throbber
Genetic Basis for Clinical Response
`to CTLA-4 Blockade in Melanoma
`Alexandra Snyder, M.D., Vladimir Makarov, M.D., Taha Merghoub, Ph.D.,
`Jianda Yuan, M.D., Ph.D., Jesse M. Zaretsky, B.S., Alexis Desrichard, Ph.D.,
`Logan A. Walsh, Ph.D., Michael A. Postow, M.D., Phillip Wong, Ph.D.,
`Teresa S. Ho, B.S., Travis J. Hollmann, M.D., Ph.D., Cameron Bruggeman, M.A.,
`Kasthuri Kannan, Ph.D., Yanyun Li, M.D., Ph.D., Ceyhan Elipenahli, B.S.,
`Cailian Liu, M.D., Christopher T. Harbison, Ph.D., Lisu Wang, M.D.,
`Antoni Ribas, M.D., Ph.D., Jedd D. Wolchok, M.D., Ph.D.,
`and Timothy A. Chan, M.D., Ph.D.
`
`ABSTR ACT
`
`Background
`Immune checkpoint inhibitors are effective cancer treatments, but molecular deter-
`minants of clinical benefit are unknown. Ipilimumab and tremelimumab are anti-
`bodies against cytotoxic T-lymphocyte antigen 4 (CTLA-4). Anti–CTLA-4 treatment
`prolongs overall survival in patients with melanoma. CTLA-4 blockade activates
`T cells and enables them to destroy tumor cells.
`
`Methods
`We obtained tumor tissue from patients with melanoma who were treated with
`ipilimumab or tremelimumab. Whole-exome sequencing was performed on tumors
`and matched blood samples. Somatic mutations and candidate neoantigens gener-
`ated from these mutations were characterized. Neoantigen peptides were tested for
`the ability to activate lymphocytes from ipilimumab-treated patients.
`
`Results
`Malignant melanoma exomes from 64 patients treated with CTLA-4 blockade were
`characterized with the use of massively parallel sequencing. A discovery set con-
`sisted of 11 patients who derived a long-term clinical benefit and 14 patients who
`derived a minimal benefit or no benefit. Mutational load was associated with the
`degree of clinical benefit (P = 0.01) but alone was not sufficient to predict benefit.
`Using genomewide somatic neoepitope analysis and patient-specific HLA typing,
`we identified candidate tumor neoantigens for each patient. We elucidated a neo-
`antigen landscape that is specifically present in tumors with a strong response to
`CTLA-4 blockade. We validated this signature in a second set of 39 patients with
`melanoma who were treated with anti–CTLA-4 antibodies. Predicted neoantigens
`activated T cells from the patients treated with ipilimumab.
`
`Conclusions
`These findings define a genetic basis for benefit from CTLA-4 blockade in melanoma
`and provide a rationale for examining exomes of patients for whom anti–CTLA-4
`agents are being considered. (Funded by the Frederick Adler Fund and others.)
`
`From the Department of Medicine (A.S.,
`T.M., M.A.P., J.D.W.), Human Oncology
`and Pathogenesis Program (A.S., V.M.,
`A.D., L.A.W., K.K., T.A.C.), Swim across
`America–Ludwig Collaborative Research
`Laboratory (T.M., Y.L., C.E., C.L., J.D.W.),
`Department of Radiation Oncology
`(T.A.C.), Department of Pathology
`(T.J.H.), and Immunology Program, Lud-
`wig Center for Cancer Immunotherapy
`(J.Y., P.W., T.S.H., J.D.W.), Memorial
`Sloan Kettering Cancer Center; Weill Cor-
`nell Medical College (A.S., M.A.P., J.D.W.,
`T.A.C.); and Department of Mathemat-
`ics, Columbia University (C.B.) — all in
`New York; the Department of Molecular
`and Medical Pharmacology (J.M.Z., A.R.)
`and the Department of Medicine, Divi-
`sion of Hematology–Oncology, Jonsson
`Comprehensive Cancer Center (A.R.),
`University of California, Los Angeles, Los
`Angeles; and Bristol-Myers Squibb,
`Princeton, NJ (C.T.H., L.W.). Address re-
`print requests to Dr. Chan at the Human
`Oncology and Pathogenesis Program,
`Memorial Sloan Kettering Cancer Center,
`1275 York Ave., Box 20, New York, NY
`10065, or at chant@mskcc.org; or to Dr.
`Wolchok at the Ludwig Center for Cancer
`Immunotherapy, Memorial Sloan Kettering
`Cancer Center, 1275 York Ave., New York,
`NY 10065, or at wolchokj@mskcc.org.
`
`Drs. Snyder, Makarov, Merghoub, and Yuan
`and Drs. Wolchok and Chan contributed
`equally to this article.
`
`This article was published on November
`19, 2014, and last updated on November
`12, 2015, at NEJM.org.
`
`N Engl J Med 2014;371:2189-99.
`DOI: 10.1056/NEJMoa1406498
`Copyright © 2014 Massachusetts Medical Society.
`
`2189
`
`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`original article
`
`
`
`Genome Ex. 1033
`Page 1 of 11
`
`

`

`Immune checkpoint blockade has led
`
`to durable antitumor effects in patients with
`metastatic melanoma, non–small-cell lung
`cancer, and other tumor types, but the factors
`determining whether a patient will have a re-
`sponse remain elusive.1,2 The fully human mono-
`clonal antibodies ipilimumab and tremelimumab
`block cytotoxic T-lymphocyte antigen 4 (CTLA-4),
`resulting in T-cell activation. Some studies have
`established correlations between outcomes with
`ipilimumab and peripheral-blood lymphocyte
`count, markers of T-cell activation,3 an “inflam-
`matory” microenvironment,4,5 and maintenance
`of high-frequency T-cell receptor clonotypes.6
`The relationship among the genomic land-
`scape of the tumor, the mutational load, and the
`benefit from treatment remains obscure. The im-
`munogenicity resulting from nonsynonymous
`melanoma mutations has been shown in a mouse
`model,7 and the antigenic diversity of human
`melanoma tumors has been modeled in silico8
`and in melanoma-specific CD8 T-cell responses
`after treatment with ipilimumab.9 Effector and
`helper T-cell function and regulatory T-cell de-
`pletion are necessary for the efficacy of CTLA-4
`blockade,10 but there is not an association be-
`tween a specific HLA type and a clinical bene-
`fit.11 Melanomas have very high mutational
`burdens (0.5 to >100 mutations per megabase)
`as compared with other solid tumors.12 Elegant
`studies have shown that somatic mutations can
`give rise to neoepitopes13 and that these may
`serve as neoantigens.14-16 We conducted a study
`to determine whether the genetic landscape of a
`tumor affects the clinical benefit provided by
`CTLA-4 blocking agents.
`
`Methods
`
`Sample Acquisition and DNA Preparation
`For the discovery set, we conducted whole-exome
`sequencing of DNA from tumors and matched nor-
`mal blood from 25 ipilimumab-treated patients. A
`validation set included an additional 39 patients, of
`whom 5 were treated with tremelimumab. Pri-
`mary tumor samples and matched normal pe-
`ripheral-blood specimens were obtained after the
`patients had provided written informed consent.
`DNA was extracted, and exon capture was per-
`formed with the use of the SureSelect Human All
`Exon 50-Mb kit (Agilent Technologies). Enriched
`
`exome libraries were sequenced on the HiSeq 2000
`platform (Illumina) to provide a mean exome cov-
`erage of more than 100× (Memorial Sloan Ketter-
`ing Cancer Center Genomics Core and Broad In-
`stitute).
`
`Immunogenicity Analysis of Somatic
`Mutations
`We created a bioinformatic tool to translate all
`mutations in exomes and then evaluate binding
`with major histocompatibility complex (MHC)
`class I molecules. The neoantigen signature was
`generated from the nonamers containing four
`amino acid strings of peptides that are common
`to tumors from patients with a long-term benefit
`from therapy. Details are provided in the Supple-
`mentary Appendix, available with the full text of
`this article at NEJM.org.
`
`Intracellular Cytokine Staining
`Candidate neoantigen peptides were synthesized
`(GenScript), cultured with autologous peripheral-
`blood mononuclear cells (PBMCs), and then ana-
`lyzed by means of intracellular cytokine staining
`for interleukin-2, CD107a, macrophage inflam-
`matory protein 1β, tumor necrosis factor α, and
`interferon-γ on restimulation of cells with the
`candidate peptides.
`
`Statistical Analysis
`The Mann–Whitney test was used to compare
`mutational loads, and the log-rank test was used
`to compare Kaplan–Meier curves. The statistical
`methods used in the study are more fully described
`in the Supplementary Appendix.
`
`R esults
`
`Mutational landscape of Melanomas
`from the Study Patients
`Baseline patient characteristics are shown in Ta-
`ble 1 (for more detailed information, see Tables
`S1 and S2 in the Supplementary Appendix). The
`study involved patients with and those without a
`long-term clinical benefit from therapy (CTLA-4
`blockade alone or CTLA-4 blockade with resec-
`tion of an isolated stable or nonresponding lesion).
`A long-term clinical benefit was defined by radio-
`graphic evidence of freedom from disease or evi-
`dence of a stable or decreased volume of disease
`for more than 6 months. Lack of a long-term ben-
`
`2190
`
`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 2 of 11
`
`

`

`Table 1. Clinical Characteristics of the Patients in the Discovery and Validation Sets, According to Clinical Benefit
`from Therapy.
`
`Characteristic
`
`Discovery Set
`
`Validation Set
`
`Long-Term
`Benefit
`(N = 11)
`
`Minimal or No
`Benefit
`(N = 14)
`
`Long-Term
`Benefit
`(N = 25)
`
`Minimal or No
`Benefit
`(N = 14)
`
`Age at start of treatment — yr
`
`Median
`
`Range
`
`Sex — no. of patients (%)
`
`Female
`
`Male
`
`Disease origin — no. of patients (%)
`
`Acral
`
`Uveal
`
`Cutaneous
`
`Unknown primary
`
`Not available
`
`BRAF or NRAS mutation — no. of
`patients (%)
`
`No
`
`Yes
`
`Lactate dehydrogenase level at start of
`therapy — no. of patients (%)
`
`63
`
`39–70
`
`3 (27)
`
`8 (73)
`
`0
`
`0
`
`10 (91)
`
`1 (9)
`
`0
`
`1 (9)
`
`10 (91)
`
`60
`
`48–79
`
`8 (57)
`
`6 (43)
`
`3 (21)
`
`0
`
`8 (57)
`
`3 (21)
`
`0
`
`6 (43)
`
`8 (57)
`
`66
`
`33–90
`
`9 (36)
`
`16 (64)
`
`1 (4)
`
`1 (4)
`
`15 (60)
`
`3 (12)
`
`5 (20)
`
`17 (68)
`
`8 (32)
`
`57
`
`18–74
`
`5 (36)
`
`9 (64)
`
`1 (7)
`
`0
`
`11 (79)
`
`0
`
`2 (14)
`
`11 (79)
`
`3 (21)
`
`8 (32)
`
`9 (64)
`
`Normal
`
`Above normal
`
`Not available
`
`Duration of response to therapy
` — wk
`
`Median
`
`Range
`
`Previous therapies — no.*
`
`Median
`
`Range
`
`Melanoma stage at time of diagnosis
`— no. of patients (%)
`
`IIIC
`
`M1a
`
`M1b
`
`M1c
`
`Overall survival — yr†
`
`Median
`
`Range
`
`8 (73)
`
`2 (18)
`
`1 (9)
`
`59
`
`42–361
`
`1
`
`0–3
`
`0
`
`0
`
`5 (45)
`
`6 (55)
`
`4.4
`
`2.0–6.9
`
`8 (57)
`
`5 (36)
`
`1 (7)
`
`14
`
`11–23
`
`1
`
`0–2
`
`0
`
`1 (7)
`
`1 (7)
`
`12 (86)
`
`0.9
`
`0.4–2.7
`
`3 (12)
`
`14 (56)
`
`130
`
`64–376
`
`0
`
`0–2
`
`3 (12)
`
`4 (16)
`
`2 (8)
`
`16 (64)
`
`3.3
`
`1.6–7.2
`
`3 (21)
`
`2 (14)
`
`11
`
`3–29
`
`0
`
`0–3
`
`0
`
`1 (7)
`
`3 (21)
`
`10 (71)
`
`0.8
`
`0.2–2.1
`
`* Previous therapies included interleukin-2 and cytotoxic chemotherapy.
`† Overall survival was calculated from the date of the first dose of ipilimumab to the date of death or censoring of data.
`
`2191
`
`Genetic Basis for Response to CTLA-4 Blockade
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 3 of 11
`
`

`

`the Supplementary Appendix). The wide ranges
`of mutational burdens (Fig. 2A, and Table S3 in
`the Supplementary Appendix) and recurrent and
`driver mutations (Fig. S2C and S2D and Table S4
`in the Supplementary Appendix) among samples
`were consistent with previously reported find-
`ings.17-19 The ratio of transitions to transversions
`(Fig. S2E in the Supplementary Appendix) and
`the frequency of nucleotide changes (Fig. S2F in
`the Supplementary Appendix) were similar in the
`discovery and validation sets.12 No gene was uni-
`versally mutated across patients with a sustained
`benefit.
`
`ASSOCIATION between Mutational Burden
`and Clinical Benefit
`We hypothesized that an increased mutational
`burden in metastatic melanoma samples would
`correlate with a benefit from CTLA-4 blockade.
`There was a significant difference in mutational
`load between patients with a long-term clinical
`benefit and those with a minimal benefit or no
`benefit, both in the discovery set (P = 0.01 by the
`Mann–Whitney test) and in the validation set
`(P = 0.009 by the Mann–Whitney test) (Fig. 2A, and
`Table S5 in the Supplementary Appendix). In the
`discovery set, a high mutational load was sig-
`nificantly correlated with improved overall sur-
`vival (P = 0.04 by the log-rank test) (Fig. 2B), and
`there was a trend toward improved survival in
`the validation set (Fig. S3A in the Supplementary
`Appendix). The latter set included eight patients
`with nonresponding tumors who otherwise had
`systemic disease control, which may confound
`the relationship between mutational load and
`survival. Further subdivision into four clinical
`categories was suggestive of a dose–response re-
`lationship in the discovery set (Fig. S3B in the
`Supplementary Appendix). These data indicate
`that a high mutational load correlates with a sus-
`tained clinical benefit from CTLA-4 blockade but
`that a high load alone is not sufficient to impart
`a clinical benefit, because there were tumors
`with a high mutational burden that did not re-
`spond to therapy.
`
`Somatic Neoepitopes in Responding Tumors
`and Efficacy of CTLA-4 Blockade
`MHC class I presentation and cytotoxic T-cell rec-
`ognition are required for ipilimumab activity.10
`Because mutational load alone did not explain a
`clinical benefit from CTLA-4 blockade, we hypoth-
`
`Figure 1. Paired Pretreatment and Post-Treatment Computed Tomographic
`Scans.
`In Panel A, the scans on the top were obtained on January 2, 2011, and Au-
`gust 26, 2013, and the scans on the bottom were obtained on September 6,
`2011, and January 14, 2013. In Panel B, the scans were obtained on August
`13, 2009, and January 9, 2010.
`
`efit was defined by tumor growth on every com-
`puted tomographic scan after the initiation of
`treatment (no benefit) or a clinical benefit lasting
`6 months or less (minimal benefit). Representa-
`tive scans are shown in Figure 1, and Figure S1
`in the Supplementary Appendix.
`To determine the genetic features associated
`with a sustained benefit from CTLA-4 blockade,
`we analyzed DNA in tumor and matched blood
`samples using whole-exome sequencing. In the
`discovery set, we generated 6.4 Gb of mapped
`sequence, with more than 99% of the target se-
`quence covered to at least 10× depth and a mean
`exome coverage of 103× (Table S3 and Fig. S2 in
`
`2192
`
`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 4 of 11
`
`

`

`A MutationalLoad
`
`P=0.009 by
`Mann–Whitney test
`
`Long-term
`benefit
`
`Minimal or
`no benefit
`
`1750
`
`1500
`
`1250
`
`1000
`
`750
`
`500
`
`250
`
`0
`
`P=0.01 by
`Mann–Whitney test
`
`Long-term
`benefit
`
`Minimal or
`no benefit
`
`1750
`
`1500
`
`1250
`
`1000
`
`750
`
`500
`
`250
`
`0
`
`No.ofExomicMissenseMutations
`
`DiscoverySet
`
`ValidationSet
`
`B SurvivalinDiscoverySet
`100
`
`>100 mutations (N=17)
`
`≤100 mutations (N=8)
`
`P=0.04 by
`log-rank test
`
`20
`
`40
`
`60
`
`80
`
`100
`
`Months
`
`80
`
`60
`
`40
`
`20
`
`0
`
`0
`
`Survival(%ofpatients)
`
`Figure 2. Mutational Landscape of Tumors According to Clinical Benefit
`from Ipilimumab Treatment.
`Panel A shows the mutational load (number of nonsynonymous mutations
`per exome) in the discovery and validation sets, according to status with re-
`spect to a clinical benefit from therapy. Panel B depicts the Kaplan–Meier
`curves for overall survival in the discovery set for patients with more than
`100 nonsynonymous coding mutations per exome and patients with 100 or
`fewer mutations.
`
`quency tetrapeptides in the combined exomes.
`Subsequent analysis is restricted to post-filtering
`peptides (see the Methods section in the Supple-
`
`2193
`
`esized that the presence of specific tumor neoan-
`tigens might explain the varied therapeutic ben-
`efit. To identify these neoepitopes, we developed
`a bioinformatic pipeline incorporating predic-
`tion of MHC class I binding, modeling of T-cell
`receptor binding, patient-specific HLA type, and
`epitope-homology analysis (see the Methods sec-
`tion and Fig. S4 in the Supplementary Appendix).
`We created a computational algorithm, called
`NAseek, to translate all nonsynonymous mis-
`sense mutations into mutant and nonmutant
`peptides (see the Methods section and Fig. S4 in
`the Supplementary Appendix). We examined
`whether a subgroup of somatic neoepitopes
`would alter the strength of peptide–MHC bind-
`ing, using patient-specific HLA types (Table S3
`in the Supplementary Appendix). We first com-
`pared the overall antigenicity trend of all mutant
`versus nonmutant peptides. In aggregate, the
`mutant peptides were predicted to bind MHC
`class I molecules with higher affinity than the
`corresponding nonmutant peptides (Fig. S5 in
`the Supplementary Appendix).
`Using only peptides predicted to bind to MHC
`class I molecules (binding affinity, ≤500 nM), we
`searched for conserved stretches of amino acids
`shared by multiple tumors. Using the methods
`described in the Methods section in the Supple-
`mentary Appendix, we identified shared, con-
`sensus sequences.20 We identified a number of
`tetrapeptide sequences that were shared by pa-
`tients with a long-term clinical benefit but com-
`pletely absent in patients with a minimal benefit
`or no benefit (Fig. 3A and 3B, and Table S6 in the
`Supplementary Appendix). It has been shown that
`short amino acid substrings comprise conserved
`regions across antigens recognized by a T-cell
`receptor.21 In these experiments, recognition of
`epitopes was driven by consensus tetrapeptides
`within the immunogenic peptides, and tetrapep-
`tides within cross-reacting T-cell receptor epit-
`opes were necessary and sufficient to drive T-cell
`proliferation, findings that are consistent with
`evidence that this polypeptide length can drive
`recognition by T-cell receptors.22 Tetrapeptides
`are used to model genome phylogeny because
`they occur relatively infrequently in proteins and
`typically reflect function.23
`We used the discovery set to generate a pep-
`tide signature from the candidate neoepitopes.
`This analysis initially pooled the aforementioned
`discovery and validation sets to remove low-fre-
`
`Genetic Basis for Response to CTLA-4 Blockade
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 5 of 11
`
`

`

`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`Figure 3. Association of a Neoepitope Signature
`with a Clinical Benefit from CTLA-4 Blockade.
`Candidate neoepitopes were identified by means of
`mutational analysis, as described in the Methods sec-
`tion in the Supplementary Appendix. Panel A shows a
`heat map of candidate tetrapeptide neoantigens that
`were present in patients with a long-term clinical ben-
`efit but absent in patients with a minimal benefit or
`no benefit in the discovery set (comprising 25 patients).
`Each row represents a neoepitope; each column repre-
`sents a patient. The vertical red line indicates the tetra-
`peptide signature associated with a response to block-
`ade of cytotoxic T-lymphocyte antigen 4 (CTLA-4).
`The exact tetrapeptides, chromosomal loci, and non-
`mutant and mutant nonamers in which they occur are
`listed in Table S6 in the Supplementary Appendix.
`Panel B shows the same information for the validation
`set (comprising 39 patients). Panel C shows the Kap-
`lan–Meier curves for overall survival in the discovery
`set for patients with the signature and those without
`the signature. Panel D shows the same data for the
`validation set.
`
`mentary Appendix). We found that the tetrapep-
`tides common to each group (candidate neoepi-
`topes) included 101 shared exclusively among
`patients in the discovery set who had a long-
`term clinical benefit; this was also indepen-
`dently observed in the validation set (Fig. 3A and
`3B, and Tables S6 and S7 in the Supplementary
`Appendix). This set of neoepitopes defines a
`signature linked to a benefit from CTLA-4
`blockade. Because of the size of our discovery
`set, we cannot exclude the possibility that addi-
`tional biologically relevant epitopes exist and
`conversely that there are biologically relevant
`epitopes that were predicted bioinformatically
`but were not expressed or presented in patients
`with a minimal benefit or no benefit (Tables S7A
`and S7B in the Supplementary Appendix).
`Shared tetrapeptide neoepitopes did not sim-
`ply result from a high mutational load. For ex-
`ample, in the discovery set, the patient with a
`minimal benefit or no benefit who had the
`greatest number of mutations (Patient SD7357,
`who had 1028 mutations) did not share any of
`the tetrapeptide signatures. This concept was
`illustrated again in the validation set, in which
`even tumors from patients with more than 1000
`mutations (Patients NR9521 and NR4631) did
`not respond (Table S3 in the Supplementary Ap-
`pendix). Simulation testing with five different
`
`A
`
`NeoepitopesinDiscoverySet
`Minimal
`Long-
`orNo
`Term
`Benefit
`Benefit
`
`B
`
`NeoepitopesinValidationSet
`Minimal
`Long-
`orNo
`Term
`Benefit
`Benefit
`
`NeoepitopeSignature
`
`NeoepitopeSignature
`
`C SurvivalinDiscoverySet
`100
`
`With signature (N=10)
`
`Without signature (N=15)
`
`P<0.001
`by log-rank
`test
`
`20
`
`40
`
`60
`
`80
`
`100
`
`Months
`
`80
`
`60
`
`40
`
`20
`
`0
`
`0
`
`Survival(%ofpatients)
`
`With signature (N=16)
`
`P=0.002
`by log-rank
`test
`
`Without signature (N=23)
`
`D SurvivalinValidationSet
`100
`
`80
`
`60
`
`40
`
`20
`
`Survival(%ofpatients)
`
`0
`
`0
`
`20
`
`40
`
`60
`Months
`
`80
`
`100
`
`AUTHOR:
`
`Snyder (Chan)
`
`FIGURE:
`
`REVISED 3 of 4 (10/22/15)
`
`2194
`
`ARTIST:
`
`ts
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`AUTHOR, PLEASE NOTE:
`
`Fig
`
`Iss
`
`
`
`Genome Ex. 1033
`Page 6 of 11
`
`

`

`models showed that the association between the
`neoepitope signature and a long-term clinical
`benefit was highly significant and was unlikely
`to have resulted from chance alone (P<0.001 for
`four methods and P = 0.002 for a fifth method)
`(Fig. S6 in the Supplementary Appendix). A high
`mutational load appeared to increase the probabil-
`ity, but not guarantee formation, of a neoepitope
`signature associated with a benefit. Consensus
`analysis revealed that the neoepitopes were not
`random. The frequencies of amino acids that made
`up the tetrapeptides in the group of patients with
`a long-term clinical benefit were different from
`those observed in the group with a minimal ben-
`efit or no benefit (Fig. S7A in the Supplementary
`Appendix).
`Presence of the neoepitope signature peptides
`correlated strongly with survival in both the
`discovery set and the validation set (P<0.001 and
`P<0.002, respectively, by the log-rank test) (Fig.
`3C and 3D). The correlation between mutational
`load and survival was not as strong (Fig. 2B, and
`Fig. S3A in the Supplementary Appendix).
`The shared tetrapeptides were encoded by
`mutations in diverse genes across the genome
`(Fig. S7B and Table S6 in the Supplementary Ap-
`pendix). Using RNA-sequencing data from the
`Cancer Genome Atlas, we confirmed that the
`genes harboring our somatic neoepitopes were
`widely expressed in melanoma (Table S8 in the
`Supplementary Appendix). In some cases, the
`amino acid change resulting from the somatic
`mutation led to a change in the tetrapeptide it-
`self. In others, the mutant amino acid was sepa-
`rate from the tetrapeptide and altered MHC
`binding, as has been described previously.24-26
`In addition, candidate neoepitopes common
`to both clinical groups were analyzed with the
`use of the Immune Epitope Database (www.iedb
`.org). This is the most comprehensive database of
`experimentally validated, published, and curated
`antigens, and it has been used to develop algo-
`rithms to identify antigens with high accuracy.14
`The candidate neoepitopes common to patients
`with a long-term clinical benefit were homolo-
`gous to many more viral and bacterial antigens
`in the database than were the neoepitopes com-
`mon to patients with a minimal benefit or no
`benefit (Table S9 in the Supplementary Appen-
`dix). For example, the tetrapeptide substring
`ESSA was shared by patients with a long-term
`
`clinical benefit (Fig. 4A) and corresponds to the
`precise antigenic portion of human cytomegalo-
`virus immediate early epitope (MESSAKRKMDP-
`DNPD).27 These data suggest that the neoepi-
`topes in patients with strong clinical benefit from
`CTLA-4 blockade may resemble epitopes from
`pathogens that T cells are likely to recognize. The
`cross-reactive peptides defined by short peptide
`consensus sequences that were discovered by
`Birnbaum et al. with the use of an unbiased
`screen also had substantial homology to antigens
`in microbes.21 Although tantalizing, these obser-
`vations will require further study to confirm.
`Using a whole-exome sequencing approach,
`we characterized the predicted antigenic peptide
`space (see the Methods section in the Supple-
`mentary Appendix). As further validation of our
`study, we reidentified melanoma antigen recog-
`nized by T cells (MART-1, also known as MelanA),
`an experimentally validated melanocytic antigen
`(Fig. S8).28 EKLS, which comprises the core
`amino acids of the MART-1 MHC class II epitope,
`was shared by patients with a long-term clinical
`benefit, and the phosphoserine moiety is critical
`for T-cell receptor recognition.29 The frequency of
`leukocyte common antigen–positive cells and ra-
`tio of CD8-positive cells to FOXP3-positive cells
`were substantially different between patients
`with a long-term clinical benefit from ipilim-
`umab and those with a minimal benefit or no
`benefit (Fig. S9 in the Supplementary Appendix).
`
`In Vitro Validation of Predicted
`Immunogenic Peptides
`Translation of next-generation sequencing into
`in vitro validation of peptide predictions has
`proven challenging, even in expert hands, with
`very low published validation rates.15 In vitro as-
`says are hampered by the paucity of clinical sam-
`ples, the sensitivity of preserved cells to the
`freeze–thaw process, the low frequency of anti-
`neoantigen T cells in clinical samples, and the
`very low sensitivity of T cells in vitro in the ab-
`sence of the complex in vivo immunogenic mi-
`croenvironment.
`We attempted to optimize prediction by inte-
`grating multiple high-throughput approaches
`(Fig. S4 in the Supplementary Appendix). On the
`basis of our prediction algorithm, we generated
`pools of peptides and performed assays of T-cell
`activation for patients for whom we had suffi-
`
`2195
`
`Genetic Basis for Response to CTLA-4 Blockade
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 7 of 11
`
`

`

`T h e ne w e ngl a nd jou r na l o f m e dicine
`
`Ipilimumab
`
`Antigen-
`presenting
`cell
`
`Tcell
`
`CTLA-4
`8
`C D 2
`T C R
`
`B 7
`
`B 7
`
`M H C
`
`Peptide
`
`Blockage of
`CLTA-4 results
`in T-cell activation
`
`PatientNo.
`
`Nonmutant
`
`Mutant
`
`SD1494
`
`YLLGSSALT
`
`YLLESSALT
`
`CR9306
`
`VGSSADILY
`
`VESSADILY
`
`PR4092
`
`YFPEESSAL
`
`YSPEESSAL
`
`TESPFEQHI
`
`1
`
`7
`
`24
`
`36
`48
`Weeks
`
`60
`
`108
`
`156
`
`GLEREGFTF
`
`Baseline
`
`7
`
`24
`
`Weeks
`
`48
`
`120
`
`1.50
`
`1.25
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`0.00
`
`0.10
`
`0.08
`
`0.06
`
`0.04
`
`0.02
`
`0.00
`
`IFN-γ–PositiveT-CellResponse(%)
`
`IFN-γ–PositiveT-CellResponse(%)
`
`TKSPFEQHI
`
`TESPFEQHI
`
`GLERGGFTF GLEREGFTF
`
`1.50
`
`1.25
`
`1.00
`
`0.75
`
`0.50
`
`0.25
`
`0.00
`
`0.08
`
`0.06
`
`0.04
`
`0.02
`
`0.00
`
`PolyfunctionalT-CellResponse(%)
`
`PolyfunctionalT-CellResponse(%)
`
`A
`
`B
`
`C
`
`Figure 4. Role of Neoantigens in Activation of T Cells from Patients Treated with CTLA-4 Blockade.
`Panel A shows an example of a tetrapeptide substring of human cytomegalovirus. In each case, the nonamer con-
`taining the mutation is predicted to bind and be presented by a patient-specific HLA. Panel B shows the dual posi-
`tive (interferon-γ [IFN-γ] and tumor necrosis factor α [TNF-α]) CD8+ T-cell response to TESPFEQHI and nonmutant
`peptide TKSPFEQHI and the increase in IFN-γ+ T cells over time. Data from Patient CR9306 are shown. T bars indi-
`cate the standard deviation. Panel C shows the dual positive (IFN-γ and TNF-α) CD8+ T-cell response to GLEREGFTF
`and nonmutant peptide GLERGGFTF and illustrates the increase in peptide-specific T cells 24 weeks after the initia-
`tion of treatment with ipilimumab relative to baseline. Data from Patient CR0095 are shown. MHC denotes major
`histocompatibility complex, and TCR T-cell receptor.
`
`2196
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 8 of 11
`
`

`

`cient lymphocytes (see the Methods section in
`the Supplementary Appendix). Positives pools
`were observed for three of five patients (Fig.
`S10A, S10B, and S10C in the Supplementary Ap-
`pendix). We identified the exact peptides for pa-
`tients with adequate PBMCs. We found a polyfunc-
`tional T-cell response to the peptide TESPFEQHI
`in Patient CR9306 (Fig. S10D in the Supplementary
`Appendix) but not to its nonmutant counterpart,
`TKSPFEQHI. This response peaked at 60 weeks
`after the initiation of treatment (Fig. 4B). T-cell
`responses were absent in healthy donors (Fig.
`S10E in the Supplementary Appendix). TESPFEQHI
`had a predicted MHC class I affinity for B4402
`of 472 nM, as compared with 18323 nM for
`TKSPFEQHI. ESPF is a common tetrapeptide
`found in the response signature and is a sub-
`string (positions 176 through 179) of the hepati-
`tis D virus large delta epitope p27 (PESPFA and
`ESPFAR).30 TESPFEQHI results from a mutation
`in FAM3C (c.A577G;p.K193E), a gene highly ex-
`pressed in melanoma (Table S8 in the Supple-
`mentary Appendix).
`We also found that peptide GLEREGFTF elicited
`a polyfunctional T-cell response in Patient CR0095
`(Fig. 4C, and Fig. S10F in the Supplementary Ap-
`pendix), whereas nonmutant GLERGGFTF did not.
`This response peaked at 24 weeks after the ini-
`tiation of treatment (Fig. 4C). GLEREGFTF arises
`from a mutation in CSMD1 (c.G10337A;p.G3446E),
`which is also highly expressed in melanoma (Ta-
`ble S8 in the Supplementary Appendix), and the
`peptide has 80% homology to a known Burkhold-
`eria pseudomallei antigen (Immune Epitope Data-
`base Reference ID: 1027043). The lack of T-cell
`activation may not rule out a given neoantigen
`because in vitro assays are limited in sensitivity,
`as described above.
`
`Discussion
`
`Anti–CTLA-4 and anti–programmed cell death 1
`antibodies have resulted in long-term disease
`control in a subgroup of patients with melano-
`ma.1,2 Here, we have illustrated the importance
`of tumor genetics in defining the basis of the
`clinical benefit from CTLA-4 blockade.
`Our observations suggest a number of prin-
`ciples relevant to immunotherapy for cancer.
`Although a high mutational load is associated
`
`with a benefit from immune checkpoint abroga-
`tion, this factor alone is not sufficient to impart
`a clinical benefit. Rather, there are somatic neo-
`epitopes that are shared by patients with a pro-
`longed benefit and are absent in those without a
`prolonged benefit. Owing to somatic mutations,
`a subset of proteins present in the tumor becomes
`recognized by the immune system as nonself,
`given their novelty in the tumor context.8,14,31,32
`These concepts were formulated in the discovery
`set and confirmed in the validation set and will
`require further prospective study before use as a
`definitive biomarker.
`It is well known in the field of infectious
`diseases that an individual amino acid within a
`peptide can affect immunogenicity by altering
`peptide–MHC or peptide–T-cell receptor interac-
`tions.33,34 In cancers, the altered amino acid resi-
`due resulting from a single missense mutation
`can create a T-cell epitope from a previously self
`peptide.31,32,35 In the patients described here,
`altered amino acids resulting from tumor muta-
`tions caused the tumors to display somatic neoepi-
`topes that elicited an antitumor response aug-
`mented by CTLA-4 blockade.
`Our study has limitations. Although large
`for a genomic study (128 exomes), our sample
`size was limited, patients had received a variety
`of previous treatments, and tumor samples
`were obtained at various time points. Further-
`more, although the panel of somatic neoepi-
`topes (Fig. 3A and 3B, and Table S6 in the
`Supplementary Appendix) may constitute the
`most important ones, the in vivo relative immu-
`nologic contribution of each peptide is unclear.
`However, data showing that functionally im-
`portant immunogenic epitopes persisted after
`treatment with expanded tumor-infiltrating
`lymphocytes suggest that the response to mu-
`tations may persist over time.16 Although the
`recapitulation of the neoantigen signature in
`the validation set suggests that this may pro-
`vide a generally applicable tool for prediction
`of a benefit from immunotherapy, further stud-
`ies will be needed to investigate the role of
`MHC class II molecules and the relative effects
`and characteristics of neoantigens in different
`cancers.
`Our use of whole-exome sequencing to iden-
`tify a genetic basis associated with a benefit from
`
`2197
`
`Genetic Basis for Response to CTLA-4 Blockade
`
`n engl j med 371;23 nejm.org december 4, 2014
`
`
`
`Genome Ex. 1033
`Page 9 of 11
`
`

`

`CTLA-4 blockade provides proof of principle that
`tumor genomics can inform responses to im-
`munotherapy. For the field of cancer genetics,
`these data suggest a need for an expanded defi-
`nition of the previous categories of driver and
`passenger mutations. Our data show that exonic
`missense mutations in general confer increased
`MHC class I binding (Fig. S5A and S5B in the
`Supplementary Appendix) and confirm the hy-
`pothesis36 that some mutations formerly catego-
`rized as passengers may in

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket