14
Review Mutational Landscape and Sensitivity to Immune Checkpoint Blockers Roman M. Chabanon 1,2 , Marion Pedrero 2 ,C eline Lefebvre 1,2 , Aur elien Marabelle 3,4 , Jean-Charles Soria 1,2,3 , and Sophie Postel-Vinay 1,2,3 Abstract Immunotherapy is currently transforming cancer treatment. Notably, immune checkpoint blockers (ICB) have shown unprec- edented therapeutic successes in numerous tumor types, includ- ing cancers that were traditionally considered as nonimmuno- genic. However, a signicant proportion of patients do not respond to these therapies. Thus, early selection of the most sensitive patients is key, and the development of predictive companion biomarkers constitutes one of the biggest challenges of ICB development. Recent publications have suggested that the tumor genomic landscape, mutational load, and tumor-specic neoantigens are potential determinants of the response to ICB and can inuence patients' outcomes upon immunotherapy. Further- more, defects in the DNA repair machinery have consistently been associated with improved survival and durable clinical benet from ICB. Thus, closely reecting the DNA damage repair capacity of tumor cells and their intrinsic genomic instability, the muta- tional load and its associated tumor-specic neoantigens appear as key predictive paths to anticipate potential clinical benets of ICB. In the era of next-generation sequencing, while more and more patients are getting the full molecular portrait of their tumor, it is crucial to optimally exploit sequencing data for the benet of patients. Therefore, sequencing technologies, analytic tools, and relevant criteria for mutational load and neoantigens prediction should be homogenized and combined in more inte- grative pipelines to fully optimize the measurement of such parameters, so that these biomarkers can ultimately reach the analytic validity and reproducibility required for a clinical imple- mentation. Clin Cancer Res; 22(17); 430921. Ó2016 AACR. Introduction Since their rst introduction into the clinic and rst approval in 2011 (1), immune checkpoint blockers (ICB) have transformed cancer treatment and allowed unprecedented improvements in overall survival (OS), progression-free survival (PFS), or overall response rates (ORR) in many aggressive diseases (2, 3). Most importantly, benets of ICB have not been limited to the "tradi- tional" immunogenic cancers, malignant melanoma and renal cell carcinoma (RCC), but have also been extended to other histologies classically described as "nonimmunogenic," such as nonsmall cell lung cancer (NSCLC) or mismatch-repairde- cient colorectal cancer (MMR-decient colorectal cancer; ref. 3). Despite these clear clinical advances, the biological mechanisms that underlie antitumor immunity and determine sensitivity to these agents, notably anti-programmed death receptor-1/-ligand 1 [antiPD-(L)1] are still poorly understood. Moreover, a statis- tically signicant proportion of patients, approximately 80%, among all tumor types included, still do not respond to these drugs, highlighting the urge for developing robust predictive biomarkers that would guide appropriate selection of patients. Recently, the tumor cell mutational burden has been correlated with clinical benets of antiPD-1 and anti-CTLA-4 therapy in various tumor types, including malignant myeloma (4, 5), NSCLC (6), and several DNA repairdecient tumors (79). Predicted neoantigen load has also emerged as an interesting selection biomarker for predicting clinical benet of these agents. Overall, a direct link between DNA repair deciency, mutational landscape, predicted neoantigen load, and clinical activity of ICB is suggested. In this review, we discuss the signicance and the relevance of this correlation in solid tumors. We also provide critical insight into the methods and techniques that have been used for per- forming analyses of tumor mutational burden, predicted neoan- tigen load, and neopeptide formation. We further propose a comprehensive approach that would allow encompassing other potential predictive biomarkers for response to antiPD-(L)1 inhibitors. Immune Escape and Carcinogenesis The original concept of immune surveillance, hypothesized in 1957 (10), and formally established in 1970 (11), postulated that the immune system alone could eliminate tumor cells in the early stages of carcinogenesis. Since then, this theory has been further enriched by the "immunoediting" notion (12), which describes how both innate and adaptive immunity contribute to carcino- genesis, notably by exerting a Darwinian selection pressure. Immunoediting classically consists of three distinct steps: (i) elimination: the innate and adaptive compartments coordinately 1 Facult e de M edicine, Universit e Paris Saclay, Universit e Paris-Sud, Le Kremlin Bic^ etre, France. 2 Inserm Unit U981, Gustave Roussy, Villejuif, France. 3 DITEP (D epartement d'Innovations Th erapeutiques et Essais Pr ecoces), Gustave Roussy,Villejuif, France. 4 Inserm Unit U1015, Gus- tave Roussy,Villejuif, France. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Sophie Postel-Vinay, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 98105, France. Phone: 3301-4211-43 43; Fax: 3301-4211-6444; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-16-0903 Ó2016 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 4309 on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

Review

Mutational Landscape and Sensitivity to ImmuneCheckpoint BlockersRoman M. Chabanon1,2, Marion Pedrero2, C�eline Lefebvre1,2, Aur�elien Marabelle3,4,Jean-Charles Soria1,2,3, and Sophie Postel-Vinay1,2,3

Abstract

Immunotherapy is currently transforming cancer treatment.Notably, immune checkpoint blockers (ICB) have shown unprec-edented therapeutic successes in numerous tumor types, includ-ing cancers that were traditionally considered as nonimmuno-genic. However, a significant proportion of patients do notrespond to these therapies. Thus, early selection of the mostsensitive patients is key, and the development of predictivecompanion biomarkers constitutes one of the biggest challengesof ICB development. Recent publications have suggested that thetumor genomic landscape, mutational load, and tumor-specificneoantigens are potential determinants of the response to ICB andcan influence patients' outcomes upon immunotherapy. Further-more, defects in theDNA repairmachinery have consistently beenassociated with improved survival and durable clinical benefit

from ICB. Thus, closely reflecting theDNAdamage repair capacityof tumor cells and their intrinsic genomic instability, the muta-tional load and its associated tumor-specific neoantigens appearas key predictive paths to anticipate potential clinical benefits ofICB. In the era of next-generation sequencing, while more andmore patients are getting the full molecular portrait of theirtumor, it is crucial to optimally exploit sequencing data for thebenefit of patients. Therefore, sequencing technologies, analytictools, and relevant criteria for mutational load and neoantigensprediction should be homogenized and combined in more inte-grative pipelines to fully optimize the measurement of suchparameters, so that these biomarkers can ultimately reach theanalytic validity and reproducibility required for a clinical imple-mentation. Clin Cancer Res; 22(17); 4309–21. �2016 AACR.

IntroductionSince their first introduction into the clinic and first approval in

2011 (1), immune checkpoint blockers (ICB) have transformedcancer treatment and allowed unprecedented improvements inoverall survival (OS), progression-free survival (PFS), or overallresponse rates (ORR) in many aggressive diseases (2, 3). Mostimportantly, benefits of ICB have not been limited to the "tradi-tional" immunogenic cancers, malignant melanoma and renalcell carcinoma (RCC), but have also been extended to otherhistologies classically described as "nonimmunogenic," such asnon–small cell lung cancer (NSCLC) or mismatch-repair–defi-cient colorectal cancer (MMR-deficient colorectal cancer; ref. 3).Despite these clear clinical advances, the biological mechanismsthat underlie antitumor immunity and determine sensitivity tothese agents, notably anti-programmed death receptor-1/-ligand1 [anti–PD-(L)1] are still poorly understood. Moreover, a statis-tically significant proportion of patients, approximately 80%,

among all tumor types included, still do not respond to thesedrugs, highlighting the urge for developing robust predictivebiomarkers that would guide appropriate selection of patients.Recently, the tumor cell mutational burden has been correlatedwith clinical benefits of anti–PD-1 and anti-CTLA-4 therapy invarious tumor types, including malignant myeloma (4, 5),NSCLC (6), and several DNA repair–deficient tumors (7–9).Predicted neoantigen load has also emerged as an interestingselection biomarker for predicting clinical benefit of these agents.Overall, a direct link between DNA repair deficiency, mutationallandscape, predicted neoantigen load, and clinical activity of ICBis suggested.

In this review, we discuss the significance and the relevance ofthis correlation in solid tumors. We also provide critical insightinto the methods and techniques that have been used for per-forming analyses of tumor mutational burden, predicted neoan-tigen load, and neopeptide formation. We further propose acomprehensive approach that would allow encompassing otherpotential predictive biomarkers for response to anti–PD-(L)1inhibitors.

Immune Escape and CarcinogenesisThe original concept of immune surveillance, hypothesized in

1957 (10), and formally established in 1970 (11), postulated thatthe immune system alone could eliminate tumor cells in the earlystages of carcinogenesis. Since then, this theory has been furtherenriched by the "immunoediting" notion (12), which describeshow both innate and adaptive immunity contribute to carcino-genesis, notably by exerting a Darwinian selection pressure.Immunoediting classically consists of three distinct steps: (i)elimination: the innate and adaptive compartments coordinately

1Facult�e de M�edicine, Universit�e Paris Saclay, Universit�e Paris-Sud, LeKremlin Bicetre, France. 2Inserm Unit U981, Gustave Roussy, Villejuif,France. 3DITEP (D�epartement d'Innovations Th�erapeutiques et EssaisPr�ecoces), Gustave Roussy,Villejuif, France. 4Inserm Unit U1015, Gus-tave Roussy, Villejuif, France.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Author: Sophie Postel-Vinay, Gustave Roussy, 114 Rue EdouardVaillant, Villejuif 98105, France. Phone: 3301-4211-43 43; Fax: 3301-4211-6444;E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-16-0903

�2016 American Association for Cancer Research.

ClinicalCancerResearch

www.aacrjournals.org 4309

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 2: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

drive immune rejection; (ii) equilibrium: through a clonal selec-tion process, the dynamic balance between tumor and immunecells results in the emergence of specific tumor cell variants withincreased resistance, which take advantage of acquiredmutations;(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses.

A variety of mechanisms can facilitate tumor immune escape(Fig. 1). Among them, deregulation of immune checkpoint sig-naling has been observed in multiple malignancies (13–21).Immune checkpoints involve the interaction between a receptorexpressed on T cells and its ligand located at the surface of antigen-presenting cells. This generates a costimulatory signal, whichtriggers either the activation or inhibition of T cells. Two majorcheckpoints regulate T-cell activation: (i) the CD28/CTLA-4 axis,which activates T cells upon engagement of CD28with CD80 and

CD86, and conversely inhibits T cells when CTLA-4 is engaged;and (ii) the PD-1 axis, which provides a strong inhibitory signalfollowing binding of PD-L1 or PD-L2 to the PD-1 receptor (22).Contrary to CTLA-4, PD-1 is thought to act predominantly in thetumor microenvironment, where PD-L1 is overexpressed by mul-tiple cell types, including dendritic cells, M2 macrophages, andtumor-associated fibroblasts (23).

As opposed to historical immune-based approaches that weredeveloped in traditionally immunogenic cancers, ICBs haveallowed significant therapeutic successes in many solid tumorsand hematologic malignancies. The anti-CTLA-4 ipilimumab(Yervoy, Bristol-Myers Squibb) was the first ICB to improve OSin malignant melanoma patients (1). In 2012, anti–PD-(L)1therapies including the anti–PD-1 pembrolizumab (Keytruda,Merck), and the anti-PD-L1 atezolizumab (MPDL-3280A,

© 2016 American Association for Cancer Research

Arginase 1TGFbIL10

NKCCL2

TNFa

CCL22

CCL5GM-CSF

CSF-1

IL4

IL1

CXCL15

CXCL12

COX-2FAS-L

TRAIL

PD-1

CTL

TCR

MHCclass I

Tumor cell

PD-L2

PD-L1

DR4/5 FAS

++++

mDC

4

2

5

3

1

PGE2

iDC

CXCL1

CXCL5

CXCL2

TregMDSCTAMTAN

Figure 1.

Mechanisms of immune escape in the tumor microenvironment. Several mechanisms, involving multiple immune components, contribute to tumor immune escape.(1) Immune recognition can be impaired following reduced expression of MHC class I molecules in malignant cells, resulting in decreased antigen presentationand consequently reduced detection by cytotoxic CD8þ T lymphocytes. (2) Cancer cells can activate immunosuppressive mechanisms by inducing immunecells' apoptosis through the expression of death signals (including FAS- and TRAIL-ligands). (3) Tumor cells release in the microenvironment a variety of immune-modulatory molecules that inhibit the immune system, such as IL6 and IL10, by inducing immunosuppressive Treg cells and MDSC, whereas the activity ofcytotoxic CD8þ T cells and NK cells is inhibited. (4) This cytokine imbalance, combined with the secretion of TGFb, COX-2, and PGE2, inhibits dendriticcell differentiation and maturation, thereby affecting antigen presentation and recognition by T cells. The release of additional immune modulators or metabolicregulators, such as IDO and arginase, also favors the establishment of an immunosuppressive tumor microenvironment. (5) Disrupted expression of immunecheckpoint ligands by cancer cells provides coinhibitory signals to CD4þ and CD8þ T lymphocytes, preventing them from building a specific antitumorimmune response. CCL, chemokine ligand; COX-2, cyclooxygenase-2; CXCL, chemokine (C-X-C motif) ligand; FAS-L, FAS-ligand; GM-CSF, granulocytemacrophage colony-stimulating factor; iDC, immature dentritic cell; IDO, indoleamine-2,3-deoxygenase; mDC, mature dentritic cell; MDSC, myeloid-derivedsuppressor cell; PD-1, programmed cell death 1; PD-L, programmed cell death ligand; PGE2, prostaglandin E2; TAN, tumor-associated neutrophil; TCR, T-cellreceptor; Treg, regulatory T cells.

Chabanon et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4310

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 3: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

Genentech/Roche), durvalumab (MEDI-4736, Astra Zeneca/MedImmune), and avelumab (MSB0010718C, Pfizer) enteredclinical development. Very promisingORR in relapsing/refractorymalignant melanoma, RCC, and NSCLC (3), associated withprolonged PFS and OS, led to their accelerated approval in2014–2015, and the outstanding activity observed in severalhistologies (Supplementary Table S1) awarded them "drugs ofthe year" in 2013 (24). Since then, an exponential number ofmonotherapy or combination trials have been launched inmultiple cancer types.

DNA Repair Deficiencies in CancerContrary to immune escape, DNA repair deficiency has been

successfully exploited as a therapeutic opportunity for more than50 years with the use of traditional cytotoxic chemotherapies. Ifthese DNA-damaging agents have initially been developed in a"one-size-fits-all" approach, DNA repair deficiencies are nowbeing exploited in a much more targeted fashion, notably usingtargetedmechanism-based approaches, such as synthetic lethality(25–28).

DNA repair deficiency is one of the main drivers of genomicinstability, a key hallmark of cancer (ref. 29; Table 1). It favors theaccumulation of DNA lesions that can arise from two distinctprocesses: (i) exogenous lesions, resulting from exposure tomutagenic agents and carcinogens and (ii) endogenous defects,which arise as a consequence of cell metabolism and the inherentinstability of DNA (30). Interestingly, some peculiar types ofexogenous DNA damage are associated with specific patterns ofmutations, also called mutational signatures. For example, thepredominance of C-to-A transitions, due to the effect of thepolycyclic hydrocarbons of tobacco smoke, is characteristicallyfound in NSCLC (31). In melanoma, UV radiation createspyrimidine dimers, which result in a high prevalence of C-to-T transitions on the untranscribed strand (32). Specificmutational signatures have also been reported in cancers withendogenous DNA damage repair defects, for example, BRCA1-or BRCA2-deficient high-grade serous ovarian and triple-nega-tive breast cancers, which harbor frequent loss of heterozygosity(28, 33); MMR-deficient colorectal cancer (34), associated witha microsatellite instable phenotype and high mutational bur-den; and POLE-deficient endometrial cancers, which exhibit anultra-mutated phenotype (7).

It is somehow intuitive that the presence of high tumormutational burden can increase the likelihood of neoantigensformation, and that the most mutated tumors may also be themost immunogenic ones (35). However, if high mutationalburden has repeatedly been associated with response andimproved outcome on ICB therapy, it would be na€�ve to con-clude on that basis that there is a general correlation betweenDNA repair deficiency and sensitivity to anti–PD-(L)1 (Fig. 2),the reality being much more complex.

Mutational Burden and Response to ICBThe description of a correlation between mutational load and

response to ICB was allowed by recent advances in next-genera-tion sequencing (NGS) technologies, notably whole-exomesequencing (WES) and RNA-sequencing (RNA-seq). High muta-tional load, defined as >100 nonsynonymous single-nucleotidevariants (nsSNV) per exome, was first associated with clinicalbenefit in melanoma patients treated with anti–CTLA-4 therapy

(4, 5). Subsequently, Rizvi and colleagues correlated high muta-tional load (defined as >178 nsSNVs per exome) and durableclinical benefit in two partially independent cohorts of NSCLCpatients receiving pembrolizumab (6). Of note, the studyreported a significantly increased ORR in tumors exhibiting asmoking molecular signature. Moreover, in responders showingthe highest mutational burden, specificmutations were identifiedin DNA repair genes, including POLD1, POLE, MSH2, BRCA2,RAD51C, and RAD17, thus supporting that DNA repair defectscan increase tumor immunogenicity by favoring somatic muta-tions. Consistently, later findings showed higher response ratesto anti–PD-1 therapy in MMR-deficient tumors (9, 36), and inBRCA2-mutatedmelanoma (37). Interestingly, in the latter study,mutational load did not correlate with tumor response but wasassociated with improved patient survival only, highlighting therole of additional factors influencing early tumor response andlong-term OS.

Now, the major challenges that remain to be addressed toimprove robustness of mutational burden include the definitionof optimal tumor purity and sequencing depth, as well as thethreshold for defining "high" and "low" mutational burden.Indeed, there is a significant overlap in mutation range betweenresponders and nonresponders (4, 5): some patients still benefitfrom ICB despite very low mutation rates, and conversely, highmutational load does not always correlate with response. This isbest illustrated by Hodgkin lymphoma, which is highly sensitiveto PD-1 blockade (38) despite carrying virtually no mutation.Mutational signatures, that are functional readouts of the past andcurrent disease biology in terms of DNA damage and DNA repair,could represent an additional genomic determinant of responseto ICB (35, 39). Their use, combined with evaluation of muta-tional load and detection of mutations in DNA repair genes, maytherefore allow better stratification of patients and identify ICB-sensitive tumors.

Importantly, the above-described analyses of the mutationallandscape only provide an "instantaneous and descriptive" pic-ture of a tumor genome. Even mutational signatures, in somecases, might exclusively reflect previous DNA repair deficienciesandmaynot be relevantmarkers of the actualDNA repair status ofthe tumor. It is therefore crucial to assess the potential for thesemutations to functionally enhance antitumor immune responsesby creating immunogenic neoantigens.

Predicted Neoantigen Load and Responseto ICB

Two main classes of tumor antigens are classically described:(i) tumor-associated antigens (TAA), which are nonmutated self-antigens that are aberrantly expressed by cancer cells followinggenetic and epigenetic alterations, and (ii) tumor-specific anti-gens, which are neoantigens that form as a result of nonsynon-ymous mutations and are generally unique to a tumor. Amongthese, the latter only have been consistently associated withantitumor T-cell reactivity and clinical efficacy of ICB (40).

Although we can anticipate that highly mutated tumors aremore prone to formneoantigens, the stochastic nature of neoanti-gen generation calls for a functional validation, as all formedneoantigensmay not be immunologically relevant. If it is obviousthat nsSNVs represent a mine of immunogenic mutations, frame-shift, splice site mutations, and intragenic fusions are also proneto generate neoepitopeswhennonfunctional proteins are directed

Mutational Landscape and Immunity in Cancer

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4311

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 4: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

Table 1. Type and frequency of DNA repair alterations in solid tumors

AlterationsCancer type Gene Type Frequency References

Non–small cell lung cancer BRCA1 Reduced mRNA and protein expression 44% (68)FANCF Promoter methylation 14% (69)ATM Somatic mutations 6% (69)MSH2 Reduced protein expression 18%–38% (69)ERCC1 Reduced protein expression 22%–66% (69)RRM1 Loss of heterozygosity 65% (69)

Small-cell lung cancer POLD4� Reduced mRNA expression N.R. (70)Clear-cell renal cell carcinoma ATM Somatic mutations 3% (71)

NSB1 Somatic mutations 0.5%MLH1 Homozygous deletion 3%–5% (72)MSH2 Promoter hypermethylation N.R. (73)

Urothelial carcinoma BRCA1 Somatic mutations 14% (74–76)BRCA2 Somatic mutations 14%PALB2 Somatic mutations 14%ATM Somatic mutations 29%MSH2 Loss of protein expression 3% (77)ERCC2 Somatic mutations 12% (78)

Head and neck cancer FANCB� Promoter methylation 31% (79)FANCF� Promoter methylation 15%FANCJ Reduced protein expression (IHC) N.R.FANCM Reduced protein expression (IHC) N.R.BRCA1 Reduced protein expression (IHC) N.R.BRCA2 Reduced protein expression (IHC) N.R.FANCD2 Reduced protein expression (IHC) N.R.

Ovarian cancer BRCA1/BRCA2 Germline mutations 15% (80, 81)Somatic mutations 35%Promoter methylation 11%–35%

FANCF Promoter methylation N.R.FANCD2 Reduced protein expression N.R.BARD1 Germline mutations 6% (82)BRIP1 Germline mutations 6%PALB2 Germline mutations 6%MRE11 Germline mutations 6%RAD50 Germline mutations 6%RAD51C Germline mutations 6%NSB1 Germline mutations 6%MSH6 Inactivating mutations 6% (82)

Triple-negative breast cancer BRCA1 Germline mutations 5%–10% (80, 83)BRCA2 Somatic mutations 10%

Gastric cancer MLH1 Loss of protein expression (IHC) 18% (84)Promoter hypermethylation 15%

MSH2 Loss of protein expression (IHC) 3%MMR-deficient colorectal cancer MRE11 Somatic mutations 75% (34, 85, 86)

RAD50 Somatic mutations 21%–46%BRCA2 Somatic mutations 2%MSH3 Somatic mutations 22%–51% (34, 85, 86)MSH6 Somatic mutations 9%–38%MLH3 Somatic mutations 9%–28%POLD3 Somatic mutations 37% (34, 85, 86)

Hepatocellular carcinoma NSB1 Somatic mutations 10% (87)MSH2 Promoter hypermethylation 25% (88, 89)

Reduced protein expression 18%PMS2 Promoter hypermethylation 15%MLH1 Promoter hypermethylation 8%

Reduced protein expression 38%Biliary tract cancer MSH2 Loss of protein expression (IHC) 7% (90, 91)

MSH6 Loss of protein expression (IHC) 7%MLH1 Loss of protein expression (IHC) 1.5%PMS2 Loss of protein expression (IHC) 1.5%

Prostate cancer BRCA2 Homozygous deletion/heterozygous deletion/frameshift mutation 14% (92, 93)ATM Frameshift mutation 12%PALB2 Frameshift mutation 4%CHK2 Homozygous deletion 4%FANCA Homozygous deletion 6%BRCA1 Homozygous deletion 2%MRE11 Frameshift mutation 2%NSB1 Frameshift mutation 2%MLH3 Frameshift mutation 4% (92, 93)

(Continued on the following page)

Chabanon et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4312

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 5: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

to the proteasome (41). The correlation between mutationalburden and predicted neoantigen load (as defined by the numberof neoantigens potentially presented by theMHC class I) has beenachieved by creating bioinformatics analysis pipelines thatmodelthe key steps of the antigen presentation process (Fig. 3 and Table2): (i) expression of mutated proteins that are processed by theproteasome, and produce neopeptides; (ii) translocation of theneopeptides through the endoplasmic reticulum and binding totheMHC class I molecule with a sufficient affinity to enable T-cellpresentation; and (iii) recognition of the presented neoantigen bya T-cell clone able to detect it.

This modeling pipeline has been overall successful in correlat-ing mutational load with predicted neoantigen load. First, muta-tional and predicted neoantigen loads were significantly corre-lated with clinical benefit in melanoma patients treated withipilimumab (5). Consistently, it was suggested that tumors dis-playing >10 nsSNVs/Mb may produce sufficient neoantigens togenerate ant-tumor immunogenicity, whereas tumors with <1nsSNV/Mb may not (41). Further consistent observations weremade in DNA repair–deficient tumors, including MSI-hightumors (7, 36),BRCA-mutated ovarian cancer (8), andmelanoma(37). However, genomic instability and accumulation of muta-tions is a double-edged sword process, which both favors thegeneration of immunogenic neopeptides, but also allows emer-gence of less immunogenic new clones that escape immunesurveillance, thereby favoring primary or acquired resistance.High intratumor heterogeneity (ITH) has indeed been correlatedwith poorer outcome, whereas sensitivity to ICB is associatedwithlow ITH and high clonal neoantigens (42). This underlines theparadoxical role of DNA repair defects in dictating response toICB. AlthoughDNA repair–deficient tumors exhibit high genomicinstability andhighmutational/neoantigen burdens, they are alsothe most likely to display high ITH due to their propensity toprovoke random mutations (43). If this observation represents astrong biological argument for treating patients with ICB early inthe course of the disease, when genomic instability is high, andITH low,we canhypothesize that each clonewithin the tumorwillretain somedegreeof intrinsic genomic instability, andparticipatein the generation of immunogenic neoantigens.

Therefore, a key issue is the determination of which epitopeswill actually prime T-cell responses, among the bulk of releasedepitopes. The study of a melanoma patient who experiencedcomplete response after 3 months of ipilimumab treatmentrevealed that, out of 1,657 nsSNVs, the tumor only displayed448 immunologically relevant epitopes, and nomore than two ofthemwere identified as able to trigger a patient-specific antitumorT-cell response (44). In a similar analysis, Rizvi and colleaguesdemonstrated that response to pembrolizumab in a NSCLC

patient was associated with the T-cell response against a singleneoantigen resulting from a nsSNV in HERC1 (6). In anotherstudy evaluating response to anti-CTLA-4 inmalignant myeloma,Snyder and colleagues identified a set of consensus tetrapeptidesequences exclusively shared by patients exhibiting long-termclinical benefit (4) and being necessary and sufficient for theactivation of an antitumor T-cell response; these results wereunfortunately not confirmed in two later studies (5, 37).

Mutational burden and predicted neoantigen load also shapethe nature and functional properties of antitumor immune infil-trates. The presence of tumor-infiltrating cytotoxic T-lymphocytes(CTL) has been correlated to higher immunogenic mutation rate,using RNA-seq data (45). Rooney and colleagues furtherdescribed that predicted neoantigen load correlated with thecytolytic activity of intratumoral CTLs and natural killer (NK)cells (46), but that a given mutation rate was associated withdistinct cytolytic activities across different histologies. Forinstance, cervical cancers exhibit higher cytolytic activity thanmelanoma, although this cancer type is not as sensitive to ICB.This suggests that both tissue-specific and tumor-specific factorscontribute to immune escape regulation. Interestingly, this workalso proposed a model for correlating the subclonal evolution oftumor genetics with the cytolytic activity of surrounding CTLs andNK cells, thereby reinforcing the link between continuous tumorgenetics drift and immune escape.

Overall, the data presented above support that highmutationalburden associates with increased neoantigens formation andtumor immunogenicity. However, the very high attrition rate,from a high mutational burden to the very few neoepitopes thatwill eventually produce an antitumor immune response, illus-trates well the complexity of predicting tumor immunogenicityusing genomic data alone. Furthermore, other mechanisms,including oncogenic stress (47–49), secretion of immunosup-pressive cytokines (e.g., IL10; ref. 50), or downregulation ofMHCclass I (51), also modulate tumor immunogenicity, and muta-tional burden is only one component of the determinants of ICBsensitivity.

Other Biomarkers of Response toAnti–PD-(L)1"Tumor-related" biomarkers

Beyond tumor "antigenome," several biomarkers are beingdeveloped to predict response to anti–PD-(L)1 therapies(52, 53). The most promising and best validated one is probablyPD-L1 expression assessment by immunohistochemical stainingon tumor and/or tumor-infiltrating immune cells (3, 54–57).However, this biomarker currently lacks sensitivity—some

Table 1. Type and frequency of DNA repair alterations in solid tumors (Cont'd )

AlterationsCancer type Gene Type Frequency References

Endometrial cancer MLH1 Promoter hypermethylation 30% (7, 94)POLE Somatic mutations 10% (7, 94)

Pancreatic cancer BRCA2 Germline mutations 1.5% (68, 95)MSH2 Loss of protein expression (IHC) 15% (96)MSH6 Loss of protein expression (IHC) 15%MLH1 Loss of protein expression (IHC) 15%PMS2 Loss of protein expression (IHC) 15%

NOTE: Genes in blue are related to DSBR, in green to MMR, in red to NER, in orange to nucleotide synthesis, and in gray to DNA replication. Genes marked with anasterisk refer to data reported in cell lines only. Mutations or alterations in genes related to cell cycle are described in Supplementary Table S2.Abbreviations: DSBR, double-strand break repair; NER, nucleotide-excision repair; N.R., not reported.

Mutational Landscape and Immunity in Cancer

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4313

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 6: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

© 2016 American Association for Cancer Research

Exogenous stress: tobacco Exogenous stress: tobacco and HPV infection

Exogenous stress: tobacco

Exogenous stress: tobacco

Exogenous stress: UV rays

DNA repair defectsfrequency (%)

PositiveNegative

Anti–PD-(L)1 response rate (%)RespondersRefractory

DNA repair defects frequency (%)

A

B

Ant

i–PD

-(L)

1 res

pons

e ra

te (

%)

0% 24%

20%

80%45%

15%

5%

15%

85%

35%65%

40%

20%20%

0%32%

68%

80%80%

60%

95%

19%

82%

N.R.

22%

78%85%

55%

76%

TP53: 75%CCNE1: 10%

MYC: 20%

DSBR: 45%MMR: 25%NER: 30%

PTEN: 25%RRM1: 65%

ATM: 6%

DSBR: 10%MMR: 20%TP53: 50%

MMR: 5%TP53: 2%

PTEN: 15%CDKN2A: 30%

MMR: 50%MRE11: 75%

POLD3: 37%ATR: 44%

BRAF: 50%TP53: 20%

Exogenous stress: asbestos

70%

30%

17%50% 50%

10%

15%19%

76%

80%

20%

80%

20%

0%24%

76%

24%

81% 85%

15%

85%

90%

83%

17%

83%

65%35%

N.R.

N.R.

DSBR: 35%

MMR: 30%POLE: 10%

DSBR: 50%MMR: 6%

TP53: 67%

MMR: 10%

MMR: 15%TP53: 35%PTEN: 10%CDKN2A: 35%

TP53: 60%MDM2: 50%

DSBR: 20%TP53: 70%PTEN: 2%CDKN2A: 20%

DSBR: 15%TP53: 80%

DSBR: 20%MMR: 3%

ERCC2: 10%TP53: 60%PTEN: 15%

CDKN2A: 50%

DSBR: 2%MMR: 15%

SCLC

NSC

LC

Hep

atoc

ellu

lar

carc

inom

a

Clea

r-ce

ll R

CCPa

ncre

atic

canc

er

MM

R-d

efici

ent

CRC

Uro

thel

ial

carc

inom

a

Mel

anom

a

Pros

tate

canc

er

Endo

met

rial

canc

er

Ova

rian

canc

erB

iliar

y tr

act

canc

erG

astr

icca

ncer

Mes

othe

liom

a

TNB

C

Hea

d an

dne

ck c

ance

r

0%

5%

10%

15%

20%

25%

30%

35%

40%

45%

50%

0% 10% 20% 30% 40% 50% 60% 70%

Chabanon et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4314

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 7: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

PD-L1-negative patients consistently experience clinical benefit(58, 59), and specificity— not all PD-L1–positive tumors benefitfrom anti–PD-(L)1 therapy (2, 60). Furthermore, the parametersof PD-L1 staining scoring are highly variable, notably the anti-PD-L1 antibody (clone SP142 and clone SP2063, Ventana; and clone28-8 and clone 22C3, Dako), the platform (PD-L1 IHC pharDx,Dako; OptiView DAB IHC Detection Kit, Ventana), the cells ofinterest (cancer cells, stromal cells, immune tumor-infiltratingcells), the positivity threshold (1%, 5%, 10%, or 50%), as well asthe tumor material used for analysis (fresh versus archivedmaterial, and primary versusmetastatic tumor; ref. 61).Moreover,PD-L1 expression can be constitutive or inducible (e.g., INFg-mediated induction; ref. 62). Together, these elements representsignificant hurdles for reaching the reproducibility and analyticvalidity that is required for any companion biomarker develop-ment and clinical implementation.

"Immune-related" biomarkersBeyond tumor-related biomarkers, the exploration of immune

infiltrate characteristics may also provide interesting biomarkers.Analysis of pretreatment samples from melanoma and NSCLCpatients responding to pembrolizumab revealed higher CD8þ T-cell levels at the tumor-invasive margin, as compared with non-responders (63, 64). Some more complex immune signatureshave also been explored: for example, Ribas and colleaguesdescribed an immune gene expression signature associated withgain in both ORR and PFS in melanoma patients treated withpembrolizumab (65), which is being explored in other histolo-gies. More recently, an eight-gene signature reflecting preexistingimmunity, the "T-effector/IFNg signature," was explored in thephase II POPLAR trial. High signature expression levels appearedto predict OS (but not PFS or ORR) benefit in atezolizumab-treated patients (66).

Immunomonitoring strategies, that is, repeated assessmentof dynamic circulating biomarkers involved in immuneresponse, have also been proposed. These dynamic biomarkers,which include notably cytokines and inflammatory mediators(Supplementary Table S3; ref. 67), can be monitored at severaltimepoints on trial using a simple blood test. If these circulatingbiomarkers have not been robust enough so far to predictresponders to ICB (52), they clearly represent a powerful andpractical tool for monitoring patient response, and deserve assuch active investigation.

Anticipating primary treatment resistanceFinally, as is the case for any targeted therapy, and especially

considering the cost of ICB and their associated biomarkers, earlyprediction of resistance is key. The very recent work by Hugo and

colleagues in melanoma (37) identified a transcriptional signa-ture associated with resistance to anti–PD-1 therapy. Exclusivelyfound in the pretreatment tumors of nonresponding patients, this"innate anti–PD-1 resistance" (IPRES) is characterized by theupregulation of genes involved in the regulation of epithelial–mesenchymal transition (EMT), cell adhesion, extracellularmatrix remodeling, angiogenesis, and wound healing. Very inter-estingly, this signature was not predictive of resistance to anti–CTLA-4 therapy, but found at variable frequencies across mostcommon cancers, suggesting that some mechanisms of ICB resis-tance might be shared by different histologies.

In the aggregate, these data highlight that a comprehensiveand integrated approach, which would encompass tumorgenetics, immune checkpoint expression, microenvironmental,and immune-monitoring data, is highly needed to best selectpatients.

Conclusions and Future ChallengesHow could we improve and expand the use of DNA repair

deficiency, mutational burden, and predicted neoantigen loadfor selecting patients that are the most likely to benefit fromanti–PD-(L)1 therapy? Targeted sequencing of hotspot muta-tions in DNA repair gene panels provides useful but limitedinformation, as it misses nongenetic forms of DNA repairdefects (e.g., secondary to epigenetic alterations), and, mostimportantly, does not functionally evaluate the tumor DNArepair capacity. The decreasing costs and expanding availabilityof NGS technologies open interesting perspectives for theirbroader use in clinical routine, and mutational load is a simpleparameter that is easily calculated and technically reproducible,allowing the comparison and/or merging of various patientseries. We can therefore reasonably hope that, with increasingnumbers and open data sharing, relevant mutational thresh-olds for predicting sensitivity to anti–PD-(L)1 therapy, as wellas tumor purity and sequencing depth that are required, willbe soon better defined in a histotype-specific fashion. Thepipeline optimization and establishment of reference guide-lines for predicting neoantigen load will also accelerate theclinical implementation of the latter work. Together, especiallyif integrated with PD-L1 IHC scoring and signatures of primaryresistance, these data might rapidly become robust enough tobe clinically implemented.

However, several challenges will still need to be addressed:(i) tumor material is not always available, and efforts shouldbe made to develop equivalent assays on circulating biomar-kers, such as cell-free tumor DNA; (ii) tumor heterogeneityneeds to be anticipated (42); (iii) the immunogenic potential

Figure 2.DNA repair defects and their association with anti–PD-(L)1 efficacy in solid tumors. A, representation, per tumor type, of the median frequency of DNA repairdeficiency (yellow pie charts) and the median efficacy of anti-PD-(L)1 (blue pie charts). For each histology, the median rate of DNA repair defects wascalculated on the basis of literature data (see Table 1 for raw data). When DNA repair defects in distinct pathways were mutually exclusive, the sumof their frequency was taken; when overlaps were observed between several DNA repair defects, the median of all DNA repair defects was chosen.The frequency of additional defects in other genes relevant for DNA repair (i.e., genes involved in cell cycle regulation or DNA replication) were alsoevaluated and are depicted on the side of the pie chart graphs. Tumor types resulting from exposure to a mutagenic agent are highlighted by a skull. ORRreported in phase I, II, or III trials performed in the corresponding histologies were taken for estimating the efficacy of anti–PD-(L)1 inhibitors (seeSupplementary Table S1 for raw data). The data cut-off for collecting anti–PD-(L)1 efficacy was January 2016. B, scatter plot illustrating the lack ofstatistically significant correlation between DNA repair mutation frequency and response to anti–PD-(L)1 therapies, highlighting the need to take intoaccount additional parameters for predicting response to these drugs. DSBR, double-strand break repair; HPV, human papillomavirus; NER, nucleotide-excision repair; N.R., not reported; TNBC, triple-negative breast cancer.

Mutational Landscape and Immunity in Cancer

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4315

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 8: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

© 2016 American Association for Cancer Research

Golgiapparatus

MHC/peptidecomplex

DNA

RNA

TAP protein

Proteasome

cellu

lar e

vent

s

Somaticmutation 3 4 5 621

Mutationalprofiling

MSI profilingData

generationNetCHOPCterm

PCleavage

FragPredict

PCM SVMTAP

Neoantigen loadMutational load and genomic instability

NetMHCpan

NetMHC

SMM

ARB

MHC multimer

GenScript

OptiType

Polysolver

AthlatesPredTAPNetCHOP 20S

TechniquesAnalysis

Software

RNA-seq

CGH

WES

WGS

Expressionprofiling

Proteasomalprocessing prediction

HLAtyping

HLA-binding

prediction

Neoantigen synthesisand T-cell reactivity

analysis

TAP transportprediction

Transcriptioninto mutated

mRNA

Proteasomalprocessing of themutated protein

TAP-mediatedpeptide transportinto the ER lumen

T-cell recognitionof cell surfaceneoantigens

Binding ofpeptides to MHCclass I complex

Tech

niqu

es a

nd to

ols

Cand

idat

esEn

dpoi

nts

Pipe

line

Peptides

ER

Tumor cell

CTL

Figure 3.

Pipeline for the identification of immune-relevant neoantigens. The typical pipeline consists of six main steps: (1) Tumor mutational load and specific mutationsare identified using WES or WGS. Additional techniques such as CGH or MSI-profiling might be of interest to evaluate genomic instability but have not beenvalidated yet in this indication. Moreover,WES is always a required starting point as DNA sequence information is required for subsequent prediction tools. (2) UsingRNA-seq, previously generated sequencing data are filtered for gene expression to restrict neoantigen prediction to the set of translated mutations("expressed nsSNV"). Subsequently, predictions for (3) proteasomal processing and (4) TAP-mediated transport of peptides are completed using dedicatedalgorithms. (5) To predict binding of peptides on MHC class I molecules, the previously selected peptides are implemented in a dedicated software that infersbinding affinity to HLA molecules according to the HLA type of the patient. (6) Eventually, the predicted peptides may be synthesized to test for T-cellreactivity in vitro using the MHC multimer technology. Key technologies most often used in the literature are highlighted in bold. Techniques exclusively usedto measure genomic instability are presented in dotted rectangles. ARB, average relative binding; CGH, comparative genomic hybridization; SMM, stabilizedmatrix method; WGS, whole-genome sequencing.

Chabanon et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4316

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 9: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

Table 2. Advantages and drawbacks of the available techniques to identify immunogenic mutations/neoantigens

Technique or software Platform Strengths Weaknesses

Relevance forantigenomeprediction References

WGS Mutational profiling Both coding and noncoding DNAsequences are analysed.

Sequencing depth is usually low,which prevents detection of somesubclonal mutations.

þþ (97)

WES Mutational profiling Provides high sequence coverageacross exome, increasing reliabilityand ability to detect subclonalmutations.

(i) Only covers the �1% codingregions of the genome.

(ii) Some mutations may be misseddue to uneven capture efficiencyacross exons.

þþþ (97)

MSI profiling Microsatelliteinstability

Several methods all well-validated. Only provides information on themicrosatellite instability.

þ (9)

CGH Genomic instability Global picture of the overall genomicinstability.

(i) Only provides copy-numbervariations and translocations oflarge portions of the genome.

(ii) No access to the DNA sequence.

þ

RNA-seq Expression profilingand codingmutation analysis

(i) Focuses on translated mutationsonly, that are the most likely tohave functional consequences.

(ii) Analysis not restricted to knowngenes: potential for discoveringnovel transcripts, splice variants orfusions.

(iii) Possibility to correlate mutationaldata with gene expression.

(i) Access to matched normal is keybut cannot be achieved in manycases: hard to distinguish tumor-specific mutations frompolymorphisms.

(ii) Limited calling of mutationswithin RNA species due to theirlow levels, either because of lowlevel gene expression or becauseof mRNA stability.

þþþ (97)

NetCHOP 20SPCM (WAPP package)FragPredict (MAPPPpackage)

Proteasomalprocessingprediction trainedon in vitro data

N.R. Predictions from in vitro data do notcapture the full complexity ofproteasomal processing.

þ (98)

PCleavageNetCHOPCterm Proteasomal

processingprediction trainedon in vivo data

(i) In vivo data provide accurateprediction as predictions are madeon the entire processingmachinery (action of severalproteasomes, cytosolicproteases. . .)

(ii) May also capture transportefficiency.

N.R. þþþ (98, 99)

PredTAP TAP transportprediction

No comparative study available. No comparative study available. N.R. (98, 100)SVMTAP (WAPPpackage)SMM (stabilized matrixmethod)

Allele-specific HLAbinding affinityprediction

N.R. (ii) Does not account for non-linearities and interdependenciesbetween amino acids.

þ (101–103)

ARB average relativebinding (matrix-basedmethods)

NetMHC [artificial neuralnetworks (ANN)-basedmethod]

Allele-specific HLAbinding affinityprediction

Nonlinear model. Does not allow prediction for allknown HLA alleles.

þþ (103, 104)

NetMHCpan [Pan-specificartificial neuralnetworks (ANN)-basedmethod]

Pan-specific HLAbinding affinityprediction

(i) Allows predictions to be made forall known HLA Class I alleles,including alleles for which noprediction is available withNetMHC.

(ii) NetMHCpan is the best-performingmethod for allele-specific HLAbinding affinity prediction.

N.R. þþþ (105)

Athlates HLA typing N.R. (i) Early tool with lower accuracythan that of up-to-date tools.Restricted to the use ofWES data

þ (98)

Polysolver HLA typing (i) Provides improved retrieval andalignment of HLA reads.Polysolver infers HLA-typeinformation with 97% sensitivityand 98% precision from exome-capture sequencing data.

(ii) Allows identification of patient-specific mutations in HLA alleles.

(ii) Restricted to the use of WESdata

þþþ (106)

(Continued on the following page)

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4317

Mutational Landscape and Immunity in Cancer

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 10: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

of mutations other than nsSNVs (including fusion transcriptsand aberrantly expressed splice variants) requires furtherexploration; (iv) developing an integrated approach, thatwould also encompass tumor microenvironment and immuneinfiltrates characteristics, as well as immunomonitoring data,warrants further investigation; and (v) last but not least, cost-efficacy and health economics studies will be needed to deter-mine which approach will eventually be the most relevant andsustainable.

Together, these challenges open very stimulating perspectivesand one can be certain that several exciting revolutions are stillto come soon in immuno-oncology.

Disclosure of Potential Conflicts of InterestA. Marabelle is a consultant/advisory board member for Amgen, Biothera

Pharmaceuticals, GlaxoSmithKline, Lytix Biopharma, Nektar, Novartis, Pfi-zer, Roche/Genentech, and Seattle Genetics. J.-C. Soria is a scientificcofounder of Gritstone Oncology and is a consultant/advisory board mem-ber for AstraZeneca, MSD, Pfizer, and Roche. No potential conflicts ofinterest were disclosed by the other authors.

Grant SupportR.M. Chabanon was supported by the Fondation Philanthropia.

Received April 8, 2016; revised May 31, 2016; accepted May 31, 2016;published OnlineFirst July 7, 2016.

References1. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB,

et al. Improved survival with ipilimumab in patients with metastaticmelanoma. N Engl J Med 2010;363:711–23.

2. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al.Pembrolizumab versus ipilimumab in advanced melanoma. N Engl JMed 2015;372:2521–32.

3. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermottDF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody incancer. N Engl J Med 2012;366:2443–54.

4. Snyder A,Makarov V,Merghoub T, Yuan J, Zaretsky JM,Desrichard A, et al.Genetic basis for clinical response to CTLA-4 blockade in melanoma. NEngl J Med 2014;371:2189–99.

5. Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, et al.Genomic correlates of response to CTLA-4 blockade in metastatic mel-anoma. Science 2015;350:207–11.

6. Rizvi NA,HellmannMD, Snyder A, Kvistborg P,Makarov V, Havel JJ, et al.Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8.

7. Howitt BE, Shukla SA, Sholl LM, Ritterhouse LL,Watkins JC, Rodig S, et al.Association of polymerase e-Mutated and microsatellite-instable endo-

metrial cancers with neoantigen load, number of tumor-infiltratinglymphocytes, and expression of PD-1 and PD-L1. JAMA Oncol 2015;1:1319–23.

8. Strickland KC, Howitt BE, Shukla SA, Rodig S, Ritterhouse L, Liu JF, et al.Association and prognostic significance of BRCA1/2-mutation status withneoantigen load, number of tumor-infiltrating lymphocytes and expres-sion of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget2016;7:13587–98.

9. LeDT,Uram JN,WangH,Bartlett BR,KemberlingH,EyringAD, et al. PD-1blockade in tumors with mismatch-repair deficiency. N Engl J Med2015;372:2509–20.

10. Burnet M. Cancer; a biological approach. I. The processes of control. BrMed J 1957;1:779–86.

11. Burnet FM. The concept of immunological surveillance. Prog Exp TumorRes 1970;13:1–27.

12. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting.Annu Rev Immunol 2004;22:329–60.

13. Hino R, Kabashima K, Kato Y, Yagi H,NakamuraM,Honjo T, et al. Tumorcell expression of programmed cell death-1 ligand 1 is a prognostic factorfor malignant melanoma. Cancer 2010;116:1757–66.

Table 2. Advantages and drawbacks of the available techniques to identify immunogenic mutations/neoantigens (Cont'd )

Technique or software Platform Strengths Weaknesses

Relevance forantigenomeprediction References

OptiType HLA typing (i) Performs fully automated HLAtypingwith four-digit resolution onNGS data from RNA-Seq, WES andWGS technologies.

(ii) OptiType showed an accuracy of99.3% on two-digit-level and of97.1% on four-digit-level typingusing datasets of RNA-Seq, WESand WGS technologies.

(i) Zygosity detection occasionallyfails in cases where alleles withhigh sequence similarityconstitute a heterozygous locus.

(ii) Not able to resolve all ambigui-ties for every genotype.

þþþ (107)

MHC multimer technology T-cell reactivityanalysis

(i) Gold-standard assay to identifyimmunogenic peptides. Can beused to detect even lowfrequencies of antigen-specific Tcells on small amounts of clinicalmaterial.

(ii) "Peptide exchange technology"allows the production of largecollections containing a lot ofdifferent peptide–MHC complexesfor T-cell staining.

N.R. þþþ (44, 97)

NOTE: Multiple NGS technologies, bioinformatics tools, and pipelines are available to analyze tumor samples and predict immunogenic mutations/potentialneoantigens in patients (see corresponding steps in Fig. 3). Primarily, genomic data are generated using various NGS technologies, most frequently including WESandRNA-seq to integrate both nsSNVs andexpressed nsSNVs. These data are then analyzedusingdedicated prediction algorithms corresponding to each step of theneoantigen generation biological process. These filtering tools guide the selection of immunogenic neoantigens among the bulk of candidate neoantigens. Althoughofficial guidelines are currently lacking on which tool should preferably be used, most often used algorithms include NetCHOPCterm for proteasomal processingprediction and NetMHC/NetMHCpan for HLA binding prediction. Eventually, a functional validation may be performed using an in vitro T-cell reactivity assay tovalidate the immunogenicity of the predicted neoantigens.Abbreviations: CGH, comparative genomic hybridization; MSI, microsatellite instability; N.R., not reported; WGS, whole-genome sequencing.

Chabanon et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4318

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 11: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

14. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al.Colocalization of inflammatory response with B7-h1 expression inhuman melanocytic lesions supports an adaptive resistance mechanismof immune escape. Sci Transl Med 2012;4:127ra37.

15. Shi S-J, Wang L-J, Wang G-D, Guo Z-Y, Wei M, Meng Y-L, et al. B7-H1expression is associated with poor prognosis in colorectal carcinoma andregulates the proliferation and invasion ofHCT116 colorectal cancer cells.PLoS One 2013;8:e76012.

16. Mu CY, Huang JA, Chen Y, Chen C, Zhang XG. High expression of PD-L1in lung cancermay contribute to poor prognosis and tumor cells immuneescape through suppressing tumor infiltrating dendritic cells maturation.Med Oncol 2011;28:682–8.

17. Yang CY, Lin MW, Chang YL, Wu CT, Yang PC. Programmed cell death-ligand 1 expression in surgically resected stage i pulmonary adenocarci-noma and its correlationwithdrivermutations and clinical outcomes. EurJ Cancer 2014;50:1361–9.

18. Gao Q, Wang XY, Qiu SJ, Yamato I, Sho M, Nakajima Y, et al. Over-expression of PD-L1 significantly associates with tumor aggressivenessand postoperative recurrence in human hepatocellular carcinoma. ClinCancer Res 2009;15:971–9.

19. Kuang D-M, Zhao Q, Peng C, Xu J, Zhang J-P, Wu C, et al. Activatedmonocytes in peritumoral stroma of hepatocellular carcinoma fosterimmune privilege and disease progression through PD-L1. J Exp Med2009;206:1327–37.

20. Ke W, Kryczek I, Chen L, Zou W, Welling TH. Kupffer cell suppressionof CD8þ T cells in human hepatocellular carcinoma is mediated byB7-H1/programmed death-1 interactions. Cancer Res 2009;69:8067–75.

21. Konishi J, Yamazaki K, AzumaM,Kinoshita I, Dosaka-AkitaH,NishimuraM. B7-H1 expression on non-small cell lung cancer cells and its relation-ship with tumor-infiltrating lymphocytes and their PD-1 expression. ClinCancer Res 2004;10:5094–100.

22. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: acommon denominator approach to cancer therapy. Cancer Cell 2015;27:450–61.

23. Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generationimmunotherapy-inhibiting programmed death-ligand 1 and pro-grammed death-1. Clin Cancer Res 2012;18:6580–7.

24. Robert C, Soria JC, Eggermont AMM. Drug of the year: programmedDeath-1 receptor/Programmed Death-1 Ligand-1 receptor monoclonalantibodies. Eur J Cancer 2013;49:2968–71.

25. FarmerH,McCabeN, LordCJ, Tutt ANJ, JohnsonDA, Richardson TB, et al.Targeting the DNA repair defect in BRCA mutant cells as a therapeuticstrategy. Nature 2005;434:917–21.

26. Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, et al.Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA muta-tion carriers. N Engl J Med 2009;361:123–34.

27. Ledermann J,Harter P,GourleyC, FriedlanderM, Vergote I, RustinG, et al.Olaparib maintenance therapy in platinum-sensitive relapsed ovariancancer. N Engl J Med 2012;366:1382–92.

28. Lord CJ, Ashworth A. The DNA damage response and cancer therapy.Nature 2012;481:287–94.

29. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell2011;144:646–74.

30. Lindahl T. Instability and decay of the primary structure of DNA. Nature1993;362:709–15.

31. Pleasance ED, Stephens PJ, O'Meara S, McBride DJ, Meynert A, Jones D,et al. A small-cell lung cancer genomewith complex signatures of tobaccoexposure. Nature 2010;463:184–90.

32. Pfeifer GP, You Y-H, Besaratinia A.Mutations induced by ultraviolet light.Mutat Res 2005;571:19–31.

33. Timms KM, Abkevich V, Hughes E, Neff C, Reid J, Morris B, et al.Association of BRCA1/2 defects with genomic scores predictive of DNAdamage repair deficiency among breast cancer subtypes. Breast Cancer Res2014;16:1–9.

34. Hewish M, Lord CJ, Martin SA, Cunningham D, Ashworth A. Mismatchrepair deficient colorectal cancer in the era of personalized treatment. NatRev Clin Oncol 2010;7:197–208.

35. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SAJr, Behjati S, BiankinA V, et al. Signatures of mutational processes in human cancer. Nature2013;500:415–21.

36. Dudley JC, Lin M-T, Le DT, Eshleman JR. Microsatellite instabilityas a biomarker for PD-1 blockade. Clin Cancer Res 2016;22:813–20.

37. Hugo W, Zaretsky JM, Sun L, Song C, Moreno Homet B, Hu-Lieskovan S,et al. Genomic and transcriptomic features of response to anti-PD-1therapy in metastatic melanoma. Cell 2016;165:35–44.

38. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M,et al. PD-1 Blockade with nivolumab in relapsed or refractory Hodgkin'slymphoma. N Engl J Med 2015;372:311–9.

39. Helleday T, Eshtad S, Nik-Zainal S. Mechanisms underlying mutationalsignatures in human cancers. Nat Rev Genet 2014;15:585–98.

40. Lennerz V, Fatho M, Gentilini C, Frye RA, Lifke A, Ferel D, et al. Theresponse of autologous T cells to a human melanoma is dominat-ed by mutated neoantigens. Proc Natl Acad Sci U S A 2005;102:16013–8.

41. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy.Science 2015;348:69–74.

42. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK,et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity toimmune checkpoint blockade. Science 2016;351:1463–9.

43. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and con-sequences of genetic heterogeneity in cancer evolution. Nature 2013;501:338–45.

44. Van Rooij N, Van Buuren MM, Philips D, Velds A, Toebes M, Heems-kerk B, et al. Tumor exome analysis reveals neoantigen-specific T-cellreactivity in an ipilimumab-responsive melanoma. J Clin Oncol2013;31:439–42.

45. Brown SD, Warren RL, Gibb EA, Martin SD, Spinelli JJ, Nelson BH, et al.Neo-antigens predicted by tumor genome meta-analysis correlate withincreased patient survival. Genome Res 2014;24:743–50.

46. RooneyMS, Shukla SA,WuCJ, Getz G,HacohenN.Molecular and geneticproperties of tumors associated with local immune cytolytic activity. Cell2015;160:48–61.

47. Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Chris-tensen CL, et al. Activation of the PD-1 pathway contributes toimmune escape in EGFR-driven lung tumors. Cancer Discov2013;3:1355–63.

48. Azuma K, Ota K, Kawahara A, Hattori S, Iwama E, Harada T, et al.Association of PD-L1 overexpression with activating EGFR mutationsin surgically resected nonsmall-cell lung cancer. Ann Oncol 2014;25:1935–40.

49. Green MR, Monti S, Rodig SJ, Juszczynski P, Currie T, O'Donnell E, et al.Integrative analysis reveals selective 9p24.1 amplification, increased PD-1ligand expression, and further induction via JAK2 in nodular sclerosingHodgkin lymphoma and primary mediastinal large B-cell lymphoma.Blood 2010;116:3268–77.

50. Berger S, Siegert A, Denkert C, K€obel M, Hauptmann S. Interleukin-10 inserous ovarian carcinoma cell lines. Cancer Immunol Immunother2001;50:328–33.

51. Reichel J, Chadburn A, Rubinstein PG, Giulino-Roth L, Tam W, Liu Y,et al. Flow-sorting and exome sequencing reveals the oncogenomeof primary Hodgkin and Reed-Sternberg cells. Blood 2015;12:1061–72.

52. Herbst RS, Soria J-C, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al.Predictive correlates of response to the anti-PD-L1 antibodyMPDL3280Ain cancer patients. Nature 2014;515:563–7.

53. Yearley JH, Gibson C, Yu N. PD-L2 expression in human tumors: rele-vance to anti-PD1 therapy in cancer [abstract]. In: Proceedings of theEuropean Cancer Congress 2015; 2015 Sep 25–29; Vienna, Austria.Brussels (Belgium): European Cancer Organisation; 2015. Abstract nr18LBA.

54. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al.Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl JMed 2015;372:2018–28.

55. Brahmer JR, Rizvi NA, Lutzky J, Khleif S, Blake-Haskins A, Li X, et al.Clinical activity and biomarkers of MEDI4736, an anti-PD-L1 anti-body, in patients with NSCLC. J Clin Oncol 32:5s, 2014 (suppl; abstr8021^).

56. Soria JC, Cruz C, Bahleda R, Delord JP, Horn L, Herbst RS, et al. Clinicalactivity, safety, and biomarkers of PD-L1 blockade in non-small cell lungcancer (NSCLC): additional analyses from a clinical study of the

Mutational Landscape and Immunity in Cancer

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4319

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 12: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

engineered antibodyMPDL3280A (anti-PDL1) [abstract]. In: Proceedingsof the European Cancer Congress 2013; 2013 Sep 27–Oct 1; Amsterdam,the Netherlands. Brussels (Belgium): European Cancer Organisation;2013. Abstract nr 3408.

57. Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, et al. Association ofPD-1, PD-1 ligands, and other features of the tumor immune microen-vironment with response to anti-PD-1 therapy. Clin Cancer Res2014;20:5064–74.

58. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancerimmunotherapy. Mol Cancer Ther 2015;14:847–56.

59. Kerr KM, Tsao M-S, Nicholson AG, Yatabe Y, Wistuba II, Hirsch FR.Programmed death-ligand 1 immunohistochemistry in lung cancer: inwhat state is this art? J Thorac Oncol 2015;10:985–9.

60. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al.Nivolumab in previously untreated melanoma without BRAF mutation.N Engl J Med 2015;372:320–30.

61. Madore J, Vilain RE,Menzies AM, KakavandH,Wilmott JS, Hyman J, et al.PD-L1 expression in melanoma shows marked heterogeneity within andbetween patients: Implications for anti-PD-1/PD-L1 clinical trials. Pig-ment Cell Melanoma Res 2015;28:245–53.

62. MacMicking JD. Interferon-inducible effector mechanisms in cell-auton-omous immunity. Nat Rev Immunol 2012;12:367–82.

63. Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJM, Robert L,et al. PD-1 blockade induces responses by inhibiting adaptive immuneresistance. Nature 2015;515:568–71.

64. Hu-Lieskovan S, Goldman J, Han M, Zaretsky J, Shintaku I, Wolf B,et al. High intratumoral T-cell infiltration correlated with mutationalload and response to pembrolizumab in non-small cell lung cancer[abstract]. In: Proceedings of the 16th World Conference on LungCancer; 2015 Sep 6–9; Denver, CO. Aurora (CO): InternationalAssociation for the Study of Lung Cancer; 2015. Abstract nrORAL3105.

65. Ribas A, Robert C, Hodi FS, Wolchok JD, Joshua AM, Hwu W-J, et al.Association of response to programmed death receptor 1 (PD-1)blockade with pembrolizumab (MK-3475) with an interferon-inflam-matory immune gene signature. J Clin Oncol 33, 2015 (suppl; abstr3001).

66. Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J,Mazieres J, et al. Atezolizumab versus docetaxel for patients withpreviously treated non-small-cell lung cancer (POPLAR): a multicen-tre, open-label, phase 2 randomised controlled trial. Lancet 2016;387:1837–46.

67. Manson G, Norwood J, Marabelle A, Kohrt H, Houot R. Biomarkersassociated with checkpoint inhibitors. Ann Oncol 2016;27:1199–206.

68. O'Sullivan CC, Moon DH, Kohn EC, Lee J, Sullivan CCO, Moon DH,et al. Beyond breast and ovarian cancers: PARP inhibitors for BRCAmutation-associated and BRCA-like solid tumors. Front Oncol 2014;4:42.

69. Postel-Vinay S, Vanhecke E, Olaussen KA, Lord CJ, Ashworth A, Soria J-C.The potential of exploiting DNA-repair defects for optimizing lung cancertreatment. Nat Rev Clin Oncol 2012;9:144–55.

70. Huang QM, Tomida S, Masuda Y, Arima C, Cao K, Kasahara TA, et al.Regulation of DNA polymerase POLD4 influences genomic instability inlung cancer. Cancer Res 2010;70:8407–16.

71. The Cancer Genome Atlas Network. Comprehensive molecular charac-terization of clear cell renal cell carcinoma. Nature 2013;499:43–9.

72. Feng C, Ding G, Jiang H, DingQ,WenH. Loss of MLH1 confers resistanceto PI3Kb inhibitors in renal clear cell carcinoma with SETD2 mutation.Tumor Biol 2015;36:3457–64.

73. Yoo KH, Won KY, Lim S-J, Park Y-K, Chang S-G. Deficiency of MSH2expression is associated with clear cell renal cell carcinoma. Oncol Lett2014;8:2135–9.

74. Nickerson ML, Dancik GM, Im KM, Edwards MG, Turan S, Brown J, et al.Concurrent alterations in TERT, KDM6A, and the BRCA pathway inbladder cancer. Clin Cancer Res 2014;20:4935–48.

75. Plimack ER, Dunbrack RL, Brennan TA, AndrakeMD, Zhou Y, SerebriiskiiIG, et al. Defects in DNA repair genes predict response to neoadjuvantcisplatin-based chemotherapy in muscle-invasive bladder cancer. EurUrol 2015;68:959–67.

76. Mullane SA, Werner L, Guancial EA, Lis RT, Stack EC, Loda M, et al.Expression levels of DNA damage repair proteins are associated with

overall survival in platinum-treated advanced urothelial carcinoma. ClinGenitourin Cancer 2016;14:352–9.

77. Bai S, Nunez AL, Wei S, Ziober A, Yao Y, Tomaszewski JE, et al. Micro-satellite instability and TARBP2 mutation study in upper urinary tracturothelial carcinoma. Am J Clin Pathol 2013;139:765–70.

78. Weinstein JN, Akbani R, Broom BM, Wang W, Verhaak RGW, McConkeyD, et al. Comprehensive molecular characterization of urothelial bladdercarcinoma. Nature 2014;507:315–22.

79. Stoepker C, Ameziane N, van der Lelij P, Kooi IE, Oostra AB, RooimansMA, et al.Defects in the Fanconi anemiapathway and chromatid cohesionin head-and-neck cancer. Cancer Res 2015;75:3543–5.

80. Lee JM, Ledermann JA, Kohn EC. PARP inhibitors for BRCA1/2 muta-tion-associated and BRCA-like malignancies. Ann Oncol 2014;25:32–40.

81. Burgess M, Puhalla S. BRCA 1/2-mutation related and sporadic breastand ovarian cancers: more alike than different. Front Oncol 2014;4:19.

82. Walsh T, Casadei S, Lee MK, Pennil CC, Nord AS, Thornton AM, et al.Mutations in 12 genes for inherited ovarian, fallopian tube, and perito-neal carcinoma identified by massively parallel sequencing. Proc NatlAcad Sci U S A 2011;108:18032–7.

83. Bryant C, Rawlinson R, Massey AJ. Chk1 inhibition as a novel therapeuticstrategy for treating triple-negative breast and ovarian cancers. BMCCancer 2014;14:570.

84. Bass AJ, ThorssonV, Shmulevich I, Reynolds SM,MillerM, Bernard B, et al.Comprehensive molecular characterization of gastric adenocarcinoma.Nature 2014;513:202–9.

85. Miquel C, Jacob S, Grandjouan S, Aime A, Viguier J, Sabourin JC, et al.Frequent alteration of DNA damage signalling and repair pathways inhuman colorectal cancers with microsatellite instability. Oncogene2007;26:5919–26.

86. The Cancer Genome Atlas Network. Comprehensive molecular char-acterization of human colon and rectal cancer. Nature 2012;487:330–7.

87. Wang Y, Hong Y, Li M, Long J, Zhao YP, Zhang JX, et al. Mutationinactivation of Nijmegen breakage syndrome gene (NBS1) in hepatocel-lular carcinoma and intrahepatic cholangiocarcinoma. PLoSOne 2013;8:e82426.

88. Hinrichsen I, KempM, Peveling-Oberhag J, Passmann S, Plotz G, ZeuzemS, et al. Promoter methylation of MLH1, PMS2, MSH2 and p16 is aphenomenon of advanced-stage HCCs. PLoS One 2014;9:e84453.

89. Wani Y, Notohara K, Tsukayama C, Okada S. Reduced expression ofhMLH1 and hMSH2 gene products in high-grade hepatocellular carci-noma. Acta Med Okayama 2001;55:65–71.

90. Moy AP, Shahid M, Ferrone CR, Borger DR, Zhu AX, Ting D, et al.Microsatellite instability in gallbladder carcinoma. Virchows Arch2015;466:393–402.

91. AbrahamSC, Lee JH, Boitnott JK, Argani P, Furth EE,WuTT.Microsatelliteinstability in intraductal papillary neoplasms of the biliary tract. ModPathol 2002;15:1309–17.

92. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al.DNA-repair defects and olaparib in metastatic prostate cancer. N Engl JMed 2015;373:1697–708.

93. Abeshouse A, Ahn J, Akbani R, Ally A, Amin S, Andry CD, et al. Themolecular taxonomy of primary prostate cancer. Cell 2015;163:1011–25.

94. The Cancer Genome Atlas Research NetworkIntegrated genomic charac-terization of endometrial carcinoma. Nature 2013;497:67–73.

95. Rustgi AK. Familial pancreatic cancer: genetic advances. Genes Dev2014;28:1–7.

96. Riazy M, Kalloger SE, Sheffield BS, Peixoto RD, Li-Chang HH, ScudamoreCH, et al. Mismatch repair status may predict response to adjuvantchemotherapy in resectable pancreatic ductal adenocarcinoma. ModPathol 2015;28:1383–9.

97. Heemskerk B, Kvistborg P, Schumacher TNM. The cancer antigenome.EMBO J 2013;32:194–203.

98. Backert L, Kohlbacher O. Immunoinformatics and epitope prediction inthe age of genomic medicine. Genome Med 2015;7:119.

99. Peters B, Bulik S, Tampe R, Van Endert PM, Holzh€utter H-G. IdentifyingMHC class I epitopes by predicting the TAP transport efficiency of epitopeprecursors. J Immunol 2003;171:1741–9.

Chabanon et al.

Clin Cancer Res; 22(17) September 1, 2016 Clinical Cancer Research4320

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 13: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

100. Zhang GL, Petrovsky N, Kwoh CK, August JT, Brusic V. PRED(TAP): asystem for prediction of peptide binding to the human transporterassociated with antigen processing. Immunome Res 2006;2:3.

101. Peters B, Sette A. Generating quantitative models describing the sequencespecificity of biological processeswith the stabilizedmatrixmethod. BMCBioinform 2005;6:132.

102. Bui HH, Sidney J, Peters B, Sathiamurthy M, Sinichi A, Purton KA, et al.Automated generation and evaluation of specificMHCbinding predictivetools: ARB matrix applications. Immunogenetics 2005;57:304–14.

103. Trolle T, Metushi IG, Greenbaum JA, Kim Y, Sidney J, Lund O, et al.Automated benchmarking of peptide-MHC class I binding predictions.Bioinformatics 2015;31:2174–81.

104. Lundegaard C, Lund O, Nielsen M. Accurate approximation methodfor prediction of class I MHC affinities for peptides of length 8, 10 and11 using prediction tools trained on 9mers. Bioinformatics 2008;24:1397–8.

105. Hoof I, Peters B, Sidney J, PedersenLE, Sette A, LundO, et al.NetMHCpan,a method for MHC class i binding prediction beyond humans. Immu-nogenetics 2009;61:1–13.

106. Shukla SA, RooneyMS, RajasagiM, TiaoG,Dixon PM, LawrenceMS, et al.Comprehensive analysis of cancer-associated somatic mutations in class IHLA genes. Nat Biotechnol 2015;33:1152–8.

107. Kohlbacher O. OptiType: precision HLA typing from next-generationsequencing data. Bioinformatics 2014;30:3310–6.

www.aacrjournals.org Clin Cancer Res; 22(17) September 1, 2016 4321

Mutational Landscape and Immunity in Cancer

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903

Page 14: Mutational Landscape and Sensitivity to Immune Checkpoint ......(iii) escape: the immune-resistant clones freely expand, circum-venting both innate and adaptive immune responses. A

2016;22:4309-4321. Published OnlineFirst July 7, 2016.Clin Cancer Res   Roman M. Chabanon, Marion Pedrero, Céline Lefebvre, et al.   BlockersMutational Landscape and Sensitivity to Immune Checkpoint

  Updated version

  10.1158/1078-0432.CCR-16-0903doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/22/17/4309.full#ref-list-1

This article cites 103 articles, 25 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/22/17/4309.full#related-urls

This article has been cited by 13 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://clincancerres.aacrjournals.org/content/22/17/4309To request permission to re-use all or part of this article, use this link

on January 21, 2021. © 2016 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst July 7, 2016; DOI: 10.1158/1078-0432.CCR-16-0903