throbber
Oncogene (1999) 18, 2241 ± 2251
`ã 1999 Stockton Press All rights reserved 0950 ± 9232/99 $12.00
`http://www.stockton-press.co.uk/onc
`
`Inhibitory e€ects of combinations of HER-2/neu antibody and
`chemotherapeutic agents used for treatment of human breast cancers
`
`Mark Pegram1, Sheree Hsu1, Gail Lewis2, Richard Pietras1, Malgorzata Beryt1, Mark Sliwkowski2,
`Daniel Coombs2, Deborah Baly2, Fairooz Kabbinavar1 and Dennis Slamon*,1
`
`1Division of Hematology-Oncology, UCLA School of Medicine, Los Angeles, California 90095, USA; 2Genentech, Inc. One DNA
`Way, South San Francisco, California, USA
`
`Previous studies have demonstrated a synergistic interac-
`tion between rhuMAb HER2 and the cytotoxic drug
`cisplatin in human breast and ovarian cancer cells. To
`de®ne the nature of the interaction between rhuMAb
`HER2 and other classes of cytotoxic drugs, we applied
`multiple drug e€ect/combination index (CI) isobologram
`analysis to a variety of chemotherapeutic drug/rhuMAb
`HER2 combinations in vitro. Synergistic interactions at
`clinically relevant drug concentrations were observed for
`rhuMAb HER2 in combination with cisplatin (CI=0.48,
`P=0.003), thiotepa (CI=0.67, P=0.0008), and etopo-
`side (CI=0.54, P=0.0003). Additive cytotoxic e€ects
`were observed with rhuMAb HER2 plus doxorubicin
`(CI=1.16, P=0.13), paclitaxel
`(CI=0.91, P=0.21),
`methotrexate
`(CI=1.15, P=0.28),
`and
`vinblastine
`(CI=1.09, P=0.26). One drug, 5-¯uorouracil, was found
`to be antagonistic with rhuMAb HER2 in vitro
`(CI=2.87, P=0.0001). In vivo drug/rhuMAb HER2
`studies were conducted with HER-2/neu-transfected,
`MCF7 human breast cancer xenografts in athymic mice.
`Combinations of rhuMAb HER2 plus cyclophosphamide,
`doxorubicin, paclitaxel, methotrexate, etoposide, and
`vinblastine in vivo resulted in a signi®cant reduction in
`xenograft volume compared to chemotherapy alone
`(P50.05). Xenografts treated with rhuMAb HER2 plus
`5-¯uorouracil were not signi®cantly di€erent from 5-
`¯uorouracil alone controls consistent with the subadditive
`e€ects observed with this combination in vitro. The
`synergistic interaction of rhuMAb HER2 with alkylating
`agents, platinum analogs and topoisomerase II inhibitors,
`as well as
`the additive
`interaction with taxanes,
`anthracyclines and some antimetabolites in HER-2/neu-
`overexpressing breast cancer cells demonstrates that these
`are rational combinations to test in human clinical trials.
`
`Keywords: HER-2/neu (c-erbB-2); chemotherapy; breast
`cancer; multiple drug e€ects analysis; synergy
`
`Introduction
`
`Overexpression of p185HER-2/neu, resulting from amplifi-
`cation of the HER-2/neu gene, is associated with poor
`clinical outcome in 25 ± 30% of carcinomas of
`the
`breast (Slamon et al., 1987), as well as in other human
`
`*Correspondence: DJ Slamon, UCLA School of Medicine,
`Department of Medicine, Division of Hematology-Oncology, 11-934
`Factor Building, Los Angeles, CA 90095, USA
`Received 13 May 1998; revised 27 October 1998; accepted 27 October
`1998
`
`malignancies (Semba et al., 1985; Slamon et al., 1989;
`Berchuck et al., 1991; Yonemura et al., 1991; Hetzel et
`al., 1992; Lukes et al., 1994; Press et al., 1994; Sa€ari
`et al., 1995). The murine monoclonal antibody 4D5 has
`speci®city
`for
`a
`juxtamembrane
`epitope
`in the
`extracellular domain (ECD) of the p185HER-2/neu protein
`(Fendly et al., 1990) and is capable of eliciting an
`antiproliferative e€ect against murine cells transformed
`by HER-2/neu as well as human malignant cell lines
`and xenografts overexpressing this oncogene (Chazin et
`al., 1992). Importantly, this growth inhibitory e€ect is
`speci®c for cells with HER-2/neu overexpression and
`does not occur with cells expressing normal amounts of
`the protein (Hudziak et al., 1989; Chazin et al., 1992).
`A recombinant, humanized form of 4D5 (rhuMAb
`HER2) has been generated by inserting the comple-
`mentary-determining regions (CDRs) of 4D5 into the
`framework of a consensus human IgG1 (Carter et al.,
`1992). When compared to murine 4D5,
`rhuMAb
`HER2
`exhibits
`a
`stronger binding
`anity
`for
`p185HER-2/neu but has similar speci®c antiproliferative
`activity against HER-2/neu-overexpressing cell
`lines
`and xenografts.
`To determine how best to use this antibody both as
`a single agent and in combination with established
`cancer therapeutics, we undertook a series of studies to
`evaluate its inhibitory e€ects in preclinical models in
`vitro and in vivo. These studies were based on a
`previous
`report of enhanced activity of cisplatin
`(CDDP) when used in combination with antibodies
`directed against the epidermal growth factor receptor
`(EGFR)
`(Aboud-Pirak et al., 1988). Initial studies
`showed that when used in combination with the drug
`CDDP, 4D5, rhuMAb HER2, as well as other anti-
`HER-2/neu antibodies, potentiate cytotoxicity of the
`chemotherapeutic by decreasing DNA repair activity
`following CDDP-induced DNA damage (Hancock et
`al., 1991; Pietras et al., 1994). This e€ect,
`termed
`receptor enhanced chemosensitivity (REC), speci®cally
`targets HER-2/neu-overexpressing cells and has no
`e€ect on cells or tissues expressing physiologic levels of
`the gene. The interaction between 4D5 and CDDP in
`inhibiting HER-2/neu-overexpressing cell lines has been
`shown to be synergistic resulting in a two-log increase
`in CDDP-induced cytotoxicity as well as pathologic
`complete remissions in experimental animals bearing
`HER-2/neu-overexpressing human breast cancer xeno-
`grafts (Pietras et al., 1994).
`Synergy, as it applies to drug-drug interactions, is
`de®ned as a combination of two or more drugs which
`achieves a therapeutic e€ect greater than that expected
`by the simple addition of the e€ects of the component
`drugs. Such synergistic interactions between drugs may
`
`1 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`2242
`
`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`improve therapeutic results in cancer treatment if the
`synergism is speci®c for tumor cells (Chou and Talalay,
`1984). Moreover, analysis of
`the nature of
`the
`interaction between two drugs (synergism, addition,
`or antagonism) may yield insight into the biochemical
`mechanisms of interaction of the drugs. For example,
`two drugs targeting the same enzyme or biochemical
`pathway may compete with one another resulting in an
`antagonistic interaction, whereas two drugs targeting
`completely independent pathways may be additive, and
`one drug which potentiates the action of another may
`result in therapeutic synergy.
`In order to characterize the e€ects of combinations
`of rhuMAb HER2 cytotoxic chemotherapeutic drugs
`commonly used in breast cancer therapy, we utilized
`the median-e€ect/combination-index
`isobologram
`method of multiple drug e€ect analysis. With this
`methodology,
`combination index (CI) values are
`calculated for di€erent dose-e€ect
`levels based on
`parameters derived from median-e€ect plots of the
`chemotherapeutic drugs alone, rhuMAb HER2 alone,
`and the combination of the two at ®xed molar ratios.
`CI values 51 indicate
`synergy, CI=1 indicates
`addition, and CI41 denotes antagonism (Chou and
`Talalay, 1984). We performed this analysis with
`rhuMAb HER2 in combination with eight drugs
`representing
`seven di€erent
`classes of
`cytotoxic
`chemotherapeutics in vitro. Assays were performed in
`for drug/rhuMAb HER2
`combinations
`at
`vitro
`clinically relevant drug/antibody concentrations using
`a cytotoxicity endpoint employing SK-BR-3 human
`breast cancer cells which contain HER-2/neu gene
`ampli®cation/overexpression. In addition, to circum-
`vent the possibility that any observed interaction might
`be unique to an individual cell
`line or to a speci®c
`method of analysis, parallel studies were conducted in
`vivo with the same rhuMAb HER2/drug combinations.
`HER-2/neu-transfected MCF7 human breast carcino-
`ma xenografts which, in contrast to SK-BR-3 cells are
`tumorigenic in athymic mice, served as the tumor
`target for the in vivo studies. Using this model we also
`investigated the e€ect of various chemotherapeutic
`drugs on the pharmacokinetics of rhuMAb HER2 in
`a subset of mice receiving either rhuMAb HER2 alone
`or rhuMAb HER-2 plus cytotoxic drug. Finally, we
`
`sought to assess the e€ect of xenograft size (i.e. tumor
`burden) on rhuMAb HER2 serum concentrations.
`
`Results
`
`Multiple drug e€ect analysis of rhuMAb HER2 in
`combination with cytotoxic chemotherapy drugs on
`SK-BR-3 breast carcinoma cells in vitro
`
`To extend the observations on anti-HER2 monoclonal
`antibodies
`in combination with CDDP, and to
`conduct a comprehensive survey of rhuMAb HER2
`in combination with other
`classes of
`cytotoxic
`chemotherapeutic drugs available for clinical use,
`rhuMAb HER2 was analysed in combination with
`seven di€erent drug classes. Representative drugs
`included:
`the anthracycline antibiotic, doxorubicin
`(DOX);
`the
`taxane drug, paclitaxel
`(TAX);
`a
`topoisomerase
`II
`inhibitor
`etoposide
`(VP-16);
`a
`platinum analog cisplatin (CDDP); a vinca alkaloid
`vinblastine (VBL);
`the alkylating agents,
`thiotepa
`(TSPA) for in vitro experiments and cyclophospha-
`mide
`(CPA)
`for
`experiments;
`and the
`in
`vivo
`antimetabolite drugs methotrexate (MTX) and 5-
`¯uorouracil (5-FU).
`curves were
`response
`In this
`analysis, dose
`constructed for each drug alone,
`rhuMAb HER2
`alone, and the combinations at ®xed molar ratios
`de®ned as
`the ratio of
`the two agents at
`their
`maximally e€ective dose. A representative example of
`the multiple drug e€ect analyses performed for all of
`the chemotherapeutic agent/rhuMAb HER2 combina-
`tions is shown for the alkylating agent TSPA (Figure
`1 and Table 1). In this analysis Fa and Fu are the
`fractions of SK-BR-3 cells a€ected or una€ected,
`respectively, by the dose (D) of either agent (drug or
`antibody). DM is the dose required to produce the
`median e€ect (analogous to the IC50), and m is the
`Hill coecient used to determine whether the dose
`e€ect
`relationships
`follow sigmoidal dose-response
`curves
`(Hill, 1913). Linear
`regression correlation
`coecients
`(r-values) of
`the median e€ect plots
`(Table 1) re¯ect that the dose-e€ect relationships for
`TSPA, rhuMAb HER2, and the combination, con-
`
`(a) Multiple drug e€ect plot of TSPA, rhuMAb HER2 and the combination where Fa = the fraction of SK-BR-3 cells
`Figure 1
`a€ected by the drugs, Fu = the fraction of cells una€ected, and D = drug dose. (b) Combination Index values for TSPA in
`combination with rhuMAb HER2 at multiple e€ect levels. CI values 51 indicate synergy
`
`2 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`2243
`
`form to the principle of mass action (in general, r-
`values 40.9 con®rm the validity of this methodology)
`(Chou and Talalay,
`1984). CI
`values
`for
`the
`combination of TSPA and rhuMAb HER2 were
`signi®cantly less
`than 1.0 across all combination
`doses
`tested (P=0.0008)
`indicating a synergistic
`interaction (Figure 1b). A summary of the data from
`the same analysis applied to each of
`the eight
`cytotoxic drug/rhuMAb HER2 combinations tested
`(Table 2) demonstrates that CDDP, TSPA, and VP-16
`(CI51;
`exhibit
`synergistic
`therapeutic
`interactions
`P50.001) with rhuMAb HER2 across a wide range
`(*0.2 ± 0.8) of Fa
`values. Additive
`interactions
`(CI=1) were observed for TAX, DOX, MTX, and
`VBL in combination with rhuMAb HER2, while only
`one drug, 5-FU, was found to exhibit an antagonistic
`(CI41; P=0.0001) interaction (Table 2).
`
`P185HER-2/neu expression and tyrosine phosphorylation
`following exposure to cytotoxic agents
`
`Previous work has demonstrated that exposure of
`several cancer cell
`lines to the anthracycline DOX
`results in an increase in expression of the EGFR and/
`ligand TGF-a (Zuckier and Tritton, 1983;
`or
`its
`Hanauske et al., 1987; Baselga et al., 1992, 1993).
`This phenomenon has been proposed to explain the
`synergistic cytotoxic e€ects of DOX used in combina-
`tion with anti-EGFR monoclonal antibodies (Baselga
`et al., 1992). To test whether p185HER-2/neu expression is
`similarly altered by DOX, protein expression levels
`were measured at various
`times
`following DOX
`exposure (Figure 2a). These studies demonstrate that
`following exposure to DOX, p185HER-2/neu expression
`levels in SK-BR-3 breast carcinoma cells are unaltered,
`unlike
`the
`reported e€ects of DOX on EGFR
`expression in A431 cells (Baselga et al., 1992). We
`next considered the possibility that cytotoxic drugs
`may impact p185HER-2/neu functional activity rather than
`expression levels. We therefore determined the e€ect of
`the various cytotoxic drugs on heregulin B-1 and 4D5-
`induced
`tyrosine
`phosphorylation
`of
`p185HER-2/neu
`
`Figure 2 (a) Expression of p185HER-2/neu
`in SK-BR-3 cells
`following exposure to DOX at the IC30 (30 nM) concentration
`for
`the
`times
`indicated.
`(b) MAb 4D5-induced tyrosine
`phosphorylation of p185HER-2/neu in SK-BR-3 cells following
`exposure to chemotherapeutic agents at the IC30 concentration at
`the indicated time points. 4D5-associated tyrosine phosphoryla-
`tion (lane 2) was observed under all of
`the chemotherapy
`conditions tested (lanes 3 ± 11) compared to control (lane 1). (c)
`Heregulin-induced p185HER-2/neu
`tyrosine phosphorylation in
`MCF7 cells following exposure to chemotherapeutic drugs at
`the IC30 concentration. These data demonstrate that p185HER-2/neu
`expression and phosphorylation state are unaltered by prior
`exposure to the chemotherapeutic agents tested
`
`Table 1 Calculated values for the Combination Index as a function of fractional inhibition of SK-BR-3 cell proliferation by a mixture of TSPA
`and rhuMAb HER2
`
`Drug
`
`TSPA
`rhuMAb HER2
`TSPA+rhuMAb HER2
`Diagnosis of combined e€ect
`
`ED30
`
`Combination Index Values
`ED50
`
`ED40
`
`ED60
`
`ED70
`
`0.52
`Synergy
`
`0.37
`Synergy
`
`0.41
`Synergy
`
`0.49
`Synergy
`
`0.60
`Synergy
`
`Parameters
`m
`
`Dm
`
`66.2 mM
`675.0 nM
`27.1 mM
`
`0.81
`0.15
`0.59
`
`r
`
`0.99
`0.96
`0.99
`
`Table 2 Mean combination index values for chemotherapeutic drug/rhuMAb HER2 combinations in vitro
`
`Drug
`
`TSPA
`CDDP
`VP-16
`DOX
`TAX
`MTX
`VBL
`5-FU
`
`rhuMAb HER2/drug
`molar ratio
`6.461075
`4.061074
`9.961074
`9.861073
`1.461071
`3.361071
`1.7
`8.861075
`P values indicate level of signi®cance compared to CI=1.0
`
`Drug Dose Range
`(mM)
`
`8.25 ± 1.066103
`6.561071 ± 1.76102
`2.661071 ± 6.86101
`2.761072 ± 6.9
`1.861073 ± 5.061071
`8.061074 ± 2.061071
`1.661074 ± 3.961072
`3.0 ± 7.656102
`
`Combination Index
`(Mean+s.e.m.)
`
`0.67+0.12
`0.56+0.15
`0.54+0.15
`1.16+0.18
`0.91+0.23
`1.36+0.17
`1.09+0.19
`2.87+0.51
`
`P value
`
`0.0008
`0.001
`0.0003
`0.13
`0.21
`0.21
`0.26
`0.0001
`
`Interaction
`
`Synergy
`Synergy
`Synergy
`Addition
`Addition
`Addition
`Addition
`Antagonism
`
`3 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`2244
`
`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`(Yarden, 1990; Holmes et al., 1992). MCF7 or SK-BR-
`3 breast carcinoma cells were treated with cytotoxic
`drugs, then allowed to incubate with heregulin (10 nM),
`or 4D5 (12.5 mg/ml). Protein lysates were then analysed
`by anti-phosphotyrosine immunoblot. These studies
`demonstrate an increase in
`p185HER-2/neu
`tyrosine
`phosphorylation following incubation with 4D5 com-
`pared to a non-speci®c isotype control antibody
`(Figure 2b, lanes 1 and 2). Prior exposure of the cells
`to the three drugs which were found to be synergistic
`with anti-HER-2/neu antibody (CDDP, TSPA, and
`VP-16) had no e€ect on 4D5-induced p185 tyrosine
`phosphorylation (Figure 2b, lanes 3 ± 7 and lanes 9 and
`10). Similarly, neither DOX which is additive, nor 5-
`FU which is antagonistic, had e€ects on 4D5-induced
`p185 tyrosine phosphorylation (Figure 2b, lanes 8 and
`11). In addition, when heregulin B-1 is used to activate
`p185HER-2/neu kinase, preincubation of MCF7 breast
`carcinoma cells with CDDP or DOX had no e€ect
`on heregulin-induced p185HER-2/neu tyrosine phosphoryla-
`tion (Figure 2c). Preincubation of MCF7 cells with
`TSPA, VP-16, TAX, MTX, VBL, or 5-FU likewise had
`no e€ect on heregulin-induced p185HER-2/neu
`tyrosine
`phosphorylation (data not shown). Taken together
`
`these data demonstrate that none of the synergistic,
`additive, or antagonistic e€ects of chemotherapeutic
`drugs with anti-HER-2/neu antibody can be explained
`on the basis of either chemotherapy-induced alteration
`of p185HER-2/neu protein expression levels or
`its
`phosphorylation.
`
`Anti-HER-2/neu antibodies alter cell cycle distribution
`of HER-2/neu-overexpressing human breast cancer cells
`
`The cytotoxic e€ects of antimetabolite drugs are cell
`cycle dependent
`(Tannock, 1978). To identify a
`possible mechanism for the antagonism of 5-FU with
`rhuMAb HER2 we investigated the e€ects of murine
`4D5 and rhuMAb HER2 on cell cycle distribution of
`exponentially growing SK-BR-3 and MCF7 cells in
`vitro (Figures 3 and 4). Both the murine 4D5 and
`rhuMAb HER2 antibodies reduce the percentage of
`cells undergoing S phase as well as
`increase the
`percentage of cells in G0/G1, and these e€ects are
`dose-dependent with the maximal antiproliferative
`activity occurring at antibody concentrations between
`1 and 10 mg/ml (Figure 4). There was no signi®cant
`di€erence in the magnitude of decrease in S phase
`
`Figure 3 DNA ¯uorescence ¯ow cytometry histograms of propidium iodide-stained nuclei obtained from MCF7 (a ± c) and SK-
`BR-3 (d ± f) breast carcinoma cells following treatment with control antibody 6E10, murine anti-p185HER-2/neu antibody 4D5, or
`humanized anti-p185HER-2/neu antibody (rhuMAb HER2) at a dose of 1 mg/ml for 72 h. These data demonstrate a signi®cant
`reduction in the fraction of breast carcinoma cells undergoing S phase following treatment with anti-HER-2 antibodies 4D5 and
`rhuMAb HER2. This e€ect is speci®c for cells with HER-2/neu-overexpression (SK-BR-3 cells)
`
`4 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`2245
`
`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`athymic mice. All of the doses, routes of administra-
`tion, and dose intervals for the various cytotoxic drugs
`and rhuMAb HER2 were based on independent dose
`®nding experiments for this speci®c strain, age, weight,
`and sex of athymic mouse. The cytotoxic drug doses
`used were at or near the maximum tolerated doses
`previously reported in the literature (Giovanella et al.,
`1977; Boven and Winograd, 1991).
`For the alkylating agent cyclophosphamide CPA,
`combination with rhuMAb HER2 resulted in a
`signi®cant reduction (P50.05)
`in day 21 xenograft
`volume compared to either agent alone (Figure 5a).
`The combination of the anthracycline antibiotic DOX
`plus rhuMAb HER2 also signi®cantly reduced MCF7/
`HER-2 xenograft volume compared to either single
`agent alone (Figure 5b). The combination of
`the
`taxane compound TAX plus rhuMAb HER2, which
`demonstrated an additive interaction in vitro, resulted
`in a signi®cant reduction in day 20 xenograft volume
`compared to treatment with TAX alone (Figure 5c).
`However,
`the di€erence between rhuMAb HER2
`alone and rhuMAb HER2 plus TAX did not reach
`statistical
`signi®cance. This
`is
`likely due
`to the
`relatively small sample size in each group and the
`fact that the dose of rhuMAb HER2 in this particular
`analysis (10 mg/kg I.P. twice weekly) yielded a marked
`reduction in xenograft growth even when used as a
`single agent.
`The following four rhuMAb HER2/drug combina-
`tions were studied in a single in vivo experiment. For
`this experiment, a `rational dose' (RD) or rhuMAb
`HER2 was
`chosen as new information became
`available based on comparative pharmacokinetic
`studies from both humans and athymic mice. RD is
`the dose of a given drug which can reproduce a serum
`level in experimental animals similar to that observed
`in human subjects (Inaba et al., 1988). The RD for
`rhuMAb HER2 resulted in a lower
`cumulative
`rhuMAb HER2 dose (16 mg/kg vs 30 ± 50 mg/kg)
`during the 21 day observation period for
`this
`experiment compared to the three in vivo studies
`reported above. With this approach, a signi®cant
`reduction in day 21 xenograft volume was observed
`for the topoisomerase II inhibitor VP-16 when used in
`combination with rhuMAb HER2 compared to either
`agent alone (Figure 6a). The combination of
`the
`microtubule inhibitor VBL with rhuMAb HER2 also
`signi®cantly reduced MCF7/HER-2 xenograft volume
`compared to treatment with VBL alone or single agent
`rhuMAb HER2 (Figure 6b). For the antimetabolite
`class of cytotoxic chemotherapeutics, two drugs with
`clinical activity against breast cancer were chosen for
`combination studies. Treatment with MTX, which
`targets dihydrofolate reductase, plus rhuMAb HER2
`resulted in a signi®cant reduction in day 21 MCF7/
`HER-2 xenograft volume when compared to either
`MTX alone or rhuMAb HER2 alone (Figure 6c).
`Finally, the antimetabolite drug 5-FU, which targets
`thymidylate synthetase, and which was found to be
`antagonistic when combined with rhuMAb HER2 in
`vitro, did not yield a signi®cant reduction in xenograft
`volume when compared to 5-FU alone in vivo (Figure
`6d). Although the combination of rhuMAb HER2 plus
`5-FU was superior to rhuMAb HER2 alone in this
`(P50.05),
`experiment
`the 5-FU dose used had
`sucient anti-tumor ecacy as a single agent such
`
`Figure 4 E€ect of anti-p185HER-2/neu MAb dose on cell cycle
`distribution of breast cells without (a) and with (b) HER-2/neu
`overexpression
`
`fraction of SK-BR-3 cells comparing 4D5 and rhuMAb
`HER2 indicating the humanization of
`the murine
`antibody did not adversely impact its antiproliferative
`activity. The lack of any e€ect on cell cycle distribution
`of MCF7 cells demonstrates the speci®city of these
`antibodies for cells with HER-2/neu overexpression.
`These data suggest that a decrease in the percentage of
`SK-BR-3 cells in S phase may result in a decreased
`sensitivity to 5-FU. An antagonistic interaction for the
`combination of rhuMAb HER2 with the antimetabo-
`lite MTX was not observed. The lack of antagonism
`between MTX and rhuMAb HER2 in vitro may be due
`to the longer incubation period required for MTX
`(120 vs 72 h) to elicit cytotoxicity in the assay used for
`the multiple drug e€ect analysis, and the fact that
`MTX exerts cytotoxic e€ects in other phases of the cell
`cycle in addition to S phase (Buick, 1994).
`
`E€ect of rhuMAb HER2 in combination with multiple
`chemotherapeutic drugs on growth of HER-2/neu-
`transfected MCF7 breast xenografts in vivo
`
`To further evaluate the potential therapeutic e€ects of
`rhuMAb HER2/chemotherapy combinations and to
`extend our observations beyond a single cell line and
`preclinical model, a series of
`in vivo studies were
`performed using human breast cancer xenografts in
`
`5 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`2246
`
`Figure 5 Combination treatment of MCF7/HER-2 breast
`carcinoma xenografts in athymic mice with rhuMAb HER2 plus
`chemotherapeutic agents CPA (a), DOX (b), and TAX (c). For
`each drug, signi®cant reduction in xenograft volume was observed
`for rhuMAb HER2/drug combinations compared to drug alone
`controls (P50.05)
`
`that it was not possible to resolve potential di€erences
`between 5-FU alone and the combination with the
`sample sizes chosen (10 mice/group).
`
`Correlation between rhuMAb HER-2 serum
`concentration and MCF7/HER-2 xenograft volume
`
`To investigate the relationship between rhuMAb HER2
`concentration and xenograft
`size,
`trough rhuMAb
`HER2 serum concentration was measured in a subset
`of mice on day 64 following extended rhuMAb HER2
`treatment at the RD (8 mg/kg loading dose and eight
`weekly i.p.
`injections of 4 mg/kg)
`(Figure 7). A
`signi®cant inverse correlation (Spearman Rank Corre-
`
`lation rho=70.543; P=0.0067) between trough
`rhuMAb HER2 concentration and xenograft volume
`was observed,
`suggesting that
`the MCF7/HER-2
`xenograft
`size signi®cantly a€ects
`rhuMAb HER2
`pharmacology. Furthermore, this e€ect is independent
`of serum shed HER-2/neu ECD concentration as this
`molecule was undetectable in any of the murine serum
`samples analysed (data not shown).
`To determine if chemotherapeutic drugs have an
`e€ect on rhuMAb HER2 pharmacology, day 64 trough
`serum rhuMAb HER2 concentrations were analysed by
`treatment group in a subset of mice used for the in vivo
`studies. Controlling for xenograft size, there was no
`signi®cant di€erence
`in rhuMAb HER2
`trough
`concentration between any of the treatment groups in
`Figure 7 (data not shown).
`
`Discussion
`
`The protein products of transforming oncogenes have
`been a target for anti-cancer drug development since
`the initial discovery of these genes, however there is
`only one currently approved drug speci®cally targeting
`these proteins in clinical use. Identi®cation of
`the
`HER-2/neu gene alteration and its association with
`aggressive forms of human breast cancer has resulted
`in its successful therapeutic targeting (Slamon et al.,
`1987, 1989; Baselga et al., 1996; Pegram et al., 1998).
`The interaction of anti-HER-2/neu antibodies with
`p185HER-2/neu results in receptor tyrosine phosphoryla-
`tion.
`(Yarden, 1990), downregulation of
`receptor
`expression (Park et al., 1992),
`internalization of the
`antibody-receptor complex (Maier et al., 1991), and a
`decrease in the association of p185HER-2/neu with its
`heterodimeric partners HER-3 and/or HER-4 (Reese et
`al., 1996; Klapper et al., 1997). These events are
`accompanied by a number of biological
`e€ects
`including most importantly a decrease in cell prolifera-
`tion (Rodriguez et al., 1993), alteration of cell cycle
`distribution, and a marked decrease in the ability of
`the cell to excise and repair DNA damage induced by
`platinum analogs (Pietras et al., 1994; Arteaga et al.,
`1994). This enhanced cytotoxic activity is speci®c for
`malignant cell
`lines or xenografts with HER-2/neu
`receptor overexpression since anti-HER-2/neu antibo-
`dies have no such e€ect on cell lines with physiologic
`HER-2/neu expression levels (Hancock et al., 1991;
`Pietras
`al.,
`1994).
`Interaction
`between
`the
`et
`p185HER-2/neu
`signaling pathway
`and CDDP-DNA
`repair mechanisms has been con®rmed using tyrosine
`kinase
`inhibitors
`to
`block
`p185HER-2/neu
`receptor
`phosphorylation which inhibits
`antibody
`induced
`attenuation of
`repair of platinum-DNA adducts
`(Arteaga et al., 1994). Moreover, reversal of CDDP
`resistance is possible through transfection and over-
`expression of HER-2/neu cDNA followed by incuba-
`tion with anti-HER-2/neu antibody (Pietras et al.,
`1994). As a result of this work, studies demonstrating
`the clinical ecacy of the combination of an anti-
`HER-2/neu antibody plus CDDP were conducted in
`breast
`cancer patients with HER-2-overexpressing
`breast cancers who previously exhibited clinical drug
`resistance to cytotoxic therapy (Pegram et al., 1998).
`To test whether this receptor enhanced chemosensi-
`tivity mechanism could be observed with other classes
`
`6 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`2247
`
`Figure 6 Treatment of MCF7/HER2 xenografts with rhuMAb HER2 in combination with VP-16 (a), VBL (b), MTX (c), and 5-
`FU (d). Combination drug/rhuMAb HER2 treatment resulted in a signi®cant reduction in xenograft volume compared to drug
`alone, or rhuMAb HER2 alone, controls (P50.05) for each of the drugs indicated with the exception of 5-FU
`
`concentrations achieved in humans (Pegram et al.,
`1997, 1998). Data from the multiple drug e€ect
`analysis methodology are useful, not only in establish-
`ing hypotheses as to the mechanism of action of multi-
`drug combinations, but can also provide insight as to
`how two drugs should be administered temporally to
`gain the maximum therapeutic e€ect. For example, two
`drugs which are synergistic might best be administered
`together whereas two antagonistic drugs would be most
`e€ective if given sequentially. Data from the current
`study demonstrate
`that
`the platinum compound
`CDDP, the alkylating agent TSPA, and the topoi-
`somerase
`II
`inhibitor VP-16
`are
`synergistic
`in
`combination with rhuMAb HER2 in treating HER-2/
`neu-overexpressing SK-BR-3 breast carcinoma cells in
`vitro. These results
`suggest
`the possibility of an
`interaction between the HER-2/neu signaling pathway
`and intracellular DNA repair mechanisms involved
`with repair of DNA damage resulting from these
`speci®c DNA damaging
`agents. Other potential
`mechanisms might also explain the synergy observed
`between rhuMAb HER2 and these agents,
`including
`the possibility that rhuMAb HER2 could impact the
`cellular pharmacology of the drugs resulting in an
`increase in their cytotoxic activity. An argument
`against this hypothesis is the fact that the anti-HER-
`2/neu antibody has no e€ect on the net cellular
`incorporation of
`14C-labeled carboplatin (Pietras et
`al., 1994) or [14C]-doxorubicin in target cells (Pegram et
`al., 1992). Another possible mechanism for
`the
`observed synergy with rhuMAb HER2 is an e€ect of
`cytotoxic drugs on the expression level and/or kinase
`
`Figure 7 Inverse relationship between MCF7/HER-2 xenograft
`volume and trough rhuMAb HER2 concentration in murine
`serum (Spearman Rank Correlation rho=70.543; P=0.0067).
`These data suggest that binding of rhuMAb HER2 to HER-2/
`neu-overexpressing xenografts reduces serum rhuMAb HER2
`concentrations
`
`of cytotoxic chemotherapeutic agents, we performed a
`series of studies evaluating combinations of cytotoxic
`agents with rhuMAb HER2 testing seven classes of
`chemotherapeutics in common clinical use. All con-
`centration ranges of cytotoxic drugs and rhuMAb
`HER2 tested in these studies were conducted at serum
`
`7 of 11
`
`Celltrion, Inc., Exhibit 1017
`
`

`

`2248
`
`Anti-HER-2/neu antibody and chemotherapy combinations
`M Pegram et al
`
`activity of p185HER-2/neu. An analogous mechanism has
`been postulated for the EGFR where low doses of
`DOX appear to increase receptor expression enhancing
`the antiproliferative activity of anti-EGFR antibody
`(Zuckier and Tritton, 1983; Hanauske et al., 1987;
`Baselga
`al.,
`1992,
`1993). The
`current data
`et
`demonstrate no change in p185HER-2/neu expression levels
`or in HER-2/neu receptor tyrosine phosphorylation
`following exposure to cytotoxic drugs, suggesting that
`unlike the EGFR, this mechanism is not operative for
`the HER-2/neu receptor.
`Most of
`the rhuMAb HER2/drug combinations
`evaluated in this study demonstrate additive rather
`than synergistic
`interactions
`suggesting
`that
`the
`majority of observed antiproliferative
`e€ects of
`rhuMAb HER2 plus cytotoxic drugs are due to a
`mechanism of action involving each agent acting
`independently.
`It
`is
`interesting to note that
`the
`mechanisms of
`action of many of
`the drugs
`demonstrating additivity do not involve direct DNA
`damage, but rather disruption of microtubule poly-
`merization/depolymerization (taxanes and vinca alka-
`loids) or inhibition of DNA synthesis (antimetabolites).
`This observation is consistent with the hypothesis that
`the synergy between cytotoxic drugs and rhuMAb
`HER2 involves an interaction between the HER-2/neu
`signaling and DNA repair pathways. Subsequent to
`our initial demonstration of the additive e€ects of
`rhuMAb HER2 with TAX (Hsu et al., 1997), studies
`con®rming this additive interaction were published
`(Baselga et al., 1998). The antimetabolite drug 5-FU is
`the only drug which demonstrated antagonism when
`used in combination with rhuMAb HER2 in vitro. We
`have not yet de®ned the mechanism of this interaction,
`but it may be the result of alterations in cell cycle
`distribution caused by rhuMAb HER2 as seen in the
`current data. It could also be the result of intracellular
`pharmacological e€ects, alteration of
`the enzymatic
`activity responsible for conversion of 5-FU to 5-
`¯uorodeoxyuridine monophosphate, or an impact on
`the level of the target enzyme thymidylate synthetase.
`Further work is needed to explore these possibilities.
`The multiple drug e€ect model is not easily applied
`to analysis of in vivo studies since such analyses, with
`the number of drugs reported in this study, would
`require at least 600 athymic mice (assuming ®ve mice
`per group, ®ve data points for each dose response
`curve, and three dose response curves ± for each drug
`alone, and in combination with rhuMAb HER2).
`Consequently we used a more conventional approach
`for analysis of
`the in vivo data (i.e. single factor
`ANOVA at ®xed time points following treatment of
`mice with optimal drug or rhuMAb HER2 doses). The
`cytotoxic drug doses chosen for these experiments are
`at or near the MTD reported in the literature for each
`of the cytotoxic drugs. The rhuMAb HER2 doses and
`schedules were designed to achieve target
`serum
`concentrations of 510 ± 20 mg/ml
`in mice bearing
`HER-2/neu-overexpressing xenografts of 50 ± 500 mm3
`in size. This antibody concentration is associated with
`our previously published maximal antiproliferative
`e€ect in vitro (De Santes et al., 1992). With this in
`vivo approach, we demonstrated signi®cantly superior
`anti-tumor ecacy of rhuMAb HER2 in combination
`with CPA, DOX, MTX, TAX, VP-16, and VBL when
`compared to e€ects of each chemotherapeutic drug
`
`alone. These results are consistent with the in vitro data
`which demonstrate that
`rhuMAb HER2 is either
`additive or synergistic with each of these drugs. For
`the drug 5-FU, which was antagonistic w

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