biomarker signatures correlate with clinical outcome in ...€¦ · 2015-03-26  · mol cancer...

10
Companion Diagnostics and Cancer Biomarkers Biomarker Signatures Correlate with Clinical Outcome in Refractory Metastatic Colorectal Cancer Patients Receiving Bevacizumab and Everolimus Yingmiao Liu 1 , Mark D. Starr 1 , John C. Brady 1 , Christel Rushing 2 , Anuradha Bulusu 2 , Herbert Pang 2,3 , Wanda Honeycutt 1 , Anthony Amara 1 , Ivy Altomare 1 , Hope E. Uronis 1 , Herbert I. Hurwitz 1 , and Andrew B. Nixon 1 Abstract A novel combination of bevacizumab and everolimus was evaluated in refractory colorectal cancer patients in a phase II trial. In this retrospective analysis, plasma samples from 49 patients were tested for over 40 biomarkers at baseline and after one or two cycles of drug administration. Analyte levels at baseline and change on-treatment were correlated with pro- gression-free survival (PFS) and overall survival (OS) using univariate Cox proportional hazard modeling. Multivariable analyses were conducted using Cox modeling. Signicant changes in multiple markers were observed following bevaci- zumab and everolimus treatment. Baseline levels of six markers signicantly correlated with PFS and OS, including CRP, Gro-a, IGFBP-1, TF, ICAM-1, and TSP-2 (P < 0.05). At C2D1, changes of IGFBP-3, TGFb-R3, and IGFBP-2 correlated with PFS and OS. Prognostic models were developed for OS and PFS (P ¼ 0.0002 and 0.004, respectively). The baseline model for OS consisted of CRP, Gro-a, and TF, while the on-treatment model at C2D1 included IGFBP-2, IGFBP-3, and TGFb-R3. These data demon- strated that multiple biomarkers were signicantly modulated in response to bevacizumab and everolimus. Several markers correlated with both PFS and OS. Interestingly, these markers are known to be associated with inammation and IGF sig- naling, key modulators of mTOR biology. Mol Cancer Ther; 14(4); 19. Ó2015 AACR. Introduction Colorectal cancer is the second leading cause of cancer-related death in the United States, and in 2015, the estimated number of new cases of colorectal cancer will reach 130,000 (1). Three anti- VEGF therapies are currently FDA approved for the treatment of metastatic colorectal cancer: bevacizumab (2), ziv-aibercept (3), and regorafenib (4). Among them, bevacizumab is the most extensively studied and has demonstrated signicant efcacy across a variety of cancers. In combination with chemotherapy, bevacizumab has been shown to improve outcomes in both the rst-line and second-line settings (2, 5, 6). However, the mechan- isms underlying tumor progression on anti-VEGF therapy remain largely unknown and more effective antiangiogenic strategies are still needed, as are biomarkers to select patients most likely to benet from these therapies (7). mTOR inhibition represents a theoretically attractive strategy to augment the antitumor effects of anti-VEGF therapy. mTOR is a serine/threonine kinase that regulates numerous cellular func- tions, including nutrient sensing and survival (8). mTOR also regulates angiogenesis and modulates inammation (9, 10). Importantly, mTOR has been shown to regulate hypoxia induc- ible factor (HIF1a; refs. 11, 12), a key player involved in resistance to anti-VEGF therapies. Everolimus is a derivative of the natural mTOR inhibitor rapamycin, with improved solubility, potency, and stability (13). In an initial phase I clinical trial that combined anti-VEGF and anti-mTOR inhibitors, greater antiangiogenic and antitu- mor effects were observed from the combination treatment than observed with either drug alone (14). Based upon the proposed mechanistic complement, as well as promising phase I data, a phase II study was conducted (15), testing the com- binatorial effect of bevacizumab and everolimus in refractory colorectal cancer patients. In this study, we observed that more than 20% of patients had stable disease of more than 6 months, suggesting a potential subset of patients who derived greater benet from this combination therapy. Notably, most patients enrolled in this trial were resistant to previous bevacizumab- containing therapies, suggesting the addition of everolimus delayed the onset of bevacizumab resistance and improved clinical outcomes (11, 12, 16). How to select the patients with the greatest potential to benet, and how to better understand 1 Department of Medicine, Division of Medical Oncology, Duke Univer- sity Medical Center, Durham, North Carolina. 2 Department of Biosta- tistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina. 3 School of Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Andrew Nixon, Duke University Medical Center, 395 MSRB, Research Drive, Durham, NC 27710. Phone: 919-613-7883; Fax: 919 668- 3925; E-mail: [email protected]. doi: 10.1158/1535-7163.MCT-14-0923-T Ó2015 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org OF1 Research. on October 17, 2020. © 2015 American Association for Cancer mct.aacrjournals.org Downloaded from Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Upload: others

Post on 03-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

Companion Diagnostics and Cancer Biomarkers

Biomarker Signatures Correlate with ClinicalOutcome in Refractory Metastatic ColorectalCancer Patients Receiving Bevacizumab andEverolimusYingmiao Liu1, Mark D. Starr1, John C. Brady1, Christel Rushing2, Anuradha Bulusu2,Herbert Pang2,3,Wanda Honeycutt1, Anthony Amara1, Ivy Altomare1, Hope E. Uronis1,Herbert I. Hurwitz1, and Andrew B. Nixon1

Abstract

A novel combination of bevacizumab and everolimus wasevaluated in refractory colorectal cancer patients in a phase IItrial. In this retrospective analysis, plasma samples from 49patients were tested for over 40 biomarkers at baseline andafter one or two cycles of drug administration. Analyte levels atbaseline and change on-treatment were correlated with pro-gression-free survival (PFS) and overall survival (OS) usingunivariate Cox proportional hazard modeling. Multivariableanalyses were conducted using Cox modeling. Significantchanges in multiple markers were observed following bevaci-zumab and everolimus treatment. Baseline levels of six markerssignificantly correlated with PFS and OS, including CRP, Gro-a,

IGFBP-1, TF, ICAM-1, and TSP-2 (P < 0.05). At C2D1, changesof IGFBP-3, TGFb-R3, and IGFBP-2 correlated with PFS and OS.Prognostic models were developed for OS and PFS (P ¼ 0.0002and 0.004, respectively). The baseline model for OS consistedof CRP, Gro-a, and TF, while the on-treatment model at C2D1included IGFBP-2, IGFBP-3, and TGFb-R3. These data demon-strated that multiple biomarkers were significantly modulatedin response to bevacizumab and everolimus. Several markerscorrelated with both PFS and OS. Interestingly, these markersare known to be associated with inflammation and IGF sig-naling, key modulators of mTOR biology. Mol Cancer Ther; 14(4);1–9. �2015 AACR.

IntroductionColorectal cancer is the second leading cause of cancer-related

death in the United States, and in 2015, the estimated number ofnew cases of colorectal cancer will reach 130,000 (1). Three anti-VEGF therapies are currently FDA approved for the treatment ofmetastatic colorectal cancer: bevacizumab (2), ziv-aflibercept (3),and regorafenib (4). Among them, bevacizumab is the mostextensively studied and has demonstrated significant efficacyacross a variety of cancers. In combination with chemotherapy,bevacizumab has been shown to improve outcomes in both thefirst-line and second-line settings (2, 5, 6). However, themechan-isms underlying tumor progression on anti-VEGF therapy remainlargely unknown and more effective antiangiogenic strategies are

still needed, as are biomarkers to select patients most likely tobenefit from these therapies (7).

mTOR inhibition represents a theoretically attractive strategy toaugment the antitumor effects of anti-VEGF therapy. mTOR is aserine/threonine kinase that regulates numerous cellular func-tions, including nutrient sensing and survival (8). mTOR alsoregulates angiogenesis and modulates inflammation (9, 10).Importantly, mTOR has been shown to regulate hypoxia induc-ible factor (HIF1a; refs. 11, 12), a key player involved in resistanceto anti-VEGF therapies. Everolimus is a derivative of the naturalmTOR inhibitor rapamycin, with improved solubility, potency,and stability (13).

In an initial phase I clinical trial that combined anti-VEGFand anti-mTOR inhibitors, greater antiangiogenic and antitu-mor effects were observed from the combination treatmentthan observed with either drug alone (14). Based upon theproposed mechanistic complement, as well as promising phaseI data, a phase II study was conducted (15), testing the com-binatorial effect of bevacizumab and everolimus in refractorycolorectal cancer patients. In this study, we observed that morethan 20% of patients had stable disease of more than 6 months,suggesting a potential subset of patients who derived greaterbenefit from this combination therapy. Notably, most patientsenrolled in this trial were resistant to previous bevacizumab-containing therapies, suggesting the addition of everolimusdelayed the onset of bevacizumab resistance and improvedclinical outcomes (11, 12, 16). How to select the patients withthe greatest potential to benefit, and how to better understand

1Department of Medicine, Division of Medical Oncology, Duke Univer-sity Medical Center, Durham, North Carolina. 2Department of Biosta-tistics and Bioinformatics, Duke University Medical Center, Durham,North Carolina. 3School of Health, Li Ka Shing Faculty ofMedicine,TheUniversity of Hong Kong, Hong Kong, China.

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

Corresponding Author: Andrew Nixon, Duke University Medical Center, 395MSRB, Research Drive, Durham, NC 27710. Phone: 919-613-7883; Fax: 919 668-3925; E-mail: [email protected].

doi: 10.1158/1535-7163.MCT-14-0923-T

�2015 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org OF1

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 2: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

the mechanisms of delayed resistance, remain two big chal-lenges in the field.

Our group has developed and applied a novel multiplex ELISAapproach that allows for a broad profiling of plasma markersrelated to angiogenesis and inflammation. This approach has thepotential to identify markers that may predict for greater or lesserbenefit from anti-VEGF therapies, other antiangiogenic agents,and combination regimens. Previously, these candidate markerswere tested in other clinical settings and specific markers havebeen identified to have potential prognostic and/or predictivevalue (17–20).

In this report, we applied the 41-analyte multiplex ELISAapproach to this phase II study of bevacizumab and everolimusin colorectal cancer. The intent was to identify candidate predic-tors of greater or lesser benefit from this regimen. Biomarker levelswere determined at baseline and on-treatment, and treatment-related changes were statistically analyzed. Baseline levels and on-treatment changes were correlated with clinical outcomes andprognostic models for OS and PFS were generated.

Materials and MethodsPatient selection, treatment, and outcome

Enrolled in this study were 50 metastatic colorectal cancerpatients (one patient censored for statistical analysis) who hadprogressed on, or could not tolerate all of the following standardof care treatments for metastatic colorectal cancer: fluoropyrimi-dines, oxaliplatin, irinotecan, bevacizumab, and cetuximab and/or panitumumab (if wild-type KRAS). This population washighly refractory, having progressed on a median of four priortreatments. It should be noted that 42 patients (84%) hadprogressed on prior bevacizumab-based therapies. Additionaldetails about the treatment regimen and clinical outcomesfor this study have previously been reported (15). Writteninformed consent was obtained from each patient regardingthe use of plasma for this correlative analysis. This study wasinstitutional review board (IRB) approved and registered withwww.clinicaltrials.gov (study number: NCT00597506). Thisretrospective analysis conforms to the reporting guidelinesestablished by the REMARK criteria.

Plasma collection, handling, and storageBlood was collected from each patient by venipuncture into a

sodium citrate vacutainer (BD Vacutainer, catalog # 369714), andmixed thoroughly. After mixing, the tubes were centrifuged at2,500� g for 15minutes. The top layer of plasma was transferredto a fresh tube and centrifuged one more time at 2,500� g for 15minutes. The double-spun, platelet-poor plasma was aliquoted,snap frozen, and stored at �80�C until use.

Multiplex and ELISA assaysAll biomarkers were measured using the SearchLight multiplex

platform (Aushon Biosystems, Inc.; Table 1), except for TGF-b R3(R&D Systems, Inc.), as previously described (18).

Statistical analysisTo evaluate on-treatment changes, L-ratio was calculated using

the formula: Log2 (post-treatment level/baseline level) for eachanalyte at two time points, cycle 2 day 1 (C2D1) and cycle 3 day 1(C3D1). Signed-rank tests were used to identify significantlymodulated markers upon treatment. Significantly modulated

markers with a P � 0.0001 were graphically illustrated usingWaterfall plots demonstrating the change from baseline to C2D1.

Spearman's rank correlations were calculated for all pairs ofanalytes at baseline, C2D1 and C3D1. Hierarchical clustering ofall markers at baseline and on-treatment was displayed asdendrograms.

Based upon their response criteria, patients were dividedinto progressive disease (PD) or stable disease/partial response(SD/PR) groups. Biomarker differences between these twopatient populations were analyzed and illustrated via Bees-warm plots to show the baseline level variations, as well as thedifferential modulation of each marker in response to treat-ment. Baseline biomarker levels, both as continuous valuesand dichotomized at the median were associated with clinicaloutcome using univariate Cox proportional hazards analysisfor each analyte for both PFS and OS. On treatment changes,represented by L-ratio 1 and L-ratio 2, were also associatedwith PFS and OS.

Multivariable analyses were performed using Cox proportionalhazardsmodels to generate baseline and on-treatment biomarkersignatures. The score selection method was used to control thenumber of markers in the signature and leave-one-out cross-validation was used to derive a prognostic signature using a SASmacro (21). Finally, Kaplan–Meier plots were used to illustratepatients' survival, and the survival curves of the low- and high-riskgroups were compared by Wilcoxon test.

ResultsChanges in biomarker levels in response to bevacizumab andeverolimus

Fifty refractory colorectal cancer patients received a combina-tion of bevacizumab (10 mg/kg/2w) and everolimus (5 or 10 mgdaily) in this single-arm, non-randomized phase II study (15). Toevaluate biomarker responses to treatment, each patient's base-line biomarker profile was used as his/her reference control.Plasma samples collected at baseline (BL), at C2D1, and atC3D1 were available for biomarker analysis from 49, 39, and25 patients, respectively.

In total, 41 biomarkers for each patient were analyzed. Threemarkers (FGFb, IL8, VEGF-C) were excluded from statisticalanalysis, as more than 10% samples fell below the limit ofdetection. The median levels, ranges, and fold changes frombaseline for each of the 38 biomarkers are shown in Table 1.Assays were highly reproducible with coefficients of variationgenerally in the 5%–20% range (data not shown). At C2D1,statistically significant changes were observed in 26 markers(P < 0.05), with 12 of them being highly statistical significant(P < 0.0001) (Supplementary Table S1). Among these 12markers, three were downregulated on treatment, includingAng-2, MCP-1, and VEGF-R2, whereas MMP-2, PAI-1 active,PAI-1 total, PlGF, SDF-1, VCAM-1, VEGF-A, VEGF-D, vWFincreased on treatment (Fig. 1). Many of the significant changesobserved with these 12 markers at C2D1 persisted at C3D1,with the direction of change being the same for every marker(Supplementary Table S1).

Correlation among biomarkersTo better understand the potential coregulation of specific

biomarkers, Spearman's rank correlationwas used to test pairwisecorrelations at baseline and on-treatment. Statistically significant

Liu et al.

Mol Cancer Ther; 14(4) April 2015 Molecular Cancer TherapeuticsOF2

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 3: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

pairs of baselinemarkers (correlation coefficients�0.7;P<0.001)included PAI-1 active and PAI-1 total, CRP and IL6, ICAM-1 andVCAM-1, VCAM-1 and TSP-2, ICAM-1 andCRP, ICAM-1 and TSP-2, PDGF-AA and PDGF-BB. Statistically significant pairs of on-treatment markers at C3D1 included CRP and IL6, TGF-b1 andPDGF-BB, PDGF-AA andPDGF-BB. All baseline andon-treatmentanalyte pairs were positively correlated, indicating that biomar-kers were either both high (increased) or both low (decreased).Hierarchical clustering maps for all markers were shown inSupplementary Fig. S1.

Biomarker difference between SD and PD patientsIn this clinical study of bevacizumab and everolimus, 23

patients (46%) exhibited stable disease (SD) as their bestresponse,with13of these patients (26%)exhibiting SD for greaterthan 6months, as specified by the Response Evaluation Criteria inSolid Tumors (RECIST version 1.0). The majority of these SDpatients achieved best responses between C2D1 and C3D1.Twenty-one patients (42%) had progress disease (PD) as their

best response on treatment with 17 patients (34%) progressedradiographically, whereas 4 patients (8%) progressed clinically.This created a balanced distribution of patients that could becategorized into SD and PD groups. After dichotomizing thepatients into responder (SD) and nonresponder (PD) groups,each marker was tested to evaluate whether there were anysignificant differences between these two groups. First, the base-line level of each marker was statistically compared between SDand PD groups, using Wilcoxon rank-sum tests. As shown in Fig.2A, baseline levels of 10 markers were significantly lower (P <0.05) in the SD group compared with the PD group, includingAng-2, CRP, D-Dimer, Gro-a, IGFBP-2, IL-6, TGF-b2, TSP-2,VCAM-1, and vWF. Note that the y-axis scale is log2, indicatingthat one unit of change reflects a 2-fold difference. After one cycleof treatment, IGFBP-3 and TGFb-R3 were observed to be upre-gulated in SD patients, as compared with very little changeobserved in the PD patients. Conversely, TSP-1 was downregu-lated in SD patients, compared with changes observed in PDpatients (Fig. 2B).

Table 1. Levels of biomarkers at baseline and on-treatment

Baseline (n ¼ 49) C2D1 (n ¼ 39) C3D1 (n ¼ 25)Biomarker Unit Median Range Median Range FC Median Range FC

Ang-2 pg/mL 371.9 146.3–7,573.8 289.2 113.5–946.3 0.8 233.7 91–1,045.8 0.7BMP-9 pg/mL 32 0.8–151.4 33.4 2.6–92.8 2.8 36 7.5–110.2 4.1CRP mg/mL 7.89 0.1–316.0 6.4 0.1–72.6 1.4 4.6 0.2–77.5 2.6D-dimer mg/mL 64.0 29.3–494.0 68.3 39.6–154.0 1.13 62.7 26.5–648.0 5.4Endoglin ng/mL 16.1 0.3–29.1 17.6 7.2–29.3 1.0 17.4 11.3–25.0 1.1E-Cadherin ng/mL 31.7 10.0–815.5 29.6 9.6–863.5 1.0 27.6 9.5–1,131.5 1.2E-Selectin ng/mL 105.1 38.0–274.8 92.6 41.5–278.8 0.95 80.0 38.3–289.5 0.9GRO-a pg/mL 34.7 12.5–166.8 34.7 13.7–120 1.34 37.9 12.6–164.2 1.7HGF pg/mL 621.5 58.7–31,799.1 920.8 69.7–14,238.6 2.4 599.2 242–13,304.3 2.9ICAM-1 ng/mL 496.7 203.4–1,600.4 530.9 218.8–1,478.7 1.1 417.6 145.3–1,132.7 1.0IGFBP-1 ng/mL 6.1 0.1–66.2 6.5 0.2–134.6 1.9 5.7 0.5–57.9 2.4IGFBP-2 ng/mL 633.0 8.0–1,360.4 572.6 98.4–2,171.6 0.9 541.9 151.4–1,688.6 1.2IGFBP-3 ng/mL 250.1 1.6–924.6 305.3 56.4–958.4 1.3 308.3 107.6–876.4 1.3IL-6 pg/mL 24.6 1.0–135.4 37.1 3.5–226.2 1.9 26.0 4.2–144.0 2.1MCP-1 pg/mL 474.6 297.1–1,238.4 397.0 214.9–641.3 0.8 397.3 124.3–1,233.0 0.9MMP-2 ng/mL 101.1 53.7–157.7 136.6 85.2–230.3 1.3 130.4 62.9–228.0 1.2MMP-9 ng/mL 104.4 36.8–207.8 89.0 43.9–579.3 1.0 78.5 38.4–231.1 0.9OPN ng/mL 110.1 15.1–273.2 108.5 12.6–360.3 0.9 96.5 11.4–268.9 0.9PAI-1 active ng/mL 3.5 0.003–25.3 11.5 0.01–75.6 11.4 7.2 0.01–90.0 6.3PAI-1 total ng/mL 13.4 3.1–85.4 37.2 3.0–333.0 3.5 28.6 6.5–175.2 2.9PDGF-AA pg/mL 10.9 0.9–542.8 14.2 1.1–527.5 2.0 18.8 3.1–615.2 16.9PDGF-BB pg/mL 25.8 0.8–613.4 13.8 1.5–661.0 1.4 19.0 2.1–466.7 3.5PEDF mg/mL 3.6 2.4–5.6 4.1 2.5–5.4 1.1 4.0 2.2–4.9 1.0PIGF pg/mL 7.4 0.9–16.6 12.6 4.9–29.7 2.0 14.6 3.8–25.8 2.2P-Selectin ng/mL 128.9 39.7–576.1 104.5 39.6–440.4 0.9 96.6 47.3–544.2 1.0SDF-1 pg/mL 632.9 38.1–1,948.8 1,083.0 38.1–3,166.6 2.0 999.0 31.2–3,060.3 2.2TF pg/mL 37.8 2.9–510.2 43.4 2.9–105.1 1.3 42.8 2.9–96.6 1.6TGF-b1 ng/mL 10.2 4.4–105.3 12.8 5.6–84.2 1.4 11.7 3.9–83.7 1.9TGF-b2 pg/mL 28.5 3.0–1,182.2 33.5 9.4–1,651.8 1.5 29.5 7.5–944.3 1.6TGFb-R3 ng/mL 80.7 39.6–164.5 78.8 48.4–149.7 1.0 79.9 53.0–175.5 1.1TSP-1 ng/mL 14.9 2.9–181.7 12.6 2.9–1,167.2 7.7 16.1 1.3–388.7 3.6TSP-2 ng/mL 35.3 13.1–124.1 35.0 15.0–94.7 1.0 29.5 14.3–83.2 1.0VCAM-1 mg/mL 2.1 1.0–7.3 3.9 1.1–9.9 1.8 3.3 0.9–8.0 1.9VEGF pg/mL 38.6 3.0–1,098.4 120.5 22.4–709.0 4.2 108.3 27.8–2,93.1 4.0VEGF-D pg/mL 690.5 173.1–3,227.8 956.9 73.9–2,737.6 1.5 928.3 237.7–1,547.3 1.4VEGF-R1 pg/mL 67.1 6.0–10,004.5 52.3 6.0–1,612.5 1.5 35.0 11.4–6,570.4 7.6VEGF-R2 ng/mL 3.7 0.3–18.3 2.2 0.3–8.2 0.7 2.1 0.3–9.0 0.6vWF U/mL 20.1 2.0–217.8 24.9 0.8–159.8 1.4 24.2 8.4–51.2 1.6

Abbreviations: Ang-2, angiopoietin-2; BMP-9, bone morphogenetic protein 9; CRP, C-reactive protein; FC, fold change from baseline; GRO-a, growth-relatedoncogene-a; HGF, hepatocyte growth factor; ICAM-1, intercellular adhesion molecule-1; IGFBP, insulin-like growth factor binding protein; MCP-1, monocytechemotactic protein-1; MMP, matrix metallopeptidase; OPN, osteopontin; PAI-1, plasminogen activator inhibitor-1; PDGF, platelet-derived growth factor; PEDF,pigment epithelium–derived factor; PlGF, placenta growth factor; SDF-1, stromal cell–derived factor-1; TF, tissue factor; TSP, thrombospondin; VCAM-1, vascular celladhesion molecule-1; sVEGF-R, soluble VEGF receptor; vWF, won Willebrand factor.

Biomarkers Change in Response to Bevacizumab and Everolimus

www.aacrjournals.org Mol Cancer Ther; 14(4) April 2015 OF3

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 4: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

Univariate correlation of biomarkers with patient outcomeTo test the prognostic and predictive value of the markers,

baseline levels and on-treatment change for each marker wereassociatedwith PFS andOS, the primary and secondary endpointsof the clinical study, respectively. At baseline, eight markers weresignificantly associated with PFS (P < 0.05): CRP, Gro-a, IGFBP-1,TF, ICAM-1, vWF, TSP-2, andTGF-b1. In all cases except for IGFBP-1, the HRs of the significant markers were >1, indicating thathigher levels of the marker correlated with shorter PFS (Table 2).Baseline levels of eight analytes were significantly associated withOS: ICAM-1, CRP, Gro-a, TSP-2, IGFBP-1, TF, MCP-1, and Ang-2(P <0.05). Aswas seen for PFS, higher baseline levels for any givenmarker were associatedwith a shorter OS. The lists ofmarkers thatcorrelatedwith PFS andOSwere quite consistent as six of the eightmarkers were statistically significant for both OS and PFS (CRP,Gro-a, IGFBP-1, TF, ICAM-1, and TSP-2).

Next, the on-treatment change for each marker was associ-ated with PFS and OS. At C2D1, PFS was significantly associ-ated with changes in IGFBP-3, TGFb-R3, and IGFBP-2 (P �0.05), with greater changes in IGFBP-3 and TGFb-R3 levelsresulting in longer PFS, while a greater change in IGFBP-2 levelsresulted in shorter PFS times (Table 3). OS was significantlyassociated with the same three markers observed in PFS(IGFBP-3, TGFb-R3, IGFBP-2) as well as with three additionalmarkers (MMP-2, MMP-9, VEGF-R2; P < 0.05). MMP-9 was alsoassociated with PFS at the trend level (P ¼ 0.0736). At C3D1,only 26 samples were available from 19 SD patients and 7 PDpatients. PFS was significantly associated with changes in TSP-1,VEGF-R1, HGF, and IGFBP-1 (P<0.05), as greater changes inTSP-1 levels associated with longer PFS, while greater changesin VEGF-R1, HGF, and IGFBP-1 levels associated with shorterPFS time.

Figure 1.Change from baseline to the end of cycle 2 for biomarkers with statistical significance (P < 0.0001). � , censored patients. Gold lines represent patients whose PFS�median; black lines represent patients whose PFS < median.

Liu et al.

Mol Cancer Ther; 14(4) April 2015 Molecular Cancer TherapeuticsOF4

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 5: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

Multivariable prognostic modelsUsing a leave-one-out, cross-validation analysis, multivariable

models were developed. High- or low-risk grouping was assignedon the basis of relationship to the median of the combination ofbiomarkers and coefficients selected in each model, a higher riskcorresponding to a higher hazard. The full method of the LOOCVanalysis is described in the macro paper (21). At baseline, themodel for predicting OS benefit consisted of CRP, Gro-a, and TF.These markers were selected in at least 89.6% of the models. The

OS of the high- and low-risk groups were 4.2 and 11.5 months(P¼ 0.0002), respectively, and this difference corresponded to anHR of 2.9.

The on-treatmentmodel at C2D1 for PFS consisted of IGFBP-2,IGFBP-3, and TGFb-R3. These markers were selected in at least85%of themodels. The PFS of the high- and low-risk groups were1.9 and 4.6 months (P ¼ 0.004), respectively, and this differencecorresponded to an HR of 1.8. Kaplan–Meier plots of thesemodels are shown in Fig. 3.

Figure 2.Distinct biomarker features of PD versus SD patients. A, baseline level comparison. B, on-treatment change comparison.

Biomarkers Change in Response to Bevacizumab and Everolimus

www.aacrjournals.org Mol Cancer Ther; 14(4) April 2015 OF5

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 6: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

DiscussionWhen this trial was first initiated, it represented one of the

first doublet combinations of a VEGF- and an mTOR-inhibitorin patients with refractory colorectal cancer. Plasma sampleswere serially collected for each patient, allowing for correlativeanalyses that explore the changes that occurred on treatment,both in the setting of tumor control and progression. Amongthe 50 patients enrolled, sample collection was excellent (49/50patients at baseline) and most assays technically performedwell. Three markers (FGFb, IL8, VEGF-C) were generally read ator below the limit of detection, and as we took a conservativeapproach to biomarker discovery, these markers were elimi-nated from analysis. Our analyses were undertaken in anexploratory and hypothesis-generating fashion, and for thisreason, P values should be considered descriptive and havenot been corrected for multiple parameter testing. In addition,our study was not randomized and for this reason, correlationswith clinical outcomes cannot differentiate the prognosticversus predictive effects of each factor. Finally, this study did

not include a monotherapy group that would be needed toisolate the effects of one drug versus another.

These caveats notwithstanding, our current analysis identi-fied several key findings. First, a large number of the angiogenicand inflammatory markers that were evaluated were modulatedby bevacizumab plus everolimus treatment, suggesting broadbiologic consequences from this therapy. Of the 38 markersanalyzed, 26 were modulated in a statistically significant fash-ion. Consistent with prior reports by our group and othersevaluating anti-VEGFmonotherapy, in this study MMP-2, PlGF,VCAM-1, and VEGF-D were increased, while Ang-2, MMP-9,and s-VEGFR2 were decreased in response to the combinationof bevacizumab and everolimus. The magnitudes of thesechanges appear to be broadly consistent with those previouslydescribed for bevacizumab monotherapy (18, 22), suggestingthese changes were driven primarily by VEGF but not mTORinhibition.

Broad angiogenesis profiling has not been commonly reportedwithmTOR inhibitors. Severalmarkers were observed to change inresponse to the combinational treatment, but were not affected inprevious analyses of anti-VEGF therapies (18), possibly reflectingmTOR-specific effects. These markers included OPN and SDF-1,

Table 2. Correlation of biomarkers at baseline with clinical outcomes

Biomarker Pa <med vs. >med HR (95% CI)

BaselinePFS

CRP 0.0032 2.09 (1.15–3.83)GRO-a 0.0073 2.31 (1.28–4.18)IGFBP-1 0.008 0.92 (0.51–1.67)TF 0.0098 1.17 (0.65–2.08)ICAM-1 0.0117 1.66 (0.92–2.99)vWF 0.0335 1.08 (0.6–1.95)TSP-2 0.0455 1.63 (0.91–2.91)TGF-b1 0.0487 1.64 (0.91–2.93)

OSICAM-1 0.0009 2.02 (1.09–3.74)CRP 0.0009 2.2 (1.19–4.03)GRO-a 0.0048 2.5 (1.33–4.69)TSP-2 0.0058 2.9 (1.44–5.04)IGFBP-1 0.0126 1.3 (0.7–2.4)TF 0.021 1.47 (0.79–2.73)MCP-1 0.0458 1.4 (0.76–2.58)Ang-2 0.0491 1.89 (1.03–3.48)

aFrom Cox proportional hazard models using continuous analyte values.

Table 3. Correlation of biomarker on-treatment changeswith clinical outcomes

Biomarker Pa HRa (95% CI)

L-ratio 1PFSIGFBP-3 0.0014 0.36 (0.2–0.68)TGFbR3 0.0084 0.13 (0.03–0.59)IGFBP-2 0.0509 1.97 (0.99–3.9)

OSIGFBP-3 0.0014 0.30 (0.15–0.63)MMP-9 0.0015 3.18 (1.55–6.52)TGFb-R3 0.0026 0.12 (0.03–0.47)MMP-2 0.0184 7.62 (1.41–41.2)IGFBP-2 0.0247 2.40 (1.11–5.14)VEGF-R2 0.0255 0.61 (0.40–0.94)

L-ratio 2PFSTSP-1 0.0059 0.77 (0.64–0.92)VEGF-R1 0.0245 1.24 (1.02–1.5)HGF 0.038 1.29 (1.01–1.65)IGFBP-1 0.0484 1.31 (1.00–1.72)

aFrom Cox proportional hazard models using the calculated L-ratio values.

Figure 3.Kaplan–Meier estimates of outcomes using leave-one-out cross-validatedCox proportional hazard models. A, prognostic model for OS using baselinelevels consists of CRP, GRO-a, and TF. B, prognostic model for PFS usingon-treatment changes consists of IGFBP-2, IGFBP-3, and TGFb-R3.

Liu et al.

Mol Cancer Ther; 14(4) April 2015 Molecular Cancer TherapeuticsOF6

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 7: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

both of which are related to IGF signaling and inflammation,processes known to be regulated by mTOR (23, 24).

As we have seen in other similar studies, many markers appearto be highly coregulated, suggesting important cross-talk amonginflammatory cytokines, growth factors, and matrix-derivedangiogenic factors. The most significant correlations were notedforCRPand IL6, ICAM-1 andTSP-2, PAI-1 active andPAI-1 total atbaseline, aswell as for PDGF-AAandPDGF-BB,CRP and IL6, TGF-b1 and PDGF-BB at C3D1. These data are consistent with thecoregulation of the IL6 and TGF-b signaling axes in these patients,a finding well described in preclinical models (25, 26).

From the perspective of clinical utility, in unselected patients,the efficacy of bevacizumab and everolimus was modest inrefractory colorectal cancer and does not merit future testing.However, 23 patients (46%)had SDand among them, 13patients(26%) achieved SD on treatment for a period lasting more than 6months, suggesting the potential to identify a subset populationof patients who may benefit more from this therapy. Despitemany biologic and technical challenges, multiplex ELISAapproaches have recently identified several strong candidate pre-dictors of benefit from anti-VEGF therapy. For example, in twoseparate phase III trials in renal cell cancer patients, the inflam-matory mediator IL6 was shown to predict for benefit for pazo-panib and for bevacizumab (20, 27). Other candidate clinicalpredictors for bevacizumab have also been described, includingVEGF ligands, HGF, and other inflammatory mediators (28). Weand others have also described reproducible patterns of change inmultiple markers with anti-VEGF therapy, as well as with otherantiangiogenic therapies (17–20, 29–31).

To explore the relationship between these markers and clinicaloutcome, patients in this trial were retrospectively dichotomizedinto PD and SD groups. When biomarkers were comparedbetween these two groups, the baseline levels of most markerswere significantly lower in SD patients (Fig. 2A), including Ang-2,CRP, IL6, etc. In response to the coadministration of bevacizumaband everolimus, SD patients tended to have larger increases inIGFBP-3 and TGFb-R3, as well as greater decreases in TSP-1.

Significant differences in biomarker profiling between PDand SD groups encouraged the approach of using biomarkerinformation to select patients with better clinical benefit. In thistrial, baseline levels and on-treatment change for each markerwere correlated with clinical outcomes (Table 2 and 3). Severalstatistically significant candidate markers that could predict forbenefit from bevacizumab and everolimus treatment wereidentified. Six baseline markers were consistently correlatedwith both PFS and OS: CRP, Gro-a, IGFBP-1, TF, ICAM-1, andTSP-2. Additional markers such as vWF and TGF-b1 weresignificantly correlated only with PFS, whereas MCP-1 andAng-2 were significantly correlated only with OS, but trendswere also noted with PFS (MCP-1, P ¼ 0.076; Ang-2, P ¼0.118). Upon treatment, changes in 3 markers were consistentlyand significantly correlated with both PFS and OS: IGFBP-2,IGFBP-3, and TGFb-R3. IGFBPs and TGFb-R3, as well as VEGFand mTOR, are known to play key roles in immune modula-tion, suggesting that clinical outcomes may be linked to favor-able changes in host immune responses induced by anti-VEGFplus anti-mTOR treatment. These associations were largelyconsistent across both PFS and OS, suggesting their effects wererobust and biologically relevant. Intriguingly, both baselineand on-treatment markers appear to share a common associ-ation with inflammation (as exemplified by CRP) and the IGF

axis signaling (such as IGFBP-1, 2, 3), suggesting the clinicalimportance of these pathways.

Manyof the currentfindings appear to be novel. Themajority ofoutcome-relatedmarkers identified here have not been previouslyreported as candidate predictors for anti-VEGF therapy, and theirbiology may be more directly related to mTOR-regulated path-ways. As with VEGF inhibitors, the development of biomarkers topredict benefit from mTOR inhibitors has been challenging. Themarkers evaluated here have not been extensively profiled in thedevelopment of mTOR inhibitors. If confirmed, our currentfindings may help identify those patients most likely to benefitfrom combined anti-VEGF plus anti-mTOR therapy and perhapsfrom anti-mTOR therapy alone. By describing potentially target-able mechanisms of resistance to these agents, these findings mayalso suggest novel combination regimens.

Finally, we generated prognosticmodels for bothOSandPFS. Abaseline model consisting of CRP, Gro-a, and TF was developedfor OS (Fig. 3A). Notably, all three markers were correlated withPFS andOS in univariate analysis, and twoof them (CRP andGro-a) were found to be significantly higher in PD patients comparedwith SD patients. As discussed above, CRP is a well-knownmediator of acute phase inflammation, Gro-a has been suggestedto play a crucial role in chemotaxis (32), whereas mTOR is alsoknown to regulate many inflammatory and immune functions(33). The on-treatment changes of IGFBP-2, IGFBP-3, and TGFb-R3 were selected for PFSmodel (Fig. 3B). Consistently, changes inall these markers were correlated with PFS and OS by univariateanalysis. Two of them (IGFBP-3 and TGFb-R3) underwent largerincreases in SD patients. Not surprisingly, sensitivity to mTORinhibitorsmaybe affected bybasal signaling in the IGF axis andbyupregulation of these pathways by feedback loops induced bymTOR inhibition (34).

These biomarker data offer potential insight as to why com-bining everolimus to bevacizumab increased drug efficacy inpreviously refractory patients and generates new hypotheses toconsider. As discussed, downregulation ofOPN and upregulationof SDF-1 appear to be specifically driven by everolimus. Bothmarkers are known to be potent regulators of inflammatorydiseases, and notably, OPN is involved in regulating T helpertype 1 and Th17 cells (35). Interestingly, Th17 cells produce thecytokine IL17, whose role in promoting tumor resistance toantiangiogenic therapy has begun to unravel recently (36). Inaddition, multiple myeloid-derived inflammatory mediatorshave been shown to mediate resistance to anti-VEGF therapy inpreclinical models (36–38). Understanding how everolimus reg-ulates OPN in these specific cell populations, and the conse-quences of intracellular versus soluble OPN (39), is of greatinterest. Another crucial player is HIF-1, a well-studied transcrip-tional factor inducing VEGF production and conveying resistancefor antitumor drugs (40). Beyond its role in angiogenesis, HIF hasbeen shown to regulate, and be regulated by,manymetabolic andinflammatory mediators, including mTOR (11, 41, 42). Ever-olimus-mediated blockage of mTOR activity may downregulateHIF-1 function, potentially impacting bevacizumab resistance.

In summary, our plasma biomarker analysis in this phase IIstudy identified multiple angiogenic markers uniquely modulat-ed by the combination of bevacizumab and everolimus, exem-plified by several factors in the IGF axis and multiple inflamma-tion mediators. Many of these markers, both at baseline andon-treatment, were significantly correlated with clinical out-comes, by both univariate analysis and multivariable models.

Biomarkers Change in Response to Bevacizumab and Everolimus

www.aacrjournals.org Mol Cancer Ther; 14(4) April 2015 OF7

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 8: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

These findings support the increasingly appreciated roles ofinflammation in cancer biology and tumor angiogenesis.

Disclosure of Potential Conflicts of InterestI. Altomare is a consultant/advisory boardmember for Genentech. H.Uronis

reports receiving commercial research support from Genentech. H.I. Hurwitzreports receiving a commercial research grant from Genentech, commercialresearch support from Genentech, Roche, and Novartis, and is a consultant/advisory board member for Genentech and Roche. A.B. Nixon reports receivingcommercial research grants from F. Hoffman-LaRoche and Genentech. Nopotential conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: H. Pang, H.I. Hurwitz, A.B. NixonDevelopment of methodology: M.D. Starr, H. Pang, H.I. Hurwitz, A.B. NixonAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): Y. Liu, M.D. Starr, J.C. Brady, W. Honeycutt,I. Altomare, H. Uronis, H.I. Hurwitz, A.B. NixonAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y. Liu, M.D. Starr, C. Rushing, A. Bulusu, H. Pang,H.I. Hurwitz, A.B. Nixon

Writing, review, and/or revision of the manuscript: Y. Liu, M.D. Starr,C. Rushing, H. Pang, I. Altomare, H.I. Hurwitz, A.B. NixonAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): Y. Liu, W. Honeycutt, A. AmaraStudy supervision: H. Pang, H. Uronis, H.I. Hurwitz, A.B. Nixon

AcknowledgmentsThe authors thank the patients and their families who participated in this

study for their invaluable contributions. The authors also thank the DukeCancer Network and Duke University GI Oncology clinical trials team, withspecial recognition to Kathy Coleman and Neal Kaplan for plasma acquisitionand processing.

Grant SupportThis study was funded by Genentech/Roche and Novartis.The costs of publication of this articlewere defrayed inpart by the payment of

page charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received October 23, 2014; revised January 27, 2015; accepted February 4,2015; published OnlineFirst February 18, 2015.

References1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, ThunMJ. Cancer statistics, 2009. CA

Cancer J Clin 2009;59:225–49.2. Hurwitz H, Fehrenbacher L, NovotnyW, Cartwright T, Hainsworth J, Heim

W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin formetastatic colorectal cancer. N Engl J Med 2004;350:2335–42.

3. VanCutsem E, Tabernero J, Lakomy R, Prenen H, Prausova J, Macarulla T,et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecanimproves survival in a phase III randomized trial in patientswithmetastaticcolorectal cancer previously treated with an oxaliplatin-based regimen.J Clin Oncol 2012;30:3499–506.

4. Grothey A, VanCutsem E, Sobrero A, Siena S, Falcone A, Ychou M, et al.Regorafenib monotherapy for previously treated metastatic colorectalcancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013;381:303–12.

5. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ, Topalian SL,et al. A randomized trial of bevacizumab, ananti-vascular endothelial growthfactor antibody, for metastatic renal cancer. N Engl J Med 2003;349:427–34.

6. Llovet JM, Ricci S,Mazzaferro V,Hilgard P, Gane E, Blanc JF, et al. Sorafenibin advanced hepatocellular carcinoma. N Engl J Med 2008;359:378–90.

7. Jayson GC, Hicklin DJ, Ellis LM. Antiangiogenic therapy–evolving viewbased on clinical trial results. Nat Rev Clin Oncol 2012;9:297–303.

8. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metab-olism. Cell 2006;124:471–84.

9. Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy.Nat Rev Cancer 2004;4:335–48.

10. GuertinDA, SabatiniDM.Defining the role ofmTOR in cancer. Cancer Cell2007;12:9–22.

11. Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC, Kaper F, et al.Regulation of hypoxia-inducible factor 1alpha expression and function bythe mammalian target of rapamycin. Mol Cell Biol 2002;22:7004–14.

12. Zhong H, Chiles K, Feldser D, Laughner E, Hanrahan C, Georgescu MM,et al. Modulation of hypoxia-inducible factor 1alpha expression by theepidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAPpathway in human prostate cancer cells: implications for tumor angio-genesis and therapeutics. Cancer Res 2000;60:1541–5.

13. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al.Efficacy of everolimus in advanced renal cell carcinoma: a double-blind,randomised, placebo-controlled phase III trial. Lancet 2008;372:449–56.

14. Zafar Y, Bendell J, Lager J, YuD,GeorgeD,NixonA, et al. Preliminary resultsof a phase I study of bevacizumab (BV) in combination with everolimus(E) inpatientswith advanced solid tumors. J ClinOncol 2006;24:18s, 2006(suppl; abstr 3097).

15. Altomare I, Bendell JC, Bullock KE, Uronis HE, Morse MA, Hsu SD, et al. Aphase II trial of bevacizumab plus everolimus for patients with refractorymetastatic colorectal cancer. Oncologist 2011;16:1131–7.

16. George DJ, Kaelin WGJr. The von Hippel-Lindau protein, vascular endo-thelial growth factor, and kidney cancer. N Engl J Med 2003;349:419–21.

17. Liu Y, Starr MD, Brady JC, Dellinger A, Pang H, Adams B, et al. Modulationof circulating protein biomarkers following TRC105 (anti-endoglin anti-body) treatment in patients with advanced cancer. Cancer Med 2014;3:580–91.

18. Liu Y, Starr MD, Bulusu A, Pang H, Wong NS, Honeycutt W, et al.Correlation of angiogenic biomarker signatures with clinical outcomes inmetastatic colorectal cancer patients receiving capecitabine, oxaliplatin,and bevacizumab. Cancer Med 2013;2:234–42.

19. Nixon AB, Pang H, Starr MD, Friedman PN, Bertagnolli MM, Kindler HL,et al. Prognostic and predictive blood-based biomarkers in patients withadvanced pancreatic cancer: results from CALGB80303 (Alliance). ClinCancer Res 2013;19:6957–66.

20. Kluger HM, Halabi S, Solomon NC, Jilaveanu L, Zito C, Sznol J, et al.Prognostic and predictive tumor-based biomarkers in patients (pts) withadvanced renal cell carcinoma (RCC) treated with interferon alpha (IFN)with or without bevacizumab (Bev): Results from CALGB (Alliance)90206. J Clin Oncol 32:5s; 2014 (suppl; abstr 4532).

21. RushingC, Bulusu A,HurwitzHI,NixonAB, PangH. A leave-one-out cross-validation SAS macro for the identification of markers associated withsurvival. Comput Biol Med 2014;57C:123–9.

22. MurukeshN, Dive C, Jayson GC. Biomarkers of angiogenesis and their rolein the development of VEGF inhibitors. Br J Cancer 2010;102:8–18.

23. Rittling SR, Chambers AF. Role of osteopontin in tumour progression. Br JCancer 2004;90:1877–81.

24. Weekes CD, Song D, Arcaroli J, Wilson LA, Rubio-Viqueira B, Cusatis G,et al. Stromal cell-derived factor 1alpha mediates resistance to mTOR-directed therapy in pancreatic cancer. Neoplasia 2012;14:690–701.

25. Zhang XL, Topley N, Ito T, Phillips A. Interleukin-6 regulation oftransforming growth factor (TGF)-beta receptor compartmentalizationand turnover enhances TGF-beta1 signaling. J Biol Chem 2005;280:12239–45.

26. Yao Z, Fenoglio S, GaoDC, Camiolo M, Stiles B, Lindsted T, et al. TGF-betaIL-6 axis mediates selective and adaptive mechanisms of resistance tomolecular targeted therapy in lung cancer. Proc Natl Acad Sci U S A2010;107:15535–40.

27. Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin AM, et al.Prognosticorpredictiveplasma cytokines andangiogenic factors forpatientstreated with pazopanib for metastatic renal-cell cancer: a retrospectiveanalysis of phase 2 and phase 3 trials. Lancet Oncol 2012;13:827–37.

28. George B, Kopetz S. Predictive and prognostic markers in colorectal cancer.Curr Oncol Rep 2011;13:206–15.

29. Backen A, Renehan AG, Clamp AR, Berzuini C, Zhou C, Oza A, et al. Thecombination of circulating Ang1 and Tie2 levels predicts progression-free

Liu et al.

Mol Cancer Ther; 14(4) April 2015 Molecular Cancer TherapeuticsOF8

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 9: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

survival advantage in bevacizumab-treated patients with ovarian cancer.Clin Cancer Res 2014;20:4549–58

30. Hernandez-Yanez M, Heymach JV, Zurita AJ. Circulating biomarkers inadvanced renal cell carcinoma: clinical applications. Curr Oncol Rep2012;14:221–9.

31. Pena C, Lathia C, ShanM, Escudier B, Bukowski RM. Biomarkers predictingoutcome in patients with advanced renal cell carcinoma: Results fromsorafenib phase III Treatment Approaches in Renal Cancer Global Evalu-ation Trial. Clin Cancer Res 2010;16:4853–63.

32. Sager R, Haskill S, Anisowicz A, Trask D, Pike MC. GRO: a novel chemo-tactic cytokine. Adv Exp Med Biol 1991;305:73–7.

33. Cobbold SP. The mTOR pathway and integrating immune regulation.Immunology 2013;140:391–8.

34. Rodrik-Outmezguine VS, Chandarlapaty S, Pagano NC, Poulikakos PI,Scaltriti M, Moskatel E, et al. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov 2011;1:248–59.

35. Uede T. Osteopontin, intrinsic tissue regulator of intractable inflammatorydiseases. Pathol Int 2011;61:265–80.

36. ChungAS,WuX,ZhuangG,NguH,Kasman I, Zhang J, et al. An interleukin-17-mediated paracrine network promotes tumor resistance to anti-angio-genic therapy. Nat Med 2013;19:1114–23.

37. Qu X, Zhuang G, Yu L, Meng G, Ferrara N. Induction of Bv8 expression bygranulocyte colony-stimulating factor in CD11bþGr1þ cells: key role ofStat3 signaling. J Biol Chem 2012;287:19574–84.

38. Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, et al. Tumorrefractoriness to anti-VEGF treatment is mediated by CD11bþGr1þ mye-loid cells. Nat Biotechnol 2007;25:911–20.

39. Shinohara ML, Kim JH, Garcia VA, Cantor H. Engagement of the typeI interferon receptor on dendritic cells inhibits T helper 17 celldevelopment: role of intracellular osteopontin. Immunity 2008;29:68–78.

40. Warfel NA, El-Deiry WS. HIF-1 signaling in drug resistance to chemother-apy. Curr Med Chem 2014;21:3021–8.

41. Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors,inflammation, and immunity. Immunity 2014;41:518–28.

42. SemenzaGL. HIF-1mediatesmetabolic responses to intratumoral hypoxiaand oncogenic mutations. J Clin Invest 2013;123:3664–71.

www.aacrjournals.org Mol Cancer Ther; 14(4) April 2015 OF9

Biomarkers Change in Response to Bevacizumab and Everolimus

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T

Page 10: Biomarker Signatures Correlate with Clinical Outcome in ...€¦ · 2015-03-26  · Mol Cancer Ther; 14(4); 1–9. 2015 AACR. Introduction Colorectal cancer is the second leading

Published OnlineFirst February 18, 2015.Mol Cancer Ther   Yingmiao Liu, Mark D. Starr, John C. Brady, et al.   Bevacizumab and EverolimusRefractory Metastatic Colorectal Cancer Patients Receiving Biomarker Signatures Correlate with Clinical Outcome in

  Updated version

  10.1158/1535-7163.MCT-14-0923-Tdoi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2015/02/18/1535-7163.MCT-14-0923-T.DC1

Access the most recent supplemental material at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://mct.aacrjournals.org/content/early/2015/03/26/1535-7163.MCT-14-0923-TTo request permission to re-use all or part of this article, use this link

Research. on October 17, 2020. © 2015 American Association for Cancermct.aacrjournals.org Downloaded from

Published OnlineFirst February 18, 2015; DOI: 10.1158/1535-7163.MCT-14-0923-T