throbber

`
`Priority Report
`
`A Model of Postsurgical Advanced Metastatic Breast Cancer
`
`More Accurately Replicates the Clinical Efficacy of
`
`Antiangiogenic Drugs
`
`Eric Guerin, Shah Man, Ping Xu, and Robert S. Kerbel
`
`Abstract
`
`The failure rate of randomized phase HI oncology clinical trials is extremely high, even when preceded by
`encouraging preclinical studies and phase II trial results of the same therapy. Thus, there is considerable effort
`being made to improve the predictive clinical potential of preclinical models, in addition to improving phase II
`trial design. With respect to the former, preclinical models have historically relied heavily on treatment ofprimary
`spontaneous or transplanted tumors rather than the more common and therapeutically challenging clinical trial
`circumstance ofadvanced metastatic disease. Here, we show that the oral antiangiogenic tyrosine kinase inhibitor
`(TKI), sunitinib, which failed to meet primary or secondary survival endpoints in 4 separate phase HI metastatic
`breast cancer (MBC) trials, either alone or with chemotherapy, similarly failed to show monotherapy or
`combination chemotherapy efficacy in a model of postsurgical advanced MBC using a metastatic variant of
`the MDA—MB—231 triple-negative human breast cancer. In contrast, the drug was effective when used to treat
`established orthotopic primary tumors. Similar results were obtained with pazopanib monotherapy, another
`antiangiogenic oral TKI. However, when an antibody targeting the VEGF pathway (DClOl) was tested, it showed a
`trend in modestly improving the efficacy of paclitaxel therapy, thus resembling to a degree prior phase HI clinical
`results of bevacizumab plus paclitaxel in MBC. Our results suggest the potential value of treating postsurgical
`advanced metastatic disease as a possible strategy to improve preclinical models for predicting outcomes in
`patients with metastatic disease. Cancer Res; 73(9); 2743—8. ©2013 AACR.
`
`Introduction
`
`An enduring problem in oncology experimental therapeu-
`tics has been the limited value ofmodels involving treatment of
`tumor-bearing mice to consistently predict outcomes later
`assessed in clinical trials, particularly at the randomized phase
`III level (1—4). A common scenario observed is positive and
`sometimes even remarkable preclinical activity, which is then
`followed by complete failure in the clinic (1—4). Such failures
`add substantially to the cost of approved agents as well as
`exposing patients with cancer enrolled in such trials to inef—
`fective therapies. As a result, there is considerable effort to
`identify potential causes for this discrepancy and develop
`significantly improved preclinical models (1—4) such as genet—
`ically engineered mouse models (GEMM) of cancer and
`patient-derived xenografts (PDX) as opposed to the historically
`
`more common use of transplantation of established cultured
`tumor cell lines grown as solid primary tumors.
`Although many factors have been proposed for the discrep-
`ant therapeutic outcomes observed between preclinical and
`clinical studies (1—4), one factor, which has received scant
`attention, is the failure to duplicate in mice treatment of
`advanced visceral metastatic disease (5, 6). Most phase I and
`II solid tumor clinical trials and the majority of phase III trials
`involve patients with such disease. In many or most cases the
`primary tumor has been surgically resected. The failure rate is
`extremely high in phase HI metastatic therapy trials (7) and
`when therapies succeed, the benefits in survival are frequently
`incremental (8). Therefore we have developed several models
`of postsurgical advanced metastatic disease using established
`human tumor cell lines grown in immunodeficient mice to
`mimic the more challenging circumstance of treating patients
`with metastatic disease (5). In most cases, the cell lines used are
`variants previously selected in viva for aggressive spontaneous
`metastatic spread after the primary orthotopic tumor has been
`surgically resected (5). One such variant, called LM2-4, was
`serially selected in Viva from the commonly used MDA-MB-231
`triple-negative human breast cancer cell line (9).
`Here, we report the use of the aforementioned postsurgical
`model of LM2—4 to evaluate the impact of several antiangio—
`genic drugs, used alone or in combination with paclitaxel
`chemotherapy, and compare the results obtained with con-
`ventional treatment of established primary tumors. One of the
`Genentech 2081
`
`Authors' Affiliations: Department of Medical Biophysics, Biological
`Sciences Platform, Sunnybrook Research Institute, University of Toronto,
`Toronto, Ontario, Canada
`Note: R.S. Kerbel holds a Tier 1 Canada Research Chair.
`
`Corresponding Author: RS. Kerbel, Biological Sciences Platform, Sun-
`nybrook Research institute, Dept. of Medical Biophysics, University of
`Toronto, 8-217, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada.
`Phone: 416-480-5711; Fax: 416—480-5884.
`
`doi: 10.1158/0008-5472.CAN-12-4183
`©2013 American Association for Cancer Research.
`
`www.aacrjourna|s.org
`
`H
`
`ir v.
`
`nnth
`
`|PR2017-00737
`
`/44(Q AmericanAssociazimzfar CancerResearch
`
`2743
`
`Genentech 2081
`Hospira v. Genentech
`IPR2017-00737
`
`1
`
`

`

`Guerin et a1.
`
`drugs we tested is sunitinib (Sutent), an oral tyrosine kinase
`inhibitor (TKI), which targets VEGF receptors (VEGFR) and
`platelet—derived growth factor receptors, among several others
`(10). Based partly on very encouraging preclinical results in 3
`different established primary breast cancer models (a trans-
`genic model, a chemically-induced rat model, and a human
`tumor xenograft model; ref. 10) and a bone colonization
`experiment (10), sunitinib was subsequently evaluated in
`patients with metastatic breast cancer (MBC); 4 independent
`phase III trials were undertaken (1 1- 15), 3 in combination with
`chemotherapy (paclitaxel, or docetaxel, or capecitabine). All 4
`trials failed to meet efficacy endpoints of survival (1 1-15). This
`stands in contrast to a phase HI trial involving the anti-VEGF
`antibody, bevacizumab (Avastin), when used with chemo-
`therapy, for example, paclitaxel, which provided a clinical
`benefit, at least in progression—free survival (PFS), though not
`in overall survival (OS; ref. 16). We also tested another anti-
`angiogenic TKI, pazopanib and a monoclonal antibody that
`targets the mouse VEGFR—2 (DC101). Sunitinib and DC101
`were also evaluated with concurrent paclitaxel chemotherapy.
`The purpose of these studies was to further validate the
`preclinical strategy of using postsurgical models of advanced
`metastatic disease to predict clinical outcomes involving
`treatment of patients with metastatic disease by addressing
`the following questions and comparing the results with prior
`phase III trial outcomes: (i) is it the case that antiangiogenic
`drug monotherapy has reduced or no therapeutic benefit when
`treating mice with advanced metastatic disease in contrast to
`established primary tumors? (ii) What is the impact on out-
`comes when chemotherapy is used in combination with the
`antiangiogenic agent? And, (iii) is there a difference in out-
`comes when using TKIs versus antibodies in combination with
`chemotherapy?
`
`Materials and Methods
`
`Female CB-17 severe combined immunodeficient (SCID)
`mice were purchased from Charles River, and female YFP
`SCID mice (17) were bred in house from breeding pairs
`generously provided by Dr. Janusz Rak (McGill University,
`Montreal). Mice at 6 to 8 weeks of age were used. MDA-MB
`231/LM2.4 is a variant cell line of MDA-MB 231 selected in
`
`Viva for aggressive spontaneous metastatic spread from
`established but resected primary tumors and was grown in
`cell culture as previously described (9). Cell line authenti-
`cation was carried out by genotyping using Illumina mouse
`linkage panel and confirmed to be human in origin. Routine
`mycoplasma screening is carried out in—house using com-
`mercial kits, which confirmed the cell line is mycoplasma
`free. Mammary fat pad injection (2 x 106 cells) was carried
`out as previously described (9). Weekly caliper measure-
`ments were carried out to determine tumor growth and
`tumor volume was calculated using the formula azb/Z where
`a is the width and b is the length. Treatment of primary
`tumors was initiated when average volume was approxi—
`mately 100 to 150 mms, that is, 12 to 15 days after cell
`injection. Surgical resection of the primary tumors was
`carried out when the average tumor size was 400 1111113.
`
`All mice were randomized just before initiation oftreatment
`Antiangiogenic drugs were generously provided by the man-
`ufacturers, namely, sunitinib (Pfizer), pazopanib (GSK), and
`DC101, the monoclonal antibody targeting mouse VEGFR—Z
`(ImClone/Eli Lilly). All drugs were prepared according to
`manufacturer's specifications. Paclitaxel was dispensed by
`Sunnybrook Pharmacy Department, Odette Cancer Center
`(Toronto, Ontario, Canada) at 60 mg/mL and further diluted
`with normal saline to the appropriate concentration. Control
`mice received either vehicle and/or normal saline as appro-
`priate. Sunitinib was administered by gavage at 60 mg/kg dose
`daily for the first 14 days followed by 5 days daily with 2 days
`break thereafter to reduce toxicity as measured by weight loss.
`Pazopanib 150 mg/kg was administered by gavage daily with-
`out interruption. Paclitaxel was administered intraperitoneally
`(i.p.) at 50 mg/kg every 3 weeks in the studies, which included
`combination with DC101 but the dose and schedule was
`
`changed to 30 mg/kg once every 2 weeks in studies, which
`involved combination with sunitinib or pazopanib to minimize
`toxicity observed in SCID mice (18).
`
`Results and Discussion
`
`We first tested sunitinib on the grth of primary estab-
`lished orthotopic (mammary fat pad) tumors. Cells from the
`established variant of MDA-MB—231 called LM2.4, which was
`selected in viva for aggressive spontaneous metastatic spread
`after surgical resection of the primary tumor (9) were injected
`into the mammary fat pad of 6- to 8-week-old female SCID
`mice, as described previously (9) in Materials and Methods. We
`did not use a luciferase-tagged clone of LM2.4 (19) because we
`have found that these cells have a reduced ability for sponta-
`neous metastasis (unpublished observations), the basis of
`which is currently unknown. When the primary tumors
`reached a volume of approximately 100 to 150 m3, sunitinib
`was administered daily by gavage at a preclinically effective
`dose of 60 mg/kg, and the treatment continued until end point.
`As shown in Fig. 1A, a robust growth delay was observed,
`similar to Abrams and colleagues using another human breast
`cancer xenograft model (called MX—I; ref. 10). In the case of
`Abrams and colleagues, this was paralleled by an increase in
`OS, the extent of which was shown to be further enhanced by
`combination with chemotherapy, for example, docetaxel (10).
`In our case, the mice were sacrificed in this preliminary
`experiment at an earlier defined endpoint, namely, when the
`control group tumors reached an average size ofapproximately
`500 mm3 (Fig. 1A). However, in another experiment, shown
`in Fig. 1B where the primary tumor was surgically resected
`(at day 20) and the same therapy initiated 3 weeks later (i.e.,
`when the mice have established visceral metastatic disease
`
`based on numerous previous studies, e.g., ref. 9 in addition to
`reproducibility and lack of variability of the short median
`survival times), no impact of the same treatment on survival
`was observed. This pattern of discrepancy in outcomes is not
`specific for sunitinib as we observed a similar pattern using
`pazopanib (as shown in Fig. 1C and D) where primary tumors
`in control mice were allowed to grow in this case to endpoint of
`1,700 mm3 and the therapy maintained until endpoint.
`
`2744
`
`
`Cancer Research
`
`Cancer Res; 73(9) May 1, 2013
`
`2
`
`

`

`
`
`
`
`
`Days
`
`Initiation
`
`of therapy
`
`Vehicle control
`
`Pazopanib
`
`_._ Vehicle control
`-I— Pazopanib 150 mg/kg
`
`
`
`D 110
`100
`90
`
`so
`70
`so
`so -----------
`40
`30
`20
`
`
`
`Percentsurvival
`
`10
`O
`
`
`
`50
`60
`70
`80
`
`Days after cell implantation
`
`
`
`
`
`Differential Drug Efficacy on Primary versus Metastatic Tumors
`
`700
`
`600
`
`500
`
`400
`300
`
`200
`100
`
`g:
`E
`v
`g

`5
`E
`:3
`
`Vehicle control
`
`Sunitinib 60 mg/kg
`
`B 110
`
`100
`_ 9°
`E 80
`E
`70
`a 60
`0
`g
`50
`._
`40
`g:
`30
`20
`
`
`
`
`— Vehicle control
`— Sunitinib 60 mg/kg
`(therapy initiated at day41)
`
`
`
`
`
`
`
`40
`
`50
`
`60
`
`70
`
`80
`
`90
`
`Days after implantation
`
`.
`
`
`
`C
`
`2000
`
`1,750
`
`isinmooo
`1.000
`750
`
`500
`
`250-
`
`
`
`
`
`Tumorvolume(mm3)
`
`5
`
`10
`
`35
`30
`25
`20
`15
`Days after cell implantation
`
`40
`
`45
`
`50
`
`Treatment
`initiated on day 14
`
`
`Figure 1. Differential impact of sunitinib or pazopanib monotherapy on primary breast tumor growth versus postsurgical advanced metastatic breast
`cancer. Sunitinib administered daily inhibits primary tumor growth (A), but has no survival benefit when treating advanced metastatic disease (B). The bottom
`shows similar results with pazopanib administered daily (C and D). For A and B, 2 x 106 MDA—MB 231/LM2-4 cells were implanted into the mammary fat
`pad of 6 08-17 SCID female mice; in the primary tumor study. treatment with sunitinib was initiated 12 days later when average tumor size was 100 mma;
`in the advanced metastasis therapy study, primary tumors were surgically resected 20 days after cell injection when average size was approximately 400 mm3
`and sunitinib treatment was initiated 21 days later. For C and D, 2 x 106 MDA-MB 231/LM2-4 cells were implanted in the primary tumor study (C),
`pazopanib treatment was initiated 14 days later when average tumorsize was 150 mm3; in the advanced metastasis therapy study (D), the primary tumors were
`surgically resected 20 days after cell injection when average size was approximately 400 mm3 and treatment was initiated 19 days later.
`
`to
`We next assessed the -impact of adding paclitaxel
`sunitinib. Shown in Fig. 2A and B is the impact on survival
`of mice with advanced metastatic disease when maximum
`
`tolerated dose (MTD) paclitaxel was combined with suniti-
`nib. Two independent experiments were carried out. If
`anything,
`there was a trend to reduced survival
`in the
`combination treatment group compared with the mice
`receiving paclitaxel alone, an observation that is consistent
`with a phase III breast cancer trial of sunitinib plus another
`taxane, docetaxel (12), where no added benefit in PPS was
`observed and OS was slightly reduced in the combination
`treatment group (12). We would note that in the 2 experi—
`ments shown in Fig. 2A and B, there was a difference in the
`therapeutic impact of paclitaxel alone. In one experiment
`(shown in Fig. 2A) the paclitaxel monotherapy treatment
`had no statistically significant benefit in median OS, whereas
`
`it did in the experiment shown in Fig. 2B. This difference
`may be due to the more aggressive tumor growth we noted
`in the experiment shown in Fig. 2A where all of the control
`mice died by day 60 compared with experiment 2B where all
`control mice died by day 70. Nevertheless in both experi-
`ments, sunitinib did not improve paclitaxel treatment out-
`comes. We also tested the effect of paclitaxel plus sunitinib
`in the established primary tumor model (Fig. 2C); sunitinib
`had a noticeable antitumor effect in contrast to paclitaxel;
`the 2 drug combination was not significantly different from
`the sunitinib-treated group.
`We next tested the antiangiogenic anti-VEGFR-2 antibody
`known as DC101 (20). The rationale for doing so was based on
`the results of the prior E2100 phase III trial evaluating bev-
`acizumab with paclitaxel in patients with MBC, which showed
`a gain in PPS of almost 6 months compared with paclitaxel
`
`
`
`www.aacrjournals.org
`
`Cancer Res; 73(9) May 1, 2013
`
`2745
`
`3
`
`

`

`Guerin et al.
`
`—-— Vehicle control
`
`—-— Sunitinib 60 mg/kg
`
`-- Paclitaxel so mg/kg*#
`-‘- Sunitinib + paclitaxel
`
`
`
`Percentsurvival
`
`-— Vehicle control
`—I— Sunitinib 60 mg/kg
`_._ Paclitaxel 30 mg/kg
`-I- Sunitinib + paclitaxel
`
`
`
`Percentsurvival
`
`75
`65
`55
`Da safterim lantation
`y
`p
`
`.
`.
`Median survnval:
`Vehicle control: 54 days
`Paclitaxel: 61 days
`Sunitinib: 65 days
`Sunitinib + paclitaxel: 50 days
`
`Therapy initiated on day 14
`
`so
`Days after implantation
`
`511
`
`4'0
`Median survival:
`Vehicle control: 65 days
`Paclitaxel: 88.5 days
`Sunitinib: 66 days
`Sunitinib + paclitaxel: 76 days
`
`Vehicle control
`Paclitaxel
`
`Sunitinib
`Sunitinib + paclitaxel
`
`10
`
`35
`3D
`25
`20
`15
`Days after cell implantation
`
`40
`
`45
`
`Figure 2. Differential impact of sunitinib plus paclitaxel chemotherapy when treating established primary tumors versus postsurgical advanced
`metastatic disease. Two independent metastatic therapy experiments are shown in A and B. Paclitaxel alone administered intraperitoneally at an MTD of 30
`mg/kg once every 2 weeks shows extensions of median survival in A and B, which was not statistically significant in one case (A). Combination of sunitinib with
`paclitaxel does not improve the survival advantages over paclitaxel alone (8) and may even worsen outcome (A). In the advanced metastasis studies,
`median survival for (i) control vehicle treatment was 54 to 65 days, (ii) paclitaxel treatment was 61 to 88.5 days, (iii) sunitinib treatment was 65 to 66 days, and (iv)
`sunitinib plus paclitaxel was 50 to 76 days; P values were not significant for A; B, ‘, paclitaxel versus control P z: 0.03; #, sunitinib + paclitaxel versus
`control P = 0.003. In established primary tumors, MTD paclitaxel alone shows no activity, whereas sunitinib alone or in combination with MTD
`paclitaxel inhibited tumor growth (C). In the advanced metastasis study (B), primary tumors were surgically resected 20 days after injection of 2 x 106 cells
`when average size was approximately 400 mm3 and treatment was initiated 20 days later; in the primary tumor study (0), treatment was initiated 14 days
`after cell injection when average tumor size was 150 mma.
`
`alone (16). As shown in Fig. 3A, DC101 had a robust antitumor
`effect when used to treat established LM2.4 primary tumors.
`The paclitaxel treatment, once again, did not have an antitu—
`mor effect when used to treat primary tumors. The 2 drug
`combination was the most effective. However, as shown in Fig.
`3B the pattern of response as assessed by survival analysis was
`somewhat different in the postsurgical advanced metastatic
`setting. Similar to sunitinib or pazopariib, DC101 was seem-
`ingly devoid ofactivity, at least when used as monotherapy and
`assessed by its impact on survival, because median survival was
`64 to 65 days in both the control and DC101 treatment groups.
`The paclitaxel monotherapy treatment showed a trend for a
`survival benefit (from 65 to 78 days) but this did not reach
`
`statistical significance. However, median survival was signif-
`icantly prolonged to 88 days in the combination treatment
`group compared with control, untreated mice. The difference
`in median survival between the DC101 + paclitaxel group
`(88 days) and the paclitaxel group (78 days) was not statistically
`significant, but would likely have led to a benefit in PPS had we
`been able to undertake such an assessment, and if so, would
`mirror the results of the E2100 phase III trial. Taken together,
`there does seem to be a modest benefit in improving the
`therapeutic impact of paclitaxel by combination with the
`VEGF pathway targeting antibody, but not with sunitinib,
`observations that seem to reflect previous phase III clinical
`trial results (13, 16), including a head-to—head comparison of
`
`2746
`
`
`Cancer Research
`Cancer Res; 73(9) May 1, 2013
`
`4
`
`

`

`Differential Drug Efficacy on Primary versus Metastatic Tumors
`
`2,500
`
`2,250
`2,000
`1,750
`
`1,500
`1,250
`1,000
`750
`500
`
`250
`
`
`
`
`
`Tumorvolume(mm3)
`
`Control
`
`
`
`
`
`Paclitaxel
`
`1 1 0
`1 00
`90
`80
`7O
`60
`50
`40
`30
`20
`1 0
`
`
`
`Percentsurvival
`
`
`
`
`-'- Control
`
`-'- Paclitaxel 50 mg/kg
`"" DC101
`
`i D0101 + paclitaxel""+
`
`
`
`40
`
`so
`
`100110120
`so
`so
`7'0
`so
`Days after cell implantation
`
`D0101+paclitaxel
`
`70
`
`80
`
`
`
`
`10
`
`20
`
`30
`
`40
`
`50
`
`60
`
`Days after cell implantation
`
`
`
`Median survival:
`Control: 65 days
`Paclitaxel: 78 days
`D0101: 64 days
`DC101+ paclitaxelz 88 days.
`_ ____——l
`
`Figure 3. Differential impact of an antibody to VEGFFt-2 (D0101) plus MTD paclitaxel on established orthotopic primary LM2-4 tumors versus postsurgical
`advanced metastatic disease. D0101 800 rig/mouse administered intraperitoneally twice per week showed marked antitumor activity when treating
`established primary tumors (A) but no survival activity in the setting of postsurgical advanced metastasis therapy (B); MTD paclitaxel 50 mg/kg i.p. once every 3
`weeks showed no activity in the primary tumor therapy model but showed an increase, although not statistically significant, in median survival in the
`postsurgical advanced metastasis model; combination of D0101 with MTD paclitaxel PTX increased the extent of inhibition of primary tumor growth (A)
`and had a significant survival benefit in advanced metastatic model (B). in the advanced metastasis studies (B), median survival for (i) control treatment was
`65 days, (ii) paclitaxel treatment was 78 days, (iin DC101 treatment was 64 days, and (iv) 00101 + paclitaxel was 88 days; ‘, D0101 + paclitaxel
`versus control P .— 0.0006; + D0101 + paclitaxel versus PTX P = 0.42. In the primary tumor study (A), treatment with D0101 was initiated 15 days later when
`average tumor size was 150 mm"; in the advanced metastasis study (B), primary tumors were surgically resected 21 days after cell injection when
`average size was approximately 400 mm3 and treatment was initiated 25 days later.
`
`sunitinib plus paclitaxel versus bevacizumab plus paclitaxel
`(13) as well as the known modest to minimal PFS activity of
`single-agent paclitaxel in metastatic triple-negative breast
`cancer (16).
`Several questions are raised by our results. First, what is the
`basis for the widely divergent effects we have observed when
`treating primary tumor-bearing mice versus mice with postsur-
`gical advanced metastatic disease with antiangiogenic drugs?
`Some possibilities include reduced expression of VEGF, or
`VEGFR—2 in the tumor or tumor vasculature of established
`
`metastases compared with the primary tumors. The qualitative
`characteristics of the vasculature in the slightly "older" metasta-
`ses may be substantially different from the primary tumors such
`that there is a greater proportion of late/mature vessels that are
`known to be less responsive to VEGF pathway targeting drugs
`(21). Metastases in vasculature rich organs such as the lung and
`liver may be more adept at co-opting the existing vasculature
`than tumors growing in the mammary fat pad (22). Second.
`would a similar pattern of results be observed if using GEMMs or
`PDXs? With respect to GEMMs, surgical resection ofthe multiple
`asynchronously arising primary tumors and the well-known
`observed lack of distant metastases in most such models make
`
`this difficult to answer. Nevertheless, some GEMM primary
`tumor therapy studies have shown a remarkable retrospective
`correlation with prior phase 111 clinical trial PFS or OS results of
`
`the respective tumor types (lung and pancreatic cancer) in part
`by using clinically relevant endpoints of tumor response (23).
`As for PDXs, some recent studies have shown the presence of
`metastases in clinically relevant patterns in nonresected primary
`tIHnor-bearing patient-derived breast cancer xenografis obtain-
`ed from patients with different breast cancer major subtypes
`(24), thus making it possible to use these models for preclinical
`adjuvant and metastatic therapy investigations.
`Finally, is there any evidence that an investigational therapy
`previously shown to be highly effective in the postsurgical
`preclinical metastatic setting shows prospective clinical
`activity? In this regard, we have reported that doublet oral
`low-dose metronomic chemotherapy using cyclophosphamide
`and a 5-fluorouracil prodrug (UFT), that is, tegafur + uracil,
`had potent activity in the postsurgical metastatic setting using
`the LM2-4 breast cancer model (9), whereas the activity was
`much less impressive when treating primary tumors in control
`unresected mice (9). A similar version of this metronomic
`chemotherapy was tested in phase II MBC trial, in combination
`with bevacizumab, with very encouraging results (25). How-
`ever, as discussed in the Introduction, such phase H trial results
`have to be confirmed in a larger randomized phase III trial,
`of which one is underway evaluating metronomic doublet
`cyclophosphamide and capecitabine with bevacizumab
`(NCT01131195; www.clinicaltrials.gov). Nevertheless, for now,
`
`
`
`www.aacrjournals.org
`
`Cancer Res; 73(9) May 1, 2013
`
`2747
`
`5
`
`

`

`Guerin et al.
`
`our results suggest the value of preclinical modeling postsur-
`gical advanced metastatic disease as a potential strategy to
`improve how they might predict clinical outcomes. The same
`may be the case for postsurgical adjuvant therapy models of
`early stage microscopic (minimal residual) metastatic disease,
`which can also respond to therapy in a manner different from
`primary tumors in control experiments (19, 26). Although not
`practical for routine drug screening, use of such models may be
`useful to confirm prior preclinical studies using conventional
`primary tumor therapy models, before embarking on expensive
`phase II or III metastatic therapy clinical trials.
`
`Disclosure of Potential Conflicts of Interest
`No potential conflicts of interest were disclosed
`
`Authors' Contributions
`Conception and design: E. Guerin, S. Man, R.S. Kerbel
`Development of methodology: S. Man, R.S. Kerbel
`Acquisition of data (provided animals, acquired and managed patients,
`provided facilities, etc.): E. Guerin, S. Man
`
`References
`
`Analysis and interpretation ofdata (e.g., statistical analysis, hiostatistics,
`computational analysis): E. Guerin, S. Man
`Writing, review, and/or revision ofthe manuscript: E. Guerin, S. Man, P. Xu,
`R.S. Kerbel
`Administrative, technical, or material support (i.e., reporting or orga—
`nizing data, constructing databases): S. Man, P. Xu
`Study supervision: R.S. Kerbel
`
`Acknowledgments
`The authors thank GSK through Dr. Rakesh Kumar for pazopanib, Pfizer
`through Dr.J. Christensen for sunitinib, and ImClone System through Dr. Bronek
`Pytowski for the DC101 antibody.
`
`Grant Support
`This work was supported by grants from the Ontario Institute for Cancer
`Research, the Canadian Breast Cancer Foundation, and the NIH (CA—41233;
`R.S. Kerbel).
`The costs ofpublication ofthis 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.
`
`Received November 14, 2012; revised February 1, 2013; accepted March 5, 2013;
`published OnlineFirst April 22, 2013.
`
`tabine (C) versus C in previously treated advanced breast cancer
`(ABC). J Clin Oncol 2010 (abstr LBA 1011).
`15. Barrios CH, Liu MC, Lee 80, Vanlemmens L, Ferrero JM, Tabei T, et al.
`Phase III randomized trial of sunitinib versus capecitabine in patients
`with previously treated HER2-negative advanced breast cancer.
`Breast Cancer Res Treat 2010;121:121—31.
`16. Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, et al.
`Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic
`breast cancer. N Engl J Med 2007;357:2666—76.
`17. Tait LR, Pauley RJ, Santner SJ, Heppner GH, Heng HH, Rak JW, et al.
`Dynamic
`stromaI-epithelial
`interactions during progression of
`MCF10DClS.com xenografts. lnt J Cancer 2007;120:2127—34.
`18. Shaked Y, Henke E, Roodhart J, Mancuso P, Langenberg M, Colleoni
`M, et al. Rapid chemotherapy-induced surge in endothelial progenitor
`cells:
`implications for antiangiogenic drugs as chemosensitizing
`agents. Cancer Cell 2008;14:263—715.
`19. Ebos JML, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG,
`Kerbel RS. Accelerated metastasis after short-term treatment with a
`potent inhibitor of tumor angiogenesis. Cancer Cell 2009;15:232—9.
`20. Klement G, Baruchel S, Rak J, Man S, Clark K, Hicklin D, et al.
`Continuous low-dose therapy with vinblastine and VEGF receptor-2
`antibody induces sustained tumor regression without overt toxicity.
`J Clin Invest 2000;105:R15—R24.
`21. Sitohy B, Nagy JA, Jaminet SC, Dvorak HF. Tumor-surrogate blood
`vessel subtypes exhibit differential susceptibility to anti-VEGF therapy.
`Cancer Res 2011;71:7021—8.
`22. Leenders WP, Kusters B, de Waal RM. Vessel co—option: how tumors
`obtain blood supply in the absence of sprouting angiogenesis. Endo-
`thelium 2002;9183—7.
`23. Singh M, Lima A, Molina R, Hamilton P, ClermontAC, Devasthali V, et al.
`Assessing therapeutic responses in Kras mutant cancers using genet-
`ically engineered mouse models. Nat Biotechnol 2010;28:585—93.
`24. DeRose YS, Wang G, Lin YC, Bemard PS, Buys 88, Ebbert MT, et al.
`Tumor grafts derived from women with breast cancer authentically
`reflect tumor pathology, growth, metastasis and disease outcomes.
`Nat Med 2011;17:1514—20.
`25. Dellapasqua S, Bertolini F, Bagnardi V, Campagnoli E, Scarano E,
`Torrisi R, et al. Metronomic cyclophosphamide and capecitabine
`combined with bevacizumab in advanced breast cancer: clinical and
`biological activity. J Clin Oncol 2008;26:4899—905.
`26. Day CP, Carter J, Bonomi C, Hollingshead M, Merlino G. Preclinical
`therapeutic response of residual metastatic disease is distinct from its
`primary tumor of origin. Int J Cancer 2012;130:190—9.
`
`7.
`
`4.
`
`5.
`
`1. Kamb A. What's wrong with our cancer models? Nat Rev Drug Discov
`2005;4z161—5.
`2. Kung AL. Practices and pitfalls of mouse cancer models in drug
`discovery. Adv Cancer Res 2007;96:191—212.
`3. Talmadge JE, Singh RK, Fidler IJ, Raz A. Murine models to evaluate
`novel and conventional therapeutic strategies for cancer. Am J Pathol
`2007;170:793-804.
`Singh M, Ferrara N. Modeling and predicting clinical efficacy for drugs
`targeting the tumor milieu. Nat Biotechnol 2012;30:648—57.
`Francia G, Cruz—Munoz W, Man 8, Xu P, Kerbel RS. Mouse models of
`advanced spontaneous metastasis forexperimental therapeutics. Nat
`Rev Cancer 2011;11:135—41.
`6. Steeg PS. Anderson RL. Bar—Eli M, Chambers AF, Eccles SA, Hunter K,
`et al. Preclinical drug development must consider the impact on
`metastasis. Clin Cancer Res 2009;15:4529—30.
`Fojo T, Amiri-Kordestani L, Bates SE. Potential pitfalls of crossover and
`thoughts on iniparib in triple-negative breast cancer. J Natl Cancer Inst
`2011;103:1738—40.
`8. Verma S, McLeod D, Batist G, Robidoux A, Martins lR, Mackey JR. in
`the end what matters most? A review of clinical endpoints in advanced
`breast cancer. Oncologist 2011;16:25—35.
`9. Munoz R, Man S, Shaked Y, Lee C, Wong J, Francia G, et al. Highly
`efficacious non-toxic treatment for advanced metastatic breast cancer
`using combination UFT—cyclophosphamide metronomic chemother-
`apy. Cancer Res 2006;66:3386—91.
`10. AbramsTJ, Murray LJ, Pesenti E, Holway VW, ColomboT, Lee LB, et al.
`Preclinical evaluation of the tyrosine kinase inhibitor SU11248 as a
`single agent and in combination with "standard of care" therapeutic
`agents for
`the treatment of breast cancer. Mol Cancer Ther
`2003;221011—21.
`11. Mackey JR, Kerbel RS, Gelmon KA, McLeod DM, Chia SK, Rayson D,
`et al. Controlling angiogenesis in breast cancer: a systematic review of
`anti-angiogenic trials. Cancer Treat Rev 2012;38:673—88.
`12. Bergh J, Bondarenko lM, Lichinitser MR, Liljegren A, Greil R, Voytko
`NL, et al. First-line treatment of advanced breast cancer with sunitinib
`in combination with docetaxel versus docetaxel alone: results of a
`prospective, randomized phase III study. J Clin Oncol 2012;30:921—9.
`13. Robert NJ, Saleh MN, Paul D, Generali D, Gressot L, Copur MS, et al.
`Sunitinib plus paclitaxel versus bevacizumab plus paclitaxel for first-
`line treatment of patients with advanced breast cancer: a phase III,
`randomized, open-label trial. Clin Breast Cancer 2011;11:82—92.
`14. Crown J, Dieras V, Starosiawska E, Yardley DA, Davidson N, Bachelot
`TD, et al. Phase III trial of sunitinib (SU) in combination with capeci-
`
`2748
`
`
`Cancer Research
`
`Cancer Res; 73(9) May 1, 2013
`
`6
`
`

`

`Cancer Research
`
`The journal of Cancer Research (1916—1930)
`
`| The American Journal of Cancer (1 931 ~1940)
`
`for Cancer Research
`AAC-R American Association
`
`A Model of Postsurgical Advanced Metastatic Breast Cancer More
`Accurately Replicates the Clinical Efficacy of Antiangiogenic Drug:
`
`Eric Guerin, Shari Man, Ping Xu, et al.
`
`Cancer Res 2013;713:2743-2748. Published OnlineFirst April 22, 2013.
`
` Updated version
`
`Access the most recent version of this article at:
`doi:10.1158/0008-5472.CAN-12-4183
`
`
`
`
`Cited articles
`This article cites 25 articles, 7 of which you can access for free at:
`http://cancerres.aacrjournals.org/content/73/9/2743.full#ref—|ist-1
`
`
`
`Citing articles
`This article has been cited by 15 HighWire-hosted articles. Access the articles at:
`http://cancerresaacrjournals.org/content/73/9/2743.full#related—urls
`
`
`
`
`S

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