Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics...

13
Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Dened Models of Colorectal Cancer Meera Raja 1 , Matt Zverev 1 , Katja Seipel 1,2 , Geraint T. Williams 3 , Alan R. Clarke 1 , and Paul H.S. Shaw 1 Abstract The objective of tailoring medicines for cancer patients accord- ing to the molecular prole of their disease holds great promise for the improvement of cancer therapy. Nevertheless, this approach has been limited, in part, due to the lack of predictive and informative preclinical studies. Herein, we describe an assessment of the therapeutic potential of targeting PI3K/mTOR and MAPK signaling in genetically dened mouse models of colorectal cancer mirroring disease subtypes targeted for novel therapy in the FOCUS4 trial. Our studies demonstrate that dual PI3K/mTOR inhibition is highly effective in invasive adenocarcinoma models characterized by combinatorial mutations in Apc and Pten; Apc and Kras; and Apc, Pten and Kras. MEK inhibition was effective in the combinatorial Apc and Kras setting, but had no impact in either Apc Pten mutants or in Apc Pten Kras triple mutants. Furthermore, we describe the importance of scheduling for com- bination studies and show that although no additional benet is gained in Apc Pten mice, combination of PI3K/mTOR and MAPK inhibition leads to an additive benet in survival in Apc Kras mice and a synergistic increase in survival in Apc Pten Kras mice. This is the rst study using robust colorectal cancer genetically engi- neered mouse models to support the validity of PI3K/mTOR and MEK inhibitors as tailored therapies for colorectal cancer and highlight the potential importance of drug scheduling in the clinic. Mol Cancer Ther; 14(10); 217586. Ó2015 AACR. Introduction The notion of stratifying therapy according to the molecular prole of a given tumor is considered to hold great promise for the improvement of therapeutic outcome. Despite this, failures of certain novel targeted therapeutic agents in clinical trials have limited the progress of this approach. This is largely attributed to a lack of accurate guidance from preclinical studies and it is thought that the increased use of reliable genetically engineered mouse models (GEMM) may lead to improvements in predicting ther- apeutic response (1). The FOCUS 4 clinical trial is currently the largest molecularly stratied multisite randomized trial to include targeted anticancer strategies against PIK3CA-mutant or PTEN (phosphatase and tensin homologue)-decient and KRAS or BRAF-mutant metastatic colorectal cancer (for More Information: http://www.focus4trial.org/). Our preclinical study has therefore mirrored some of the major tumor genetic backgrounds of the FOCUS 4 trial using GEMMs and matched novel targeted ther- apeutic agents to help determine the rationale for treatment combinations likely to ultimately improve patient outcome. The genetically engineered mouse models used in this study faithfully recapitulate molecular pathways involved in the initiation and progression of tumorigenesis and are therefore powerful tools for the assessment of novel agents and proof-of-principle studies for molecularly targeted therapies. Mutational activation of the KRAS oncogene is found in 40% to 50% of patients with colorectal cancer and, similarly, mutations activating the closely associated PI3K pathway occur in 40% of colorectal cancer cases (2). Both are known to be associated with poor prognosis and also predict poor outcome to conventional chemotherapy (3, 4). Moreover, KRAS and PI3K pathway muta- tions are established predictors of nonresponse to anti-EGFR (epidermal growth factor receptor)targeted therapies in colorec- tal cancer (5, 6). This has led to a surge in the development of targeted agents against PI3K and MAPK pathways and their assessment in preclinical studies. A number of recent studies have evidenced the benets of targeting these pathways using potent agents such as dual PI3K/mTOR and MEK1/2 inhibitors as single agents and in combination therapy using robust in vivo models of human cancer (710). For colorectal cancer however, these eva- luations have been restricted to cell-based assays and xenotrans- plantation studies that carry certain limitations, such as the lack of an intact immune system or tumor microenvironments (11). To augment these approaches, we have used three GEMMs of colo- rectal cancer, each bearing combinatorial mutations in Apc, Pten, and Kras (1214), to evaluate the benets of targeted agents against the relevant pathways activated in these tumors. Despite many advantages, one argument against the use of GEMMS as preclinical models is the impracticability of primary bowel resec- tion prior to palliative drug therapy. However, for many patients with metastatic colorectal cancer, the primary site of the disease 1 European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, United Kingdom. 2 University Hospital Bern, Bern, Switzerland. 3 School of Medicine, Cardiff University, Cardiff, United Kingdom. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Alan Clarke, European Cancer Stem Cell Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, United Kingdom. Phone: 44-29-208-74609; Fax: 44-29-208-74116; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-15-0223 Ó2015 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 2175 on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Transcript of Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics...

Page 1: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

Small Molecule Therapeutics

Assessment of the In Vivo Activity of PI3K andMEK Inhibitors in Genetically Defined Models ofColorectal CancerMeera Raja1, Matt Zverev1, Katja Seipel1,2, Geraint T.Williams3, Alan R. Clarke1, andPaul H.S. Shaw1

Abstract

The objective of tailoring medicines for cancer patients accord-ing to themolecular profile of their diseaseholds great promise forthe improvement of cancer therapy. Nevertheless, this approachhas been limited, in part, due to the lack of predictive andinformative preclinical studies. Herein, we describe an assessmentof the therapeutic potential of targeting PI3K/mTOR and MAPKsignaling in genetically definedmousemodels of colorectal cancermirroring disease subtypes targeted for novel therapy in theFOCUS4 trial. Our studies demonstrate that dual PI3K/mTORinhibition is highly effective in invasive adenocarcinoma modelscharacterized by combinatorial mutations in Apc and Pten; Apcand Kras; and Apc, Pten and Kras. MEK inhibition was effective in

the combinatorial Apc and Kras setting, but had no impact ineither Apc Pten mutants or in Apc Pten Kras triple mutants.Furthermore, we describe the importance of scheduling for com-bination studies and show that although no additional benefit isgained in Apc Pten mice, combination of PI3K/mTOR andMAPKinhibition leads to an additive benefit in survival in Apc Krasmiceand a synergistic increase in survival in Apc Pten Krasmice. This isthe first study using robust colorectal cancer genetically engi-neered mouse models to support the validity of PI3K/mTOR andMEK inhibitors as tailored therapies for colorectal cancer andhighlight the potential importance of drug scheduling in theclinic. Mol Cancer Ther; 14(10); 2175–86. �2015 AACR.

IntroductionThe notion of stratifying therapy according to the molecular

profile of a given tumor is considered tohold great promise for theimprovement of therapeutic outcome. Despite this, failures ofcertain novel targeted therapeutic agents in clinical trials havelimited the progress of this approach. This is largely attributed to alack of accurate guidance frompreclinical studies and it is thoughtthat the increased use of reliable genetically engineered mousemodels (GEMM) may lead to improvements in predicting ther-apeutic response (1). The FOCUS 4 clinical trial is currently thelargestmolecularly stratifiedmultisite randomized trial to includetargeted anticancer strategies against PIK3CA-mutant or PTEN(phosphatase and tensin homologue)-deficient and KRAS orBRAF-mutant metastatic colorectal cancer (for More Information:http://www.focus4trial.org/). Our preclinical study has thereforemirrored some of the major tumor genetic backgrounds of theFOCUS 4 trial using GEMMs and matched novel targeted ther-apeutic agents to help determine the rationale for treatment

combinations likely to ultimately improve patient outcome. Thegenetically engineeredmousemodels used in this study faithfullyrecapitulate molecular pathways involved in the initiation andprogression of tumorigenesis and are therefore powerful tools forthe assessment of novel agents and proof-of-principle studies formolecularly targeted therapies.

Mutational activation of the KRAS oncogene is found in 40% to50% of patients with colorectal cancer and, similarly, mutationsactivating the closely associated PI3K pathway occur in 40% ofcolorectal cancer cases (2). Both are known to be associated withpoor prognosis and also predict poor outcome to conventionalchemotherapy (3, 4). Moreover, KRAS and PI3K pathway muta-tions are established predictors of nonresponse to anti-EGFR(epidermal growth factor receptor)–targeted therapies in colorec-tal cancer (5, 6). This has led to a surge in the development oftargeted agents against PI3K and MAPK pathways and theirassessment in preclinical studies. A number of recent studies haveevidenced the benefits of targeting these pathways using potentagents such as dual PI3K/mTOR and MEK1/2 inhibitors as singleagents and in combination therapy using robust in vivomodels ofhuman cancer (7–10). For colorectal cancer however, these eva-luations have been restricted to cell-based assays and xenotrans-plantation studies that carry certain limitations, such as the lack ofan intact immune system or tumor microenvironments (11). Toaugment these approaches, we have used three GEMMs of colo-rectal cancer, each bearing combinatorial mutations in Apc, Pten,and Kras (12–14), to evaluate the benefits of targeted agentsagainst the relevant pathways activated in these tumors. Despitemany advantages, one argument against the use of GEMMS aspreclinical models is the impracticability of primary bowel resec-tion prior to palliative drug therapy. However, for many patientswith metastatic colorectal cancer, the primary site of the disease

1European Cancer Stem Cell Research Institute, Cardiff University,Cardiff, United Kingdom. 2University Hospital Bern, Bern, Switzerland.3School of Medicine, Cardiff University, Cardiff, United Kingdom.

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

Corresponding Author: Alan Clarke, European Cancer Stem Cell ResearchInstitute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF244HQ, United Kingdom. Phone: 44-29-208-74609; Fax: 44-29-208-74116; E-mail:[email protected]

doi: 10.1158/1535-7163.MCT-15-0223

�2015 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 2175

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 2: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

will often remain in situ in the absence of attributable symptoms;an approach being examined in the SYNCHRONOUS trial (15).Given this, we would argue that the GEMMs used in this studydirectly serve to reflect patients with colorectal cancer.

Mutations in genes that encode components of PI3K/AKT andRAS/MEK/ERK signaling frequently occur in colorectal cancer andcoexist in approximately a third of all cases (16). In addition,sequencing analysis reveals that mutations in adenomatous poly-posis coli (APC), KRAS, and PI3K are themost frequently observedgenetic events in human colorectal cancer (17). Loss of the tumor-suppressor protein PTEN has been shown to occur in 12% to 80%of human colorectal cancer cases (18–20). Moreover, PTEN loss isassociated with poor prognosis and predicts nonresponse to anti-EGFR agents currently available for KRAS wild-type colorectalcancer patients (21, 22). In light of this, we used three previouslyestablished and described GEMMs with combinatorial mutationsinApc andPten;ApcandKras; andApc,Pten, andKras (12–14).Wehave used these models of intestinal tumorigenesis, all of whichlead to invasive intestinal adenocarcinoma, to investigate theeffects of PI3K/mTOR inhibition using NVP-BEZ235; MEK1/2inhibition using MEK162 (Novartis Pharma); and combinatorialtherapy using both NVP-BEZ235 and MEK162, as stratified ther-apeutic strategies. Although it is widely known that TP53 muta-tions also contribute to the pathogenesis of human colorectalcancer, this study aimed to directly emulate current clinical trialsevaluating PI3K and MAPK pathway inhibitors by using only therelevant pathway status as a stratified approach to targeted therapy.

Materials and MethodsExperimental animals and tissue harvesting

Procedures were carried out in accordance with UK HomeOffice regulations. All mice were maintained on an outbredbackground and genotyped by PCR for appropriate alleles.Mousestrains harboring conditional deletion of Apc and Pten andoncogenic activation of Kras using the AhCreERT andVillinCreERT transgenes were induced as described previously(12, 13). For long-term survival studies, mice were aged until adefined treatment start point, at which they were randomized toreceive either 0.5% methyl cellulose (vehicle), 35 mg/kg NVP-BEZ235 twice daily (T-D; Novartis Pharmaceuticals), 30mg/kg ofMEK162 twice daily (Novartis Pharmaceuticals), or combinationat doses described in the text. Mice were treated daily untilsymptomatic of disease or to an endpoint of 500 days afterinduction (DPI) whereupon they were harvested 4 hours afterfinal dose. Kaplan–Meier survival analysis was performed usingSPSS Statistics 18.0. For short-term pharmacodynamic experi-ments, induced mice were aged until symptomatic of disease.Mice were then administered a single dose of vehicle (equivalentvolume per weight), 35 mg/kg of NVP-BEZ235, 30 mg/kg ofMEK162, or combinations (as outlined in the text) and harvested4 hours following the final dose. Mice used for short-term experi-ments were also administered a dose of bromodeoxyuridine(BrdUrd) 2 hours prior to killing. Intestinal tissues were flushedwith water, opened longitudinally, rolled into swiss-roll struc-tures, and fixed in 10% neutral buffered formalin (Sigma) over-night before processing and embedding in paraffin wax.

Western blot analysisProteins were extracted from snap-frozen tumors and Western

blot analysis was carried out as previously described (12). Equiv-

alent protein (30 mg) from at least 3 to 6 tumors from n� 3 micewere pooled per cohort and run in triplicate. Antibodies againstpAKT Ser473 (4060), pAKT Thr308 (4056), pS6RP (4858),p4EBP1 (2855), pERK (4370), total extracellular signal–regulatedkinase (ERK; 4377) from Cell Signaling Technology and b-actin(A5216) from Sigma were used. Appropriate horseradish perox-idase (HRP)–linked secondary antibodies (GE Healthcare) andECL reagents (GE Healthcare) were used to develop blots.

Histology, immunohistochemistry, and scoringFormalin-fixed paraffin-embedded tissues were sectioned (5

mm), stained with hematoxylin and eosin (H&E) for tumornumber analysis or, used for immunohistochemistry (IHC). Totaltumor numberwas scoredblind from threeH&E-stained slides foreach sample. IHC against cleaved caspase-3 (1:200; 9664; CellSignaling Technology) and BrdUrd (1:150; C47580; BD Bios-ciences) was performed using standard methodologies. Positivestainingwas scored as a percentage of total epithelial cells per fieldof view. Typically, five fields were assessed in all tumors in aminimum of 3 mice per cohort. Error bars represent standarddeviation of the mean. Statistical analysis was performed usingthe Mann–WhitneyU test. All analysis on scoring were two-tailedtests where n equals the number of lesions scored.

ResultsDescription and validation of colorectal cancer models

Inactivation of Apc and Pten was used to mimic activation ofWnt signaling and PI3K signaling, respectively. Activation ofoncogenic Kras was used to mimic activation of MAPK signaling.Conditional mutation of these genes within the intestinal epi-thelium was achieved using tamoxifen and b-napthoflavone(BNF) for Apcf/þ Ptenf/f mice bearing the AhCreERT transgene;or tamoxifen dissolved in corn oil for Apcf/þ KrasV12DLSL/þ andApcf/þ Ptenf/f KrasV12DLSL/þ mice bearing the VillinCrERT trans-gene. These mice are referred hereon as, Apcf/þ Ptenf/f, Apcf/þ

KrasLSL/þ, and Apcf/þ Ptenf/f KrasLSL/þ, respectively. Left untreated,mice develop invasive intestinal malignancies with median sur-vivals of 100, 160, and 40 DPI, respectively. Although Apcf/þ

Ptenf/f andApcf/þPtenf/f KrasLSL/þmice develop lesionswithin thesmall intestine only, Apcf/þ KrasLSL/þ mice present with colonlesions additionally (12–14). Analysis of themolecular pathwaysin tumors harvested from these mice confirms activation ofrelevant pathways compared with Apcf/þ controls (Supplemen-tary Fig. S1), reinforcing credence of these models for therapeuticintervention. A cohort of mice for each tumor subgroup was alsoeuthanized at the relevant treatment start points to confirm thepresence of tumors with varying degrees of severity. As such,Apcf/þ Ptenf/f mice presented with adenomas and early invasiveadenocarcinomas characterized by sub-mucosal invasion, where-as Apcf/þ KrasLSL/þ and Apcf/þ Ptenf/f KrasLSL/þ mice primarilypresented with adenomas at the start of treatment (Supplemen-tary Figs. S2–S5).

Antagonism of PI3K/mTOR signaling in colorectal cancermodels

Given that PI3K signaling is increased in all three tumormodels(Supplementary Fig. S1), we first antagonized the pathway usingthe dual PI3K/mTOR inhibitor NVP-BEZ235. At 4 hours, follow-ing a single dose of NVP-BEZ235, PI3K/mTOR signaling wasreduced in Apcf/þ Ptenf/f tumors (Fig. 1A). At this time point, we

Raja et al.

Mol Cancer Ther; 14(10) October 2015 Molecular Cancer Therapeutics2176

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 3: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

also observed increased apoptosis (Fig. 1D; P � 0.05; Mann–Whitney U test) and reduced proliferation (Fig. 1E; P � 0.05;Mann–Whitney U test) as scoring by cleaved caspase-3 andBrdUrd positivity, respectively (Supplementary Figs. S2 andS3). Continuous daily treatment from 77 DPI was found tosignificantly increase survival of Apcf/þPtenf/fmice fromamedianof 99 to 266DPI (Fig. 2A; n� 15mice per cohort; P� 0.0001; log-rank test). Taken together, these data illustrate favorable antitu-mor activity of NVP-BEZ235 in Pten-deficient tumors.

We next evaluated the effect of NVP-BEZ235 exposure in theKras-mutant setting, given that Kras is known to activate PI3Ksignaling through direct interaction with p110a (23), and ourconfirmatory observation of this activation (Supplementary Fig.S1) in Apcf/þ KrasLSL/þ colon tumors. Western blot analysis ofshort-term treatment of NVP-BEZ235 revealed modest inhibitionof AKT signaling at Ser473 and Thr308, but substantial inhibition

of signaling downstream of mechanistic target of rapamycin(mTOR) complex 1 as assessed by levels of pS6RP (Fig. 1B).These molecular changes were accompanied by increased apo-ptotic signaling through cleaved caspase-3 (Fig. 1D; P � 0.05;Mann–Whitney U test; Supplementary Fig. S2), suggesting asignificant antitumor effect despite variable inhibition of PI3Ksignaling. In the long-term setting, NVP-BEZ235 treatment from100 DPI significantly increased survival of Apcf/þ KrasLSL/þ micefrom amedian of 153 to 343 DPI (Fig. 2B; n� 12 per cohort; P�0.0001; log-rank test), indicating considerable dependence ofKras-mutant tumors on PI3K/mTOR signaling. These data high-light the potential benefit of targeting PI3K signaling in Kras-mutant colorectal cancers.

Mutations in KRAS and those activating PI3K signaling coexistin a third of all colorectal cancers. In our Apcf/þ Ptenf/f KrasLSL/þ

mousemodel, although neither activation ofMAPK signaling nor

A B

D*

0123456789

SIT Colon polyps

Colon polyps

SIT SIT

APC PTEN APC KRAS APC PTENKRAS

% C

leav

ed c

aspa

se-3

*

* *

Colon polyps SITC

SIT SIT

05

1015202530354045

SIT SIT SIT

APC PTEN APC KRAS APC PTENKRAS

% B

rdU

rd+

cells

*

VehicleNVP-BEZ235

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

VehicleNVP-BEZ235

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

VehicleNVP-BEZ235Vehicle

NVP-BEZ235

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

Apcf/+ Ptenf/f Apcf/+ KrasLSL/+ Apcf/+ Ptenf/f KrasLSL/+

E

Figure 1.NVP-BEZ235 leads to differential inhibition of PI3K/mTOR signaling across tumor models and activation of apoptosis. Western blot analysis of tumor lysates fromApcf/þ Ptenf/f small intestinal tumors (SIT; A), Apcf/þ KrasLSL/þ colon polyps and SITs (B), and Apcf/þ Ptenf/f KrasLSL/þ SITs (C) exposed to vehicle or 35-mg/kgNVP-BEZ235 for 4 hours. Immunoblotting with antibodies against effectors of the PI3K/mTOR pathway revealed a marked reduction in signaling across thegenotypes. Cleaved caspase-3 (D) and BrdUrd (E)-positive cells per microscopic field were scored from short term (4 hours) vehicle (blue bar) and NVP-BEZ235(red bar)–treated tumors. A significant increase in cleaved caspase-3 staining was observed for all genotypes and a significant reduction in BrdUrd-positivecells was observed in Apcf/þ Ptenf/f tumors. Data represent average of five different fields per tumor; n � 3 mice per cohort; error bars, standard deviation.(� , P � 0.05; Mann–Whitney U test).

PI3K and MEK Inhibitors in Genetic Models of Colorectal Cancer

www.aacrjournals.org Mol Cancer Ther; 14(10) October 2015 2177

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 4: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

hyper-activation of PI3K signaling was observed in intestinaltumors (Supplementary Fig. S1),mice have a significantly reducedlifespan and present with more tumors at death (13). Despite theincreased number, Apcf/þ Ptenf/f KrasLSL/þ mice predominantlypresent with noninvasive adenomas similar to Apcf/þ KrasLSL/þ

mice (Supplementary Figs. S5A, S6A, and S7A), and as reflected inBrdUrd scoring of vehicle-treated tumors (Fig. 1e). Short-termexposure to NVP-BEZ235 resulted in minimal perturbation ofpAKT473 and pAKT308 levels, but pS6RP and p4EBP1 weresuppressed as similarly observed in the Apc Pten and Apc Krasmodels. Our data therefore show that concurrent activation ofboth PI3K and Kras diminishes the ability of NVP-BEZ235 toreduce signaling immediately downstream of PI3K (Fig. 1C).Nevertheless, levels of apoptosis as scored through cleaved

caspase-3 were found to be increased (Fig. 1D; P � 0.05;Mann–Whitney U test; Supplementary Fig. S2). Long-term inter-vention using NVP-BEZ235 from 22DPI in this tumormodel wasfound to significantly increase survival of mice from a median of40 to 104 DPI (Fig. 2C; n � 13 per cohort; P � 0.0001; log-ranktest).

Despite the favorable effects on survival in all three tumormodels, NVP-BEZ235 only reduced the total number of lesions inApcf/þ Ptenf/f mice as assessed at the time of death (Fig. 2D).However, such tumors were of increased size as assessed by totalarea (Supplementary Fig. S4A) and an increased proportion were"advanced invasive adenocarcinomas", defined as showing inva-sion through themusclewall (Supplementary Fig. S4B). Althoughthese observations may indicate a phenotype of resistant tumor

100

80

60

40

20

0

100

80

60

40

20

0

100

80

60

40

20

0

B C

DPI

Apcf/+ Ptenf/f Apcf/+ KrasLSL/+ Apcf/+ Ptenf/f KrasLSL/+VehicleNVP-BEZ235 T-DTreatmentstart

Sur

viva

l pro

babi

lity

(%)

A

*

V N-BSIT

Apcf/+

V N-BColon polyps

V N-BSIT

V N-BSIT

Tota

l num

ber o

f les

ions

D

20

40

60

80

100

0

0 100 200 300 400 500 0 100 200 300 400 500 0 50 100 150 200 250

Ptenf/f Apcf/+ KrasLSL/+

KrasLSL/+Apcf/+ Ptenf/f

Figure 2.Continuous NVP-BEZ235 treatment extends survival of all tumor models and reduces tumor number in Apcf/þ Ptenf/f mice. The Kaplan–Meier survival analysis ofApcf/þ Ptenf/f (A), Apcf/þ KrasLSL/þ (B), and Apcf/þ Ptenf/f KrasLSL/þ (C) mice on vehicle (blue line) or 35-mg/kg NVP-BEZ235 (green line) twice daily by oralgavage revealed significantly increased survival across all genotypes (median survivals: Apcf/þPtenf/f vehicle¼99days vs. NVP-BEZ235¼ 266DPI, Apcf/þKrasLSL/þ

vehicle ¼ 153 vs. NVP-BEZ235 ¼ 343 DPI, Apcf/þ Ptenf/f KrasLSL/þ vehicle ¼ 40 vs. NVP-BEZ235 ¼ 104 DPI; P � 0.0001, log-rank test) Treatment start points:Apcf/þ Ptenf/f ¼ 77DPI, Apcf/þ KrasLSL/þ ¼ 100DPI, Apcf/þ Ptenf/f KrasLSL/þ ¼ 22DPI. D, tumors at death were scored from H&E stained intestinal sectionsfor each cohort. A significant reduction in tumor number was observed for Apcf/þ Ptenf/f mice on NVP-BEZ235 (N-B) treatment compared with vehicle (V).� , P � 0.05; n � 12 per cohort, Mann–Whitney U test.

Raja et al.

Mol Cancer Ther; 14(10) October 2015 Molecular Cancer Therapeutics2178

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 5: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

growth in response to chronic NVP-BEZ235 dosing, this inter-pretation is confounded by the animals' longevity—treated micesurvived much longer than controls (median, 266 vs. 100 days).In contrast, analysis of tumors arising in Apcf/þ KrasLSL/þ micerevealed that while NVP-BEZ235 treatment had no effect on thetotal burden of colon tumors, it was associated with a significantreduction in the total tumor area of small intestinal tumors, albeitwithout any change in the total number of lesions present or intheir invasive characteristics (Supplementary Figs. S5A, S5B, S6A,and S6B). The later was also the case for Apcf/þ Ptenf/f KrasLSL/þ

lesions following long-term NVP-BEZ235 exposure (Supplemen-tary Fig. S7A). Interestingly, histologic examination showed thatmany of the intestinal tumors in the treated Apcf/þ Ptenf/f

KrasLSL/þ cohort contained areas of incipient or frank ulceration

(Supplementary Fig. S7B and S7C). A smallminority of thesewereulcerating adenocarcinomas, but most were noninvasive adeno-mas showing a spectrum of degenerative changes that wereinterpreted to represent progressive tumor destruction/regressionleading to loss of mucosal barrier integrity and ulceration.

MEK inhibition throughMEK162 inmodels of colorectal cancerGiven that activation of oncogenic Kras reduced the ability of

NVP-BEZ235 to completely inhibit PI3K signaling, we next inves-tigated the consequences of inhibitingMAPK signaling though theMEK1/2 inhibitor MEK162 in all three tumor models.

In Apcf/þ Ptenf/f mice, short-term exposure toMEK162 reducedlevels of pERK and surprisingly also pAKT308 (Fig. 3A), highlight-ing the complexity of targeting closely associated signaling

A B CSIT Colon polyps SIT SIT

Apcf/+ Ptenf/f Apcf/+ KrasLSL/+ Apcf/+ Ptenf/f KrasLSL/+

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

Vehicle MEK162 Vehicle MEK162 Vehicle MEK162 Vehicle MEK162

D E *

0

2

4

6

8

10

12

14

SIT Colon polyps

SIT SIT

APC PTEN APC KRAS APC PTENKRAS

% C

leav

ed c

aspa

se-3

**

05

1015202530354045

SIT Colon polyps

SIT SIT

APC PTEN APC KRAS APC PTENKRAS

% B

rdU

rd+

cells

*

Figure 3.MEK inhibition reduces MAPK signaling but results in differential modulation of PI3K signaling across the tumor models. Western blot analysis of tumor lysates fromApcf/þ Ptenf/f small intestine tumors (SITs; A), Apcf/þ KrasLSL/þ colon polyps and SITs (B), and Apcf/þ Ptenf/f KrasLSL/þ SITs (C) exposed to vehicle or 30-mg/kgMEK162 for 4 hours. Immunoblotting with antibodies against the MAPK effector pERK revealed decreased signaling across the genotypes. An increasein PI3K/mTOR signaling was observed in some Apcf/þ KrasLSL/þ tumors; however, a reduction in signaling was observed in Apcf/þ Ptenf/f KrasLSL/þ tumors.Cleaved caspase-3 (D) and BrdUrd-positive cells (E) per microscopic field were scored from short-term (4 hours) vehicle (blue bar) and MEK162 (red bar)-treatedtumors. A significant increase in cleaved caspase-3 staining was observed in Apcf/þ KrasLSL/þ colon polyps and Apcf/þ Ptenf/f KrasLSL/þ SITs. A reduction inBrdUrd-positive cells was observed in Apcf/þ Ptenf/f tumors, and a significant increase in BrdUrd-positive cells in Apcf/þ Ptenf/f KrasLSL/þ tumors. Data representaverage of five different fields per tumor; n � 3 mice per cohort; error bars, standard deviation. � , P � 0.05, Mann–Whitney U test.

PI3K and MEK Inhibitors in Genetic Models of Colorectal Cancer

www.aacrjournals.org Mol Cancer Ther; 14(10) October 2015 2179

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 6: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

cascades. Although acute MEK inhibition significantly reducedproliferation, (Fig. 3E; P � 0.05; Mann–Whitney U test; Supple-mentary Fig. S3), there was no effect on survival of Apcf/þ Ptenf/f

mice when administered MEK162 continuously from 77 DPI(Fig. 4A, median survival: vehicle ¼ 99 days vs. MEK162 ¼101 DPI; n � 15; P � 0.05, log-rank method)

In Apcf/þ KrasLSL/þ mice, short-term MEK162 exposureincreased apoptosis through cleaved caspase-3 in Apcf/þ

KrasLSL/þ colon tumors (Fig. 3d; P� 0.05; Mann–WhitneyU test)and abolished detectable ERK signaling (Fig. 3B). Interestingly,Apcf/þ KrasLSL/þ colon tumors displayed variable alterations inPI3K/mTOR signaling following MEK inhibitor treatment asassessed by Western blot analysis of pAKT308 and pS6RP, sug-gesting heterogeneous compensatory activation of the closely

associated PI3K signaling cascade (Fig. 3B) and may reflect inter-tumor heterogeneity. Long-term MEK162 administration from100 DPI significantly increased survival of mice from amedian of153 to 287 days (Fig. 4b; P � 0.0001; n � 12; log-rank test).Although median survival of Apcf/þ KrasLSL/þ mice exposed toNVP-BEZ235 is approximately 60 days (343 DPI; Fig. 2b) longer,the difference is not statistically significant (P � 0.05; log-ranktest), indicating equipotent effects of NVP-BEZ235 and MEK162with regard to long-term survival in this genetic setting.

Short-term exposure to MEK162 in Apcf/þ Ptenf/f KrasLSL/þ

tumor-bearing mice led to a reduction in ERK signaling (Fig.3C), an induction of apoptosis (Fig. 3D; P� 0.05;Mann–WhitneyU test; Supplementary Fig. S2) and also a reduction in PI3K/mTOR signaling indicated by reduced pAKT308, pS6RP, and

100

80

60

40

20

0

A Vehicle

MEK162T-DTreatmentstart

100

80

60

40

20

0

100

80

60

40

20

0

DPI

Sur

viva

l pro

babi

lity

(%)

B C

V MSIT

Apcf/+ Ptenf/f

V MColon polyps

V MSIT

Apcf/+ KrasLSL/+

V MSIT

Apcf/+ Ptenf/f

KrasLSL/+

Tota

l num

ber o

f les

ions

**

D

Apcf/+ Ptenf/f Apcf/+ Kras LSL/+ Apcf/+ Ptenf/f KrasLSL/+

20

40

60

80

100

0

0 20 40 60 80 100 120 140 0 100 200 300 400 500 0 20 40 60 80 100 120

Figure 4.Long-term MEK inhibition only is beneficial for Apc Kras mice. The Kaplan–Meier survival analysis of Apcf/þ Ptenf/f (A), Apcf/þ KrasLSL/þ (B), and Apcf/þ

Ptenf/f KrasLSL/þ (C) mice on vehicle (blue line) or 30-mg/kg MEK162 (red line) twice daily by oral gavage revealed significantly increased survival ofApcf/þ KrasLSL/þ mice only (median survivals: Apcf/þ Ptenf/f vehicle ¼ 99 days vs. MEK162 ¼ 101 DPI, Apcf/þ KrasLSL/þ vehicle ¼ 153 vs. MEK162 ¼ 287 DPI;P � 0.0001, log-rank test, Apcf/þ Ptenf/f KrasLSL/þ vehicle ¼ 40 vs. MEK162 ¼ 36 DPI). Treatment start points: Apcf/þ Ptenf/f ¼ 77DPI, Apcf/þ KrasLSL/þ ¼ 100DPI,Apcf/þ Ptenf/f KrasLSL/þ ¼ 22DPI. Tumors at death were scored from H&E-stained intestinal sections for each cohort (D). A significant reduction in colonand small intestinal tumor number was observed for Apcf/þ KrasLSL/þ mice on MEK162 treatment (M) compared with vehicle (V). � , P � 0.05; n � 12 per cohort,Mann–Whitney U test.

Raja et al.

Mol Cancer Ther; 14(10) October 2015 Molecular Cancer Therapeutics2180

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 7: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

p4EBP1 levels (Fig. 3C). However, this was also accompanied byincreased cellular proliferation at this time point (Fig. 3e; P �0.05; Mann–Whitney U test, Supplementary Fig. S3). Long-termMEK162 exposure from 22 DPI did not deliver a significantsurvival benefit (Fig. 4c, median survival vehicle ¼ 40 days vs.MEK162 ¼ 36 days; P � 0.05; n � 13; Mann–Whitney U test),indicating that additional Pten loss negated the beneficial effect ofMEK162 observed in Apcf/þ KrasLSL/þ tumors.

Analysis of tumor burden at death following MEK162 treat-ment in all three tumor models echo the observations fromsurvival analysis, in that a significant reduction in the totalnumber of lesions was only observed in the responding Apcf/þ

KrasLSL/þ mice (Fig. 4D). Furthermore, significant alterations intumor area were only detected in the small intestine despite theincrease in invasive lesions (Supplementary Figs. S5A, S5B, S6A,and S6B). For Apcf/þ Ptenf/f mice, MEK162 had no significanteffect on tumor number, tumor area, or invasive characteristics(Fig. 4D; Supplementary Fig. S4A and S4B), whereas for Apcf/þ

Ptenf/f KrasLSL/þmice, although total tumor number appears to bereduced, the proportion of invasive lesions following MEK162treatment is slightly increased (Fig. 4D and Supplementary Fig.S7A), potentially attributable to the increased proliferation intumors observed from acute MEK inhibition.

Combinatorial therapy in models of colorectal cancerThe observations from long-term therapeutic and short-term

antitumor experiments of NVP-BEZ235 and MEK162 as singleagents, suggest combinatorial inhibition could provide addition-al therapeutic benefits in all tumormodels, in particular for Apcf/þ

KrasLSL/þ and Apcf/þ Ptenf/f KrasLSL/þ mice, as elements of cross-talk were more apparent in these settings. First, however, it wascrucial to establish an effective method of administering the twocompounds to achieve themost favorable antitumor effects. Threecombination strategies were chosen which involved administra-tion of 30 mg/kg MEK162 1 hour prior to (combo 1), after(combo 2), or at the same time (combo 3) as 35 mg/kg NVP-BEZ235. For all cohorts, mice were euthanized 4 hours followingthe final dose and samples were collected for evaluation of shortterm pharmacodynamics and antitumor activity. Probing oftumor lysates for effectors downstreamof PI3K/mTOR andMAPKsignaling revealed that Apcf/þ Ptenf/f tumors were particularlysensitive to the order of drug sequencing. In this tumor setting,combination strategy 2 (combo2) resulted inmaximal inhibitionof pERK and PI3K/mTOR effectors (Fig. 5A), as well as increasedapoptosis and reduced proliferation (Fig. 5E and F). Interestingly,MEK inhibition prior to or concurrently with NVP-BEZ235,reduced its ability to inhibit PI3K and mTOR signaling inPTEN-deficient tumors (Fig. 5A). These findings are unlikely dueto feedback activation of PI3K signaling as single-agent MEKinhibition in Apcf/þ Ptenf/f mice did not lead to compensatoryactivation of PI3K signaling in tumors (Fig. 3A).

Apcf/þ KrasLSL/þ tumors responded to the different combina-tion strategies in similar ways through comparably reduced levelsof pERK and pS6RP (Fig. 5C and S5C), and increased levels ofcleaved caspase 3 (Fig. 5e). No discernible alterations in the levelsof pAKT at either Ser473or Thr308, or in the levels of p4EBP1wereobserved (Fig. 5B and C). One potential explanation for thisapparent contradiction is that the inhibition of pS6RP may bethrough interactions between terminal ERK signaling and mTORrather than through AKT, as the level of p4EBP1 (which is alsodownstream of AKT/mTOR signaling) was unaltered. Neverthe-

less, the pharmacodynamic effects observed in Apcf/þ KrasLSL/þ

colon polyps may be due to an additive effect of combininginhibitors, given that MEK inhibition results in increased levels ofpAKT (Fig. 3b) and PI3K/mTOR inhibition in this setting pre-dominantly alters signaling downstream of mTOR (Fig. 1b),further providing rationale for long-term combination therapyin this tumor setting.

Similarly, Apcf/þ Ptenf/f KrasLSL/þ tumors displayed parallels inresponse to the varied combination strategies. Although levels ofpERK and pS6RP were found to be reduced with all three strat-egies, pAKT473 was only reduced following combo 3 (Fig. 5D). Amarked increase in apoptosis through cleaved caspase-3 wasobserved for all three strategies (Fig. 5E), indicating proapoptoticsignaling. Proliferation was increased with both combo 1 and 2,similar to our observations with single-agent MEK162, possiblysignifying an adaptive response (Fig. 5F). Nevertheless, given thatApcf/þ Ptenf/f KrasLSL/þ mice responded dramatically to single-agent NVP-BEZ235 in the long term but acutely showed onlymodest pathway inhibition, the lack of complete pathway inhi-bition from the short-term combination studies was not regardedas unfavorable. Therefore, despite some differences in response tothe combination strategies between the tumor models, combi-nation strategy 2,which appeared to bebeneficial for all cohorts interms of favorable pharmacodynamics and antitumor activity,was chosen as the strategy for long-term combination treatment.

In the Apcf/þ Ptenf/f setting, mice were exposed using a dailyregimen of combo 2 (NVP-BEZ235 1 hour prior to MEK162),followed 8 hours later by an additional daily administration ofNVP-BEZ235. This regimen is termed Combo R1 and was used toensure tolerability, as twice-daily combo 2 exposure was found tocause weight loss. Although combo R1 was found to significantlyincrease survival of mice compared with vehicle controls, (Figure6a, median survival: vehicle¼ 99 days vs. combo¼ 270 DPI; n�15; P � 0.0001; log-rank method) this did not provide anyadditional benefit to single agent NVP-BEZ235 (Fig. 6A, mediansurvival: NVP-BEZ235¼ 266 days vs. combo¼ 270DPI; n� 15; P� 0.05; log-rank test). Assessment of tumors at death revealedfurther similarities to single-agent NVP-BEZ235: total tumornumbers (Fig. 6D; P � 0.05; Mann–Whitney U test), tumor area(Supplementary Fig. S4A), and the proportion of invasive lesions(Supplementary Fig. S4B) were found to be similar to NVP-BEZ235, indicating that the addition of MEK162 in this settingprovided no additional benefits for survival or tumor burden.These data strongly suggest that PI3K and mTOR inhibition iseffective and sufficient for a therapeutic response in PTEN-deletedintestinal tumors.

For combination treatment experiments inApcf/þKrasLSL/þ andApcf/þ Ptenf/f KrasLSL/þ mice, the dosing regimen was furtherreduced to only a single daily combo 2 regimen (NVP-BEZ2351 hour prior to MEK162) to ensure tolerability. This is referred toas combo R2. Subsequently, additional control arms of singleagentswere undertaken to establish the full effects of combinationtreatment. In Apcf/þKrasLSL/þmice, the reduced once-daily (O-D)NVP-BEZ235 and MEK162 remained equipotent (Fig. 6b). Inaddition, NVP-BEZ235 elicited effects in a dose-dependent man-ner, but MEK162 reached maximal effects with the once-dailydosing regimen, in terms of survival benefit (Figs. 2B, 4B, and 6B).Although the reduced single-agent doses resulted in similar colontumor severity profiles, the total tumor area appears increasedcompared with higher dose regimens, suggesting that singletreatments are more permissive for colon tumor growth

PI3K and MEK Inhibitors in Genetic Models of Colorectal Cancer

www.aacrjournals.org Mol Cancer Ther; 14(10) October 2015 2181

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 8: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

(Supplementary Fig. S5A and S5B). In contrast, small intestinallesions fromonce daily–treatedmice appeared similarwith regardto tumor area and severity in comparison with tumors frommice

on twice-daily treatments, suggesting comparable sensitivities ofsmall intestinal tumors to the long-term treatments (Supplemen-tary Fig. S6A and A6B).

Apcf/+Ptenf/+ SIT Apcf/+ KrasLSL/+colon polyps

Apcf/+ Ptenf/f KrasLSL/+SIT

A

C

F

B

E *

Apcf/+ KrasLSL/+ SIT D

−505

10152025303540

SIT Colon polyps

SIT SIT

APC PTEN APC KRAS APC PTENKRAS

% C

leav

ed c

aspa

se-3

* * *

*

05

1015202530354045

SIT Colon polyps

SIT SIT

APC PTEN APC KRAS APC PTENKRAS

% B

rdU

rd+

cells

* *

Vehicle Combo 1 Combo 2 Combo 3 Vehicle

Vehicle Combo 1 Combo 2 Combo 3 Vehicle

Vehicle Combo 1 Combo 2 Combo 3 Vehicle

Vehicle Combo 1 Combo 2 Combo 3 Vehicle

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

pERK

T ERK

pAKT 473

pAKT 308

pS6RP

p4EBP1

β-Actin

Figure 5.Sequencing of combination is crucial for effective inhibition of MEK–ERK and PI3K–AKT signaling in Apc Pten tumors. Western blot analysis of tumor lysates fromApcf/þ Ptenf/f small intestinal tumors (SITs; A), Apcf/þ KrasLSL/þ colon polyps and SITs (B and C), and Apcf/þ Ptenf/f KrasLSL/þ SITs (D) mice exposed to thedifferent combination strategies. Immunoblottingwith antibodies against downstreameffectors ofMAPK and PI3K/mTORpathways revealed complete inhibition ofsignaling only with combo 2 in Apcf/þ Ptenf/f tumors. Apcf/þ KrasLSL/þ and Apcf/þ Ptenf/f KrasLSL/þ tumors were less sensitive to the order of sequencingand all combinations reduced signaling MAPK and PI3K/mTOR signaling through pERK and pS6RP, respectively. Scoring of cleaved caspase-3 (E) andBrdUrd-positive cells (F) in tumors from mice exposed to vehicle (blue bar), combo 1 (red bar), 2 (green bar), and 3 (purple bar). A significant increase incleaved caspase-3 scoring was observed with combo 2 and 3 in Apcf/þ Ptenf/f tumors, with combo 2 and 3 in Apcf/þ KrasLSL/þ colon polyps and with all threecombination strategies in Apcf/þ KrasLSL/þ and Apcf/þ Ptenf/f KrasLSL/þ SITs. A significant reduction in BrdUrd-positive cells was observed with combo 1and 2 in Apcf/þ Ptenf/f tumors and a significant increase in staining with combo 2 and 3 in Apcf/þ KrasLSL/þ and combo 1 and 2 in Apcf/þ Ptenf/f KrasLSL/þ SITs.Data represent average of five different fields per tumor, n � 3 mice per cohort, error bars represent standard deviation. � , P � 0.05, Mann–Whitney U test.

Raja et al.

Mol Cancer Ther; 14(10) October 2015 Molecular Cancer Therapeutics2182

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 9: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

Combination treatment in Apcf/þ KrasLSL/þ mice was found tobe well tolerated and resulted in an additive increase in mediansurvival compared with single agents (Fig. 6b, median survivals:vehicle¼ 153 days vs. NVP-BEZ235 O-D¼ 238 days vs. MEK162O-D ¼ 286 days vs. combo R2 ¼ 389 DPI, all comparisons aresignificant at P� 0.0001 except forMEK162O-D vs. NVP-BEZ235O-D P � 0.05; log-rank method). In the colon, combinationtreatment was found to result in reduced tumor number, but nosignificant change in tumor area or severity, despite the increase inelapsed time from treatment start point, potentially indicatingthat treatment results in restriction of both tumor growth and

progression (Fig. 6D and Supplementary Fig. S5A and A5B).Contrastingly, in the small intestine, combined NVP-BEZ235 andMEK162 administration resulted in reduced tumor number aswell as reduced tumor area, indicating potent effects on tumorgrowth (Fig. 6D and Supplementary Fig. S6A and S6B).

Finally, combination treatment (combo R2) administered toApcf/þPtenf/f KrasLSL/þmice also resulted in an increase in survivalwhen compared with vehicle cohorts (Fig. 6C, median survivals:vehicle¼40days vs. combo¼125DPI;P�0.0001; log-rank test).Analysis of tumors at death indicated no difference in the numberof lesions (Fig. 6D), but did also uncover the presence of tumor

100

80

60

40

20

0

A

DPI

Sur

viva

l pro

babi

lity

(%)

Vehicle NVP-BEZ235 T-D MEK162 T-D Combo R1 Treatmentstart

Apcf/+ Ptenf/f

Apcf/+ Ptenf/f

KrasLSL/+

Apcf/+ KrasLSL/+

100

80

60

40

20

0

B

DPI

Sur

viva

l pro

babi

lity

(%)

100

80

60

40

20

0

C

DPI

Sur

viva

l pro

babi

lity

(%)

Vehicle NVP-BEZ235 O-D MEK162 O-D Combo R2 Treatmentstart

Vehicle NVP-BEZ235 O-D MEK162 T-D Combo R2 Treatmentstart

D

V C SIT

Apcf/+ Ptenf/f

V C Colon polyps

V C SIT

Apcf/+ KrasLSL/+

V C SIT

Apcf/+ Ptenf/f

KrasLSL/+

*

* *

NS To

tal n

umbe

r of l

esio

ns

20

40

60

80

100

0

0 100 200 300 400 500

0 50 100 200 300150 250

0 100 200 300 400 500

Figure 6.Long-term evaluation of combination treatment in Apc Pten, Apc Kras, and Apc Pten Kras mice. The Kaplan–Meier survival analysis of Apcf/þ Ptenf/f (A),Apcf/þ KrasLSL/þ (B), and Apcf/þ Ptenf/f KrasLSL/þ (C) mice on vehicle (blue line) or combination (purple line) with control NVP-BEZ235 (green line) andMEK162 (red line) treatments. Administration of combination treatment significantly increased survival of Apcf/þ Ptenf/f mice compared with vehicle, howevernot compared with single-agent NVP-BEZ235. For Apcf/þ KrasLSL/þ mice, combination treatment prolonged longevity and resulted in an additive increasein survival when compared with single-agent MEK162 and NVP-BEZ235. In Apcf/þ Ptenf/f KrasLSL/þ mice, combined treatment synergistically increased survival ofmice compared with NVP-BEZ235 and MEK162 alone. (Median survivals: Apcf/þ Ptenf/f vehicle ¼ 99 days vs. combo R1 ¼ 270 DPI; P � 0.0001, log-rank test;Apcf/þKrasLSL/þ vehicle¼ 153 vs. comboR2¼ 389DPI;P�0.0001, log-rank test; Apcf/þPtenf/f KrasLSL/þ vehicle¼40vs. combo¼ 125 DPI;P�0.001, log-rank test.)Treatment start points: Apcf/þ Ptenf/f ¼ 77DPI, Apcf/þ KrasLSL/þ ¼ 100DPI, Apcf/þ Ptenf/f KrasLSL/þ ¼ 22DPI. Scoring of tumors from H&E-stained sections ofintestines (D) revealed a significant reduction in tumor number in those receiving combination therapy (C) compared with vehicle (V) in the Apcf/þ Ptenf/f

and Apcf/þ KrasLSL/þ cohorts (� , P � 0.05; n � 12 per cohort, Mann–Whitney U test).

PI3K and MEK Inhibitors in Genetic Models of Colorectal Cancer

www.aacrjournals.org Mol Cancer Ther; 14(10) October 2015 2183

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 10: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

ulceration suggestive of signs of tumor destruction/regression, asdescribed previously (Supplementary Fig. S7B and S7C). Despitethis, proportionally more invasive lesions characterized by sub-mucosal invasion were observed in the combination-treatedcohort, which may be due to either resistant tumor growth orsimply the increase in time elapsed (Supplementary Fig. S7A).Nevertheless, combination therapy resulted in a synergistic benefitin survival when compared with single-agents NVP-BEZ235 andMEK162, as when reduced to once-daily administration NVP-BEZ235 no longer elicited any survival benefit in Apcf/þ Ptenf/f

KrasLSL/þ mice (Fig. 6C; median survival: vehicle ¼ 40 days vs.NVP-BEZ235 O-D ¼ 36 DPI, P � 0.05; log-rank test). These datafurther suggest dose-dependent effects of reduced NVP-BEZ235similar to those seen in the Apcf/þ KrasLSL/þ mice. Furthermore,analysis of tumors at the end of treatment revealed that propor-tionally more superficially invasive carcinomas were present inNVP-BEZ235 once daily–treated mice in comparison with vehicleandNVP-BEZ235 twice-dailymice. Together, thesedata suggest thedose of NVP-BEZ235 used for therapeutic antagonism is critical.

DiscussionAlthough the promise of stratified medicine is currently far

from fulfilled, support for the notion of tailoring medicines for aspecific patient population remains strong, for example as evi-denced by the development of imatinib to specifically treatchronic myeloid leukemia and gastrointestinal stromal tumors.Here, we use robust and reliable GEMMs to assess the effect ofgenetic variation on drug response, in the context of murineintestinal neoplasia, with likely relevance to human colorectalcancer to inform clinical trials such as the FOCUS 4 trial. Giventhat mutations activating PI3K/AKT and RAS/MEK/ERK signalingcoexist in a third of all colorectal cancers, the efficacy of the dualPI3K/mTOR inhibitor NVP-BEZ235 and the MEK1/2 inhibitorMEK162 as single agents and in combination was addressed inthree autochthonous tumor models with differing combinatorialmutations in Apc, Pten, and Kras.

We have shown that dual PI3K and mTOR inhibition usingNVP-BEZ235 is beneficial therapeutically in the Apcf/þ Ptenf/f

setting in particular, but also for Apcf/þKrasLSL/þ andApcf/þPtenf/f

KrasLSL/þmice, evidenced by increasedmedian survivals (Fig. 2A–C). Thismaybe attributable to the role of PI3K/mTOR signaling ina number of crucial cellular processes, including cell survival andproliferation through activation of Akt. Phosphorylation andactivation of Akt through PDK1 at Thr308 and at Ser473 bymTORcomplex 2 (TORC2) leads to subsequent activation of TORC1signaling which regulates protein synthesis through activation ofribosomal protein S6 kinases and the eukaryotic initiation factor4E binding protein (4E-BP1; refs. 24, 25). In addition, Akt hasinhibitory effects on negative regulators of the cell cycle, includingp27 (kip1; ref. 26) and p21 (cip1; ref. 27), but also elicitsinhibitory effects on GSK-3, thus activating c-Myc and cyclinD1 that promote progression of the cell cycle (28). PI3K signalingis also critical for regulating cell death through inhibition of theproapoptotic factors Fas, Bim, and Bad (29–31) and regulatingautophagy and cellular metabolism, processes implicated inmediating increased longevity. A number of these properties ofPI3K signalingwere evaluated immediately after exposure toNVP-BEZ235 in our three models to determine the pharmacodynamicand antitumor effects of NVP-BEZ235. Here, Apcf/þ Ptenf/f micedisplayed substantial sensitivity to NVP-BEZ235 as complete

pathway inhibition was coupled with favorable increases inapoptotic signaling and reduced proliferation (Fig. 1A, D, andE). Interestingly, Kras-mutant tumors exhibited greater sensitivityfor TORC1 inhibition than PI3K inhibition (Fig. 1B), which maybe a result of promiscuous Kras activation of PI3K signalingthrough interactions with P110 (23). Furthermore, this effect wasexacerbated in the presence of concurrent Pten deletion andsuggests that oncogenic Kras prevents the ability of NVP-BEZ235to completely reduce AKT signaling (Fig. 1c). If indeed this effect isattributable to the presence of oncogenic Kras signaling, thishighlights the potential advantages of MEK inhibition in thesetumor settings.

Given the above observations, MEK inhibition was next inves-tigated usingMEK162 as single-agent therapy in our threemodelsof intestinal tumorigenesis. MEK inhibition in Apcf/þKrasLSL/þ

mice was found to be equipotent with PI3K/mTOR inhibition interms of survival (Figs. 2B, 4B, and 6B), suggesting equivalentdependence of KRAS-mutant tumors on PI3K/AKT and RAS/ERKsignaling. This property of KRAS-mutant tumors appears to becontext dependent. Similarly to the observations we report here,independent MEK and PI3K inhibition have been shown to beeffective in KRAS melanoma models (9); however, KRAS-mutantpancreatic and lung tumors were found to be more responsive toMEK rather than PI3K inhibition (8, 10). Our data also show thatadditional PTEN deletion renders otherwise sensitive KRAS-mutant tumors nonresponsive to MEK inhibition (Fig. 4C). Thisfinding is in accordance with previous studies and providesfurther evidence for the notion that KRASmutational status aloneis not sufficient as a prognostic marker for response to MEKinhibition (32–34).Moreover, given that Apcf/þPtenf/fmice showno response to long-term MEK inhibition (Fig. 4A), mutationsactivating the PI3K pathway, such as PTEN deletion, can also beused to predict nonresponse to MEK inhibitors in the KRAS wild-type tumor setting.

Although single-agents NVP-BEZ235 andMEK162 substantial-ly improved survival of mice as described previously, it wasanticipated that combination therapy may further increase thisbenefit. Despite the attractiveness of combinatorial therapy, thereis currently little data in the literature to direct the most appro-priate dosing schedule for combination of PI3K and MEK inhi-bitors. This is crucial, as while agents may be effective as singleagents, antagonism between two agents when combined, espe-cially given cross-talk between the two pathways may be evident.Furthermore, due to overlapping sensitivities, the combinationmay result in no net clinical gain when administered jointly (35).To address some of these issues, we chose to investigate threevaried combination strategies that differed in the order of com-pound administration. Here, MEK162 was administered 1 hourprior to (combo 1), after (combo 2), or at the same time (combo3) as NVP-BEZ235 to determine whether scheduling is key toachieve concomitant pathway inhibition. Interestingly, sensitivityto the scheduling was primarily detected in Apcf/þ Ptenf/f tumors,whereby MEK162 administered prior to or at the same time asNVP-BEZ235 diminished sensitivity to complete PI3K andmTORinhibition (Fig. 5a). Given that the most favorable antitumoreffects, in terms of increased apoptosis and reduced proliferation,were also observed by this strategy (combo 2), it was surprisingthat long-term administration provided no additional benefits tosingle-agent NVP-BEZ235 (Figs. 5E and F and 6A). It is possiblethat while MEK inhibition may lead to favorable pharmacody-namic effects in combination acutely, MAPK signaling is not

Raja et al.

Mol Cancer Ther; 14(10) October 2015 Molecular Cancer Therapeutics2184

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 11: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

required for tumormaintenance inApcf/þPtenf/fmiceand thereforepathway inhibition does not lead to a synergistic effect in the longterm. In direct contrast to the Apcf/þ Ptenf/f setting, Apcf/þKrasLSL/þ

and Apcf/þ Ptenf/f KrasLSL/þ tumors in response to the short termcombination strategies displayed less effective and less variation ininhibition of both pathways (Fig. 5B–D). Nevertheless, the obser-vation that long-term combo 2 administration increased mediansurvival additively in Apcf/þKrasLSL/þ mice and synergistically inApcf/þ Ptenf/f KrasLSL/þmice indicates that while some antagonismmay be prevalent as suggested by the short-term combinationstudies, the two pathways are essential in tumor maintenance(Fig. 6B and C). These data suggest that combination therapy inKRAS-mutant settings could provide substantial benefits.

In summary, we have performed a systematic preclinical studythat effectively evaluates rational therapeutic strategies for Apc,Pten, and Kras mutant colorectal cancer with the aim of identi-fying optimal clinical strategies. Our data show that PI3K/mTORinhibition enhances survival in both Kras-mutant and Pten-mutant settings. In contrast, MEK inhibition is only effective ina Kras-mutant background and this is overridden by additionalPten mutation. Critically, we also demonstrate true synergybetween the two therapies, but only in the presence of all three(Apc, Kras, and Pten)mutations. Taken together, our data confirmthe notion that specific pathway targeting is effective, at least withthe GEMMs used here, and so support the general concept ofstratified approaches to therapy. Our data also highlight bothsynergies and limitations in the use of therapeutic combinations,which occur in a genotype specific manner. Such studies toidentify stratified approaches have been previously conductedfor lung and ovarian cancers (7, 33); however, this is the first suchstudy conducted using GEMMs for colorectal cancer. As such, ourstudies should inform human clinical trials such as the FOCUS 4trial (http://www.focus4trial.org/).

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: M. Raja, A.R. Clarke, P.H.S. ShawDevelopment of methodology: M. Raja, P.H.S. ShawAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): M. Raja, M. Zverev, K. SeipelAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): M. Raja, K. Seipel, G.T. Williams, A.R. Clarke, P.H.S.ShawWriting, review, and/or revision of the manuscript: M. Raja, G.T. Williams,A.R. Clarke, P.H.S. ShawAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): M. Zverev, K. SeipelStudy supervision: A.R. Clarke, P.H.S. Shaw

AcknowledgmentsThe authors thank Mark Bishop and Lucie Taylor for genotyping of animals,

Fangli Chen for microscopy assistance, Derek Scarborough and Mark Isaac forhistology services, andWolfgangHackl andNovartis Pharmaceuticals for kindlysupplying NVP-BEZ235 and MEK162.

Grant SupportM. Raja was funded by Tenovus and NISCHR, M. Zverev was funded by

Cancer Research UK, and P.H.S. Shaw was funded by Cancer Research UK(Bobby Moore Fellowship), Welsh Deanery as Walport Clinical Lecture, andNISCHR (Welsh Government).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received March 18, 2015; revised July 13, 2015; accepted July 18, 2015;published OnlineFirst July 23, 2015.

References1. Robles AI, Varticovski L. Harnessing genetically engineered mouse models

for preclinical testing. Chem Biol Interact 2008;171:159–64.2. Bos JL, Fearon ER, Hamilton SR, Verlaan-de Vries M, van Boom JH, van der

Eb AJ, et al. Prevalence of ras gene mutations in human colorectal cancers.Nature 1987;327:293–7.

3. Richman SD, SeymourMT, Chambers P, Elliott F, Daly CL, Meade AM, et al.KRAS and BRAFmutations in advanced colorectal cancer are associated withpoor prognosis but do not preclude benefit from oxaliplatin or irinotecan:results from the MRC FOCUS trial. J Clin Oncol 2009;27:5931–7.

4. Ogino S, Nosho K, Kirkner GJ, Shima K, Irahara N, Kure S, et al. PIK3CAmutation is associated with poor prognosis among patients with curativelyresected colon cancer. J Clin Oncol 2009;27:1477–84.

5. Li�evre A, Bachet JB, Boige V, Cayre A, Le Corre D, Buc E, et al. KRASmutations as an independent prognostic factor in patients with advancedcolorectal cancer treated with cetuximab. J Clin Oncol 2008;26:374–9.

6. Yang ZY, Wu XY, Huang YF, Di MY, Zheng DY, Chen JZ, et al. Promisingbiomarkers for predicting the outcomes of patients with KRAS wild-typemetastatic colorectal cancer treated with anti-epidermal growth factorreceptor monoclonal antibodies: a systematic review with meta-analysis.Int J Cancer 2013;133:1914–25

7. Kinross KM, Brown DV, Kleinschmidt M, Jackson S, Christensen J, Culli-nane C, et al. In vivo activity of combined PI3K/mTOR andMEK inhibitionin aKras(G12D);Pten deletionmousemodel of ovarian cancer.Mol CancerTher 2011;10:1440–9.

8. Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, et al.Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D andPIK3CA H1047R murine lung cancers. Nat Med 2008;14:1351–6.

9. Roberts PJ, Usary JE, Darr DB, Dillon PM, Pfefferle AD, Whittle MC, et al.Combined PI3K/mTOR and MEK inhibition provides broad antitumoractivity in faithful murine cancer models. Clin Cancer Res 2012;18:5290–303.

10. Hofmann I,Weiss A, ElainG, SchwaederleM, SterkerD, Romanet V, et al. K-RASmutant pancreatic tumors showhigher sensitivity toMEK than to PI3Kinhibition in vivo. PLoS One 2012;7:e44146.

11. de Jong M, Maina T. Of mice and humans: are they the same?—Implica-tions in cancer translational research. J Nucl Med 2010;51:501–4.

12. Marsh V, Winton DJ, Williams GT, Dubois N, Trumpp A, SansomOJ, et al.Epithelial Pten is dispensable for intestinal homeostasis but suppressesadenoma development and progression after Apc mutation. Nat Genet2008;40:1436–44.

13. Davies E, Marsh V, Meniel V, Williams G, Clarke A.R. PTEN loss and KRASactivation leads to the formation of serrated adenomas and metastaticcarcinoma in the mouse intestine. J Pathol 2014;233:27–38.

14. Janssen KP, Alberici P, Fsihi H, Gaspar C, Breukel C, Franken P, et al. APCand oncogenic KRAS are synergistic in enhancing Wnt signaling in intes-tinal tumor formationandprogression.Gastroenterology 2006;131:1096–109.

15. Rahbari NN, Lordick F, Fink C, Bork U, Stange A, J€ager D, et al. Resection ofthe primary tumour versus no resection prior to systemic therapy inpatients with colon cancer and synchronous unresectable metastases(UICC stage IV): SYNCHRONOUS–a randomised controlled multicentretrial (ISRCTN30964555). BMC Cancer 2012;12:142.

16. Cancer Genome Atlas Network. Comprehensive molecular characteriza-tion of human colon and rectal cancer. Nature 2012;487:330–7.

PI3K and MEK Inhibitors in Genetic Models of Colorectal Cancer

www.aacrjournals.org Mol Cancer Ther; 14(10) October 2015 2185

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 12: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

17. Wood LD, Parsons DW, Jones S, Lin J, Sj€oblom T, Leary RJ, et al. Thegenomic landscapes of human breast and colorectal cancers. Science2007;318:1108–13.

18. Yip WK, Choo CW, Leong VC, Leong PP, Jabar MF, Seow HF. Molecularalterations of Ras-Raf-mitogen-activated protein kinase and phosphatidy-linositol 3-kinase-Akt signaling pathways in colorectal cancers from atertiary hospital at Kuala Lumpur, Malaysia. APMIS 2013;121:954–66.

19. Neumann J, Wehweck L, Maatz S, Engel J, Kirchner T, Jung A. Alterations inthe EGFR pathway coincide in colorectal cancer and impact on prognosis.Virchows Arch 2013;463:509–23.

20. Chong ML, Loh M, Thakkar B, Pang B, Iacopetta B, Soong R. Phosphati-dylinositol-3-kinase pathway aberrations in gastric and colorectal cancer:meta-analysis, co-occurrence and ethnic variation. Int J Cancer 2014;134:1232–8.

21. Perrone F, Lampis A, OrsenigoM, Di BartolomeoM, Gevorgyan A, LosaM,et al. PI3KCA/PTEN deregulation contributes to impaired responses tocetuximab in metastatic colorectal cancer patients. Ann Oncol 2009;20:84–90.

22. Frattini M, Saletti P, Romagnani E, Martin V, Molinari F, Ghisletta M, et al.PTEN loss of expression predicts cetuximab efficacy inmetastatic colorectalcancer patients. Br J Cancer 2007;97:1139–45.

23. Kodaki T, Woscholski R, Hallberg B, Rodriguez-Viciana P, Downward J,Parker PJ. The activation of phosphatidylinositol 3-kinase by Ras. Curr Biol1994;4:798–806.

24. Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB, et al.Characterization of a 3-phosphoinositide-dependent protein kinase whichphosphorylates and activates protein kinase Balpha. Curr Biol 1997;7:261–9.

25. Rini BI. Temsirolimus, an inhibitor of mammalian target of rapamycin.Clin Cancer Res 2008;14:1286–90.

26. Medema RH, Kops GJ, Bos JL, Burgering BM. AFX-like Forkhead transcrip-tion factorsmediate cell-cycle regulation by Ras and PKB through p27kip1.Nature 2000;404:782–7.

27. Li Y, Dowbenko D, Lasky LA. AKT/PKB phosphorylation of p21Cip/WAF1enhances protein stability of p21Cip/WAF1 and promotes cell survival. JBiol Chem 2002;277:11352–61.

28. Cantley LC. The phosphoinositide 3-kinase pathway. Science 2002;296:1655–7.

29. Mayo LD, Donner DB. A phosphatidylinositol 3-kinase/Akt pathwaypromotes translocation of Mdm2 from the cytoplasm to the nucleus. ProcNatl Acad Sci U S A 2001;98:11598–603.

30. Osaki M, Kase S, Adachi K, Takeda A, Hashimoto K, Ito H. Inhibition of thePI3K-Akt signaling pathway enhances the sensitivity of Fas-mediatedapoptosis in human gastric carcinoma cell line, MKN-45. J Cancer ResClin Oncol 2004;130:8–14.

31. Van Der Heide LP, Hoekman MF, Smidt MP. The ins and outs of FoxOshuttling: mechanisms of FoxO translocation and transcriptional regula-tion. Biochem J 2004;380:297–309.

32. Wee S, Jagani Z, Xiang KX, Loo A, Dorsch M, Yao YM, et al. PI3K pathwayactivation mediates resistance to MEK inhibitors in KRAS mutant cancers.Cancer Res 2009;69:4286–93.

33. Chen Z, Cheng K, Walton Z, Wang Y, Ebi H, Shimamura T, et al. A murinelung cancer co-clinical trial identifies genetic modifiers of therapeuticresponse. Nature 2012;483:613–7.

34. Balmanno K, Chell SD, Gillings AS, Hayat S, Cook SJ. Intrinsic resistance tothe MEK1/2 inhibitor AZD6244 (ARRY-142886) is associated with weakERK1/2 signalling and/or strong PI3K signalling in colorectal cancer celllines. Int J Cancer 2009;125:2332–41.

35. Dancey JE, Chen HX. Strategies for optimizing combinations of molecu-larly targeted anticancer agents. Nat Rev Drug Discov 2006;5:649–59.

Mol Cancer Ther; 14(10) October 2015 Molecular Cancer Therapeutics2186

Raja et al.

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223

Page 13: Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in ... · Small Molecule Therapeutics Assessment of the In Vivo Activity of PI3K and MEK Inhibitors in Genetically Defined

2015;14:2175-2186. Published OnlineFirst July 23, 2015.Mol Cancer Ther   Meera Raja, Matt Zverev, Katja Seipel, et al.   Genetically Defined Models of Colorectal Cancer

Activity of PI3K and MEK Inhibitors inIn VivoAssessment of the

  Updated version

  10.1158/1535-7163.MCT-15-0223doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2015/07/23/1535-7163.MCT-15-0223.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/14/10/2175.full#ref-list-1

This article cites 35 articles, 13 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/14/10/2175.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mct.aacrjournals.org/content/14/10/2175To request permission to re-use all or part of this article, use this link

on January 15, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst July 23, 2015; DOI: 10.1158/1535-7163.MCT-15-0223