`MDA-MB-23 l-derived, cisplatinum-resistant variant CDDP—S4
`was not affected until dose concentrations of 2,600 nM (770
`ng/ml) were reached in monolayer, and 27,773 nM (8,330 ng/ml)
`in spheroid culture. This in vitro “screen” suggests that actively
`proliferating endothelial cells may be sensitive to a much lower
`range of dose concentrations of chemotherapeutic agents than
`actively proliferating tumor cells. If so,
`there is at
`least a
`theoretical possibility of substantially lowering the present clin-
`ically used MTDs of chemotherapeutic drugs to specifically
`target dividing endothelial cells present in tumors.
`The Effect of Tubulin Inhibitors and Anti-VEGFR-2
`
`Antibodies on HUVECs Grown in Monolayer Culture. De-
`spite the limitations of in vitro culture as a model, including the
`use of (large vein) HUVECS rather than microvascular endothe-
`lial cells, it provides an approach for direct observation of the
`effects of tubulin inhibitors, a monoclonal antibody against the -
`VEGFR-Z, or a combination of the two on endothelial cells. We
`observed no appreciable effect of IMC-lCll, a monoclonal
`neutralizing antibody, against the human VEGFR-2/KDR recep-
`tor, when used alone (Fig. 2). However,
`in combination with
`low-dose concentration vinblastine,
`the effects were striking.
`For example, in combination with 0.5 nM (0.5 ng/ml) vinblas-
`tine, IMC-lCll caused retraction of the cellular membrane and
`full coagulation of the cytoskeleton (Fig. 2). Interestingly, a
`potent effect was observed with the lower dose concentration,
`and doubling the dose concentration to 1 ng/ml provided no
`additional benefits (Fig. 2). This suggests that, at least in com-
`bination with a specific inhibitor of EC survival and other
`functions, lower doses of tubulin inhibitors may produce ade-
`quate anti-endothelial effects.
`Drug Resistance Testing. Before proceeding to study
`the efficacy of low-dose continuous chemotherapy in vivo
`against multidrug-resistant tumors, we verified the functional
`presence of the ng membrane pump. Using rhodamine chloride
`(a ng substrate that is easily detected by flow cytometry), we
`found a clear shift of the fluorescent population to the left (Fig.
`3), confirming the presence of ng-mediated efflux of rhoda-
`mine chloride from the MVB9, MPAHS, MD22, and T01 cells,
`but not, as expected, from the ng-negative parental cell line,
`MDA-MB-231, or the cisplatinum-resistant CDDP—S4 variant.
`As expected, we also found that this drug efflux is reliably
`
`in vitro, by ng reversal agents such as
`least
`inhibited, at
`cyclosporin A or verapamil (data not shown).
`In Vivo Tumor Growth Assessment. Building upon the
`striking in vitro differences in relative chemotherapeutic drug
`sensitivities between the tumor and endothelial cells tested, we
`evaluated the effects of a continuous low-dose regimen of each
`of these agents in inhibiting growth of orthotopic tumor xe-
`nografts in vivo. The cell lines were intentionally selected for
`their high levels of acquired resistance, e.g., up to lOO-fold, to
`negate the possibility that any antitumor effects were due to
`direct antitumor cell activity.
`The first group,
`treated with the neutralizing antibody
`directed against mouse VEGFR-Z/flk-l (DC101) shown previ-
`ously to inhibit the s.c. growth of many different kinds of human
`xenografi in immune-deficient mice (23), displayed the ex-
`pected effectiveness in inhibiting tumor growth (Fig. 4, green
`line, filled circles in all panels). Over the first weeks of therapy
`the effectiveness of DC101 was comparable with that of the
`combination treatment group, such that if this experiment had
`been terminated at 30—60 days, erroneous conclusions might
`have been drawn regarding the apparent equal efficacy of the
`two treatment groups. In most of the long-term experiments, the
`tumor volume curves diverge beyond 60—70'days and the ben-
`efit of adding the chemotherapeutic agent becomes apparent.
`Further confirmation of this delayed divergence effect was seen
`upon analysis of histological specimens at 3 weeks (see below
`and Fig. 5).
`The findings in the second treatment group (chemothera-
`peutic agent alone) showed minimal to no growth inhibition
`(Fig. 4, blue lines, filled squares), and combining the chemo-
`therapeutic agent with a ng pump inhibitor failed to improve
`this lack of an antitumor effect (Fig. 4, mauve diamonds).
`Regardless of the lack of effect of the chemotherapeutic
`agent alone,
`in most instances addition of the anti-flk-l anti-
`body, DC101, produced tumor growth suppressions over and
`above those observed with either agent alone (Fig. 4, red line,
`filled squares in all panels). Most importantly, this combination
`therapy did not result in detectable drug resistance to the treat-
`ment even after >100 days of continuous treatment. With the
`exception of those treated with Adriamycin and cisplatinum, the
`mice remained healthy throughout the course of treatment, and
`in all cases the tumor remnants, when assessed by histology,
`appeared to contain mainly fibrous scar tissue.
`Histopathological Analysis.
`In the multiple in viva pre-
`clinical trials undertaken in our laboratory, we have frequently
`observed that even through significant differences can be seen
`with tumor volume measurements,
`the true degree of tumor
`regression is not always appreciated. However controlled, the
`gross measurement of tumor volume reflects a delayed response,
`and resorption, of the tissues and not necessarily the actual
`degree of the tissue damage inflicted. We have therefore eval-
`uated representative samples of tumor tissues from all groups at
`arbitrary time points and observed, surprisingly. that significant
`differences were apparent as early as 3 weeks post initiation of
`therapy. Care was taken to include reference mammary gland
`tissue in all tumor samples (visible on the left in Fig. 5, aee), to
`avoid concerns about sampling bias. The MPAHS tumor histol-
`ogy, shown in Fig. 5, represents a typical example of the
`histopathology of all of the different tumor subtypes and their .
`
`Downloaded from clincancerres.aacrjoumals.org on December 6, 2017. © 2002 American Association for Cancer
`
`5
`
`

`

`226 Chemotherapy and Anti-VEGFR-Z Antibody in Breast Cancer
`
` Fig. 2 Sensitivity
`
`of HUVECs
`grown in monolayer to vinblastine
`alone or in combination with [MC-
`lCIl (a monoclonal antibody to hu-
`man VEGFR-Z/KDR). HUVECs
`grown on glass slides coated with 1%
`gelatin,
`and
`supplemented with
`growth factors, were treated with ei-
`ther 0.5 ng/ml vinblastine or a com-
`bination of vinblastine and 1C11(25
`ug/ml),
`a monoclonal
`antibody
`against
`the VEGFR-Z. The culture
`was then fixed and stained with anti-
`B-tubulin/Cy-3 conjugate. The mild
`polymerization of tubulin evident in
`the cultures treated with 0.5 ng/ml
`vinblastine is visibly enhanced by
`combining the agent with IMC-ICI l,
`and no further escalation of this effect
`is achieved by doubling the vinblas-
`tine dose concentration.
`
`1 nglml Vb]
`
`1 ng/ml Vbl + 25 ug/ml 1C1]
`
`respective changes. No easily appreciated differences were seen
`in mice treated with saline or vinblastine alone. Healthy looking
`cancer cells with high nuclearzcytoplasmic ratio, high mitotic:
`karyorrhetic index, and high invasive potential are the prevalent
`component in tissues from these two groups (Fig. 5, a, f—h,
`k—m). In contrast. cells with pyknotic nuclei and a high degree
`of cytoplasmic blebbing, features consistent with apoptosis,
`predominate in the combination group (Fig. 5e) and are preva-
`
`lent in the DC101 alone group (Fig. 5d). In fact, it appears that
`the degree of tissue necrosis and apoptosis is over and above
`that expected on the basis of tumor size measurements at 3
`weeks (Fig. 4, day 51 of MPAHS panel). Similar differences are
`evident for mitoticzkaryorrhetic indexes. Whereas a typical field
`in the control and vinblastine alone group may manifest many
`mitotic figures (Fig. 5, fand g), these are hard to find in samples
`treated with DC101 (Fig. 51'), and none were found in the
`
`Downloaded from clincancerresaacrjournalsorg on December 6, 2017. © 2002 American Association for Cancer
`
`6
`
`

`

`Clinical Cancer Research 227
`
`MDA231
`Parental cell line
`
`MDA231/MD22
`Adriamycin-Resistant clone
`
`Fig.3 In vitro inhibition of
`MDR of selected cancer cell
`lines. The MDA-MB-23l pa-
`rental cell
`line and its drug-
`resistant variants were grown in
`monolayer,
`incubated with 5
`rig/ml
`rhodamine chloride (a
`fluorescent ng pump sub-
`strate) for 30 min, and assessed
`for
`relative
`fluorescence by
`flow cytometry. The human
`mammary carcinoma parental
`cell lines MDA-MB-23l and its
`cisplatinum-resistant
`variant
`CDDP—S4 readily take up rho-
`damine chloride, shifting the
`curve to the right, and this up-
`take cannot be inhibited by cy-
`closporin A (a ng inhibitor). In
`contrast, its drug-resistant vari-
`ants show less rhodamine chlo-
`ride uptake, or efflux of rhoda-
`mine chloride from the cells,
`and this is, at
`least
`in vitro,
`readily inhibited by cyclosporin
`A or verapamil, suggesting a
`ng-mediated resistance.
`
`Rhod+ ClA
`
`
`
`"‘
`
`m
`
` m-u
`
`MDA23l/MPAHS
`mdr.‘ transfected clone
`
`MDA23l/MVB9
`in vitro selected Vinblastinc

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