throbber
' t: .«.y.n",. 4,
`
`{MD I
`
`

`

`Volume 24 Number 5 2014
`
`Executive Editor Hillary E. Sussman
`
`Assistant Editor Laura E. DeMare
`
`Aravinda Chakravarti
`
`Editors
`
`Evan E. Eichler
`
`Richard A. Gibbs
`
`Tara Kulesa
`
`Eric Green
`
`Richard M. Myers
`
`William]. Pavan
`
`Administrative Assistant
`Peggy Calicchia
`
`Production Manager
`Linda Sussman
`
`Production Editor
`Marie Cotter
`
`Production Assistant
`
`Editorial Board
`
`I. Akey (Univ. of Washington)
`B. Andersson (Karolinska Institute)
`S.£. Antonarakis (Univ. of Geneva Medical School)
`B.E. Bernstein (Broad Institute)
`W. Blckmore (Medical Research Council)
`M. Boehnke (Univ. of Michigan)
`M. Brudno (Univ. of Toronto)
`M. Buiyk (Brigham 5t Women's Hospital and Harvard
`Medical School)
`L. Carrel (Penn State College of Medicine)
`N.P. Carter (Wellcome Trust Sanger Institute)
`G.A. Churchill (The lackson Laboratory)
`B. Cohen (Washington Univ. in St. Louis School of Medicine)
`G.M. Cooper (HudsonAlpha Institute for Biotechnology)
`G.E. Crawford (Duke Univ.)
`A. DI Rlenzo (Univ. of Chicago)
`M. Dunham (Univ. of Washington)
`P.|. Farnham (Univ. of California, Davis)
`5. Gabriel (Broad Institute)
`M.B. Cersteln (Yale Univ.)
`M. Hahn (lndiana Univ.)
`I.M. Hall (Univ. of Virginia)
`D.B. Iaffe (Broad Institute)
`L. Iin (Fudan University)
`S. Iones (BC Cancer Agency)
`1. Korbei (European Molecular Biology Laboratory)
`].D. Lleb (Univ. of North Carolina in Chapel Hill)
`E. Liu (Genome Institute of Singapore)
`|.R. Lupski (Baylor College of Medicine)
`T.F.C. Mackay (North Carolina State Univ.)
`P. Maiurnder (Indian Statistical Institute)
`
`K. Makova (Pennsylvania State Univ.)
`E. Mardls (Washington Univ. in St. Louis School of Medicine)
`E.H. Marguiles (National Human Genome Research Institute)
`G.T. Marti-I (Boston College)
`A.S. McCalllon (Johns Hopkins Univ. School of Medicine)
`M.L. Meyerson (Dana-Farber Cancer Institute and Harvard
`Medical School)
`A. Milosavlievic (Baylor College of Medicine)
`ll. MItra (Washington Univ. in St. Louis School of Medicine)
`|.V. Moran (Univ. of Michigan Medical School)
`M.A. Nobrega (Univ. of Chicago)
`|.P. Noonan (Yale Univ. School of Medicine)
`J. Parkhili (The Wellcome Trust Sanger Institute)
`W.R. Pearson (Univ. of Virginia)
`].H. Postlethwait (Univ. of Oregon)
`0. Rando (Univ. of Massachusetts Medical School)
`A. Regev (Broad Institute)
`D.A. Relrnan (Stanford Univ.)
`I. Rogers (Baylor College of Medicine)
`S.L. Salzberg (Johns Hopkins Univ. School of Medicine)
`P.C. Scacheri (Case Western Reserve Univ.)
`E. Segal (Weizmann Institute)
`1. Shendure (Univ. of Washington)
`].A. Stamatoyannopoulos (Univ. of Washington)
`M.R. Stratton (Wellcome Trust Sanger Institute)
`S. Tishkoff (Univ. of Pennsylvania)
`A.].M. Walhout (Univ. of Massachusetts Medical School)
`S.T. Warren (Emory Univ. School of Medicine)
`D.A. Wheeler (Baylor College of Medicine)
`R. Wolfe (University College, Dublin)
`K. Zhao (National Heart, Lung, and Blood Institute)
`
`
`
`Genome Research (ISSN 1088-9051) is published monthly by Cold Spring Harbor Laboratory Press, 500 Sunnyside Blvd, Woodbury, NY 11797-2924.
`
`
`Periodicals paid at Woodbury, NY and additional mailing offices. Canada Post International Publications Mail Product (Canadian distribution) Sales Agreement
`No. 1321846. POSTMASTER: Send address changes to Cold Spring Harbor Laboratory Press, 500 Sunnyside Boulevard, Woodbury, NY 11 797-2924.
`Subscriptions: Kathleen Cirone, Subscription Manager. Individual subscribers have a choice of "online only" or "print + online" subscriptions for this journal. Oniine
`
`
`only: 385; Print + Online: U.S., 5135; Canada and Mexico, $220; R.O.W., 5250 (includes airlift). For 2014 institutional pricing, visit http:f/genome.org/site/
`
`
`subscriptions/costdtl. Orders may be sent to Cold Spring Harbor Laboratory Press, Fulfillment Department, 500 Sunnyside Boulevard, Woodbury, New York
`
`
`117974924. Telephone: Continental U.S. and Canada 1-800-843—4388; all other locations 516-422-4100. Fax 516-422-4097. Personal subscriptions must be
`
`
`prepaid by personal check, credit card, or money order. Claims for missing issues must be received within four months of issue date.
`
`
`Advertising: Marcie Siconolfi, Advertising Manager, Cold Spring Harbor Laboratory Press,
`I Bungtown Rd, Cold Spring Harbor, New York 11724-2203. Phone:
`
`
`516-422—4010: fax: 516-422-4092.
`
`
`Information for Contributors: Author instructions are available at our website, http:/fwwwgenomeorg.
`
`
`Dnllne Manuscript Submission: http://submiLgenomeorg.
`
`
`Copyright Information: Authors of articles published in Genome Research retain copyright of the articles (except US Government employees) but grant Cold
`
`
`Spring Harbor Laboratory Press exclusive right to publish the articles for a period of six months following full-issue publication. This includes the rights to publish,
`
`
`reproduce, distribute, display, and store the article in all formats; to translate the article into other languages; to create adaptations, summaries, extracts, or
`
`
`derivations of the article; and to license others to do any or all of the above. Authors can reuse their articles in their work as long as Genome Research is credited as
`
`
`the place of original publication. Six months following publication, these articles are made available under a CC—BY-NC 4.0 Creative Commons license.
`
`
`Articles that carry the Open Access designation are available immediately under a CC-BY 4.0 0r CC-BY-NC 4.0 Creative Commons license. Detailed copyright terms
`
`
`are online at http:/lgenomecshlp.org/site/misc/terms.xhtm|. Permission information is online at http:/rgenomecshlp.org[site/miscfperrnissionsjanding.xhtml.
`
`
`Copyright (9) 2014 by Cold Spring Harbor Laboratory Press
`
`
`

`

` Volume 24 Issue 5 May 2014
`
`Perspecfive
`Can I be sued for that? Liability risk and the disclosure of clinically significant genetic
`research findings
`Amy L. McGuire, Bartha Maria Knoppers, Ma'n H. Zawati, and Ellen Wright Clayton
`
`Research
`
`PRDM? binding organizes hotspot nucleosomes and limits Holliday Junction migration
`Christopher L. Baker, Michael Walker, Shimpei Kaiita, Petko M. Petkov, and Kenneth Paigen
`
`Somatic mutations found in the healthy blood compartment of a llS-yr—oid woman
`demonstrate oligoclonal hematopoiesis
`Henne Holstege, Wayne Pfeiffer, Daoud Sie, Marc Hulsman, Thomas J. Nicholas,
`Clarence C. Lee, Tristen Ross, Jue Lin, Mark A. Miller, Bauke Ylstra, Hanne Meijers-Heijboer,
`Martijn H. Brugman, Frank J.T. Staal, Gert Holstege, Marcel |.T. Reinders, Timothy T. Harkins,
`Samuel Levy, and Erik A. Sisterrnans
`
`Neo—antigens predicted by tumor genome meta-analysis correlate with increased patient
`survival
`
`Scott D. Brown, Rene L. Warren, Ewan A. Gibb, Spencer D. Martin, John J. Spinelli,
`Brad H. Nelson, and Robert A. Holt
`
`Enhancer-targeted genome editing selectively blocks innate resistance to oncokinase
`inhibition
`
`Dan E. Webster, Brook Barajas, Rose T. Bussat, Karen J. Yan, Poornima H. Neela, Ross I. Flockhart,
`Joanna Kovalski, Ashley Zehnder, and Paul A. Khavari
`
`Recurrent epimutations activate gene body promoters in primary glioblastoma
`Raman P. Nagaraian, 30 Zhang, Robert J.A. Bell, Brett E. Johnson, Adam B. Olshen,
`Vasavi Sundaram, Daofeng Li, Ashley E. Graham, Aaron Diaz, Shaun D. Fouse, Ivan Smirnov,
`Jun Song, Pamela L. Paris, Ting Wang, and Joseph F. Costello
`
`Identifying mRNA sequence elements for target recognition by human Argonaute proteins
`Jingiing Li, TaeHyung Kim, Razvan Nutiu, Debashish Ray, Timothy R. Hughes,
`and Zhaolei Zhang
`
`Evolution of splicing regulatory networks in Drosophila
`C. Joel McManus, Joseph D. Coolon, Jodi Eipper—Mains, Patricia J. Wittkopp,
`and Brenton R. Graveley
`
`Tempo and mode of regulatory evolution in Drosophila
`Joseph D. Coolon, C. Joel McManus, Kraig R. Stevenson, Brenton R. Graveley,
`and Patricia J. Wittkopp
`
`719
`
`724
`
`733°A
`
`7430A
`
`75]
`
`761
`
`775
`
`786
`
`7970"
`
`(continued)
`
`

`

`High-resolution mapping definES the cooperative architecture of Polycomb response elements
`Guillermo A. Orsi, Sivakanthan Kasinathan, Kelly T. Hughes, Sarah Saminadin—Peter,
`Steven Henikoff, and Kami Ahmad
`
`Genome methylation in D. melanogasrer is found at specific short motifs and is independent
`of DNMT2 activity
`Sachiko Takayama, Joseph Dhahbi, Adam Roberts, Guanxiong Mao, Seok—Jin Heo, Lior Pachter,
`David l.l<. Martin, and Dario Boffelli
`
`Widespread and frequent horizontal transfers of transposable elements in plants
`Moaine E| Baidouri, Marie-Christine Carpentier, Richard Cooke, Dongying Ciao, Eric Lasserre,
`Christel Llauro, Marie Mirouze, Nathalie Picault, Scott A. Jackson, and Olivier Panaud
`
`Predicting the virulence of MRSA from its genome sequence
`Maisem Laabei, Mario Recker, Justine K. Rudkin, Mona Aldeljawi, Zeynep Gulay,
`Tim J. Sloan, Paul Williams, Jennifer L. Endres, Kenneth W. Bayles, Paul D. Fey,
`Viiaya Kumar Yajjala, Todd Widhelm, Erica Hawkins, Katie Lewis, Sara Pan‘ett, Lucy Scowen,
`Sharon J. Peacock, Matthew Holden, Daniel Wilson, Timothy D. Read, Jean van den Elsen,
`Nicholas K. Priest, Edward J. Feil, Laurence D. Hurst, Elisabet Josefsson, and Ruth C. Massey
`
`809
`
`82]
`
`3310A
`
`8390A
`
`A genomic portrait of the genetic architecture and regulatory impact of microRNA
`expression in response to infection
`
`850
`
`Katherine J. Siddle, Matthieu Deschamps, Ludovic Tailleux, Yohann Nédélec, Julien Pothiichet,
`Geanncarlo Lugo-Villarino, Valentina Libri, Brigitte Gicquel, Olivier Neyrolles, Guillaume Laval,
`Etienne Patin, Luis B. Barreiro, and Lluis Quintana-Murci
`
`Methods
`
`General approach for in vivo recovery of cell type-specific effector gene sets
`Julius C. Barsi, Qiang Tu, and Eric H. Davidson
`
`ISMARA: automated modeling of genomic signals as a democracy of regulatory motifs
`Piotr J. Balwierz, Mikhail Pachkov, Phil Arnold, Andreas J. Gruber, Mihaela Zavolan,
`and Erik van Nimwegen
`
`860
`
`8690A
`
`0AOpen Access pa per
`
`Cover Horizontal transfer of genetic material has been demonstrated on a small scale
`for several organisms. In this issue, this phenomenon is demonstrated to be widespread
`in the plant kingdom. On the cover is a Pointillist-Iike view of the horizontal transfer of
`transposable elements between palm (top left) and grape (bottom right). The two circles
`depict the respective DNA molecules and leaf colors indicate the chimeric nature of
`plants. Horizontal branches on the "tree of life“ at the bottom left illustrate frequent
`transfer during the evolution of plants. (Cover illustration by Abdelkebir El Baidouri.
`[For details, see El Baidouri et al., pp. 831—838.])
`
`FSCmiscw MIX
`
`Paper from
`responsible sources
`FSC“ 001294?
`
`

`

`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`Research ,,
`
`Neo—antigens predicted by tumor genome meta-analysis
`correlate with increased patient survival
`
`Scott D. Brown}2 Rene L. Warren,1 Ewan A. Gibb,1'3 Spencer D. Martin,1'3'4
`John J. Spinelli,5'6 Brad H. Nelson,3'4'7 and Robert A. Holt1'3'8'9
`'Canada’s Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia V52 1L3, Canada; ZGenome
`Science and Technology Program, University of British Columbia, Vancouver, British Columbia V6 T 124, Canada; 3Department of
`Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 124, Canada; 4Deeley Research Centre, BC Cancer
`Agency, Victoria, British Columbia V8R 6V5, Canada,- 5Cancer Control Research Program, BC Cancer Agency, Vancouver, British
`Columbia V52 1L3, Canada; “School of Population and Public Health, University of British Columbia, Vancouver, British Columbia
`V6 T 124, Canada; 7Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia VBP 5 C2,
`Canada,- SDepartment of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 156, Canada
`
`Somatic missense mutations can initiate tumorogenesis and, conversely, anti‘tumor cytotoxic T cell (CTL) responses.
`Tumor genome analysis has revealed extreme heterogeneity among tumor missense mutation profiles, but their relevance
`to tumor immunology and patient outcomes has awaited comprehensive evaluation. Here, for 5l5 patients from six tumor
`sites, we used RNA-seq data from The Cancer Genome Atlas to identify mutations that are predicted to be immunogenlc
`in that they yielded mutational epitopes presented by the MHC proteins encoded by each patient’s autologous HLA—A
`alleles. Mutational epitopes were associated with increased patient survival. Moreover, the corresponding tumors had
`higher CTL content, inferred from CDBA gene expression, and elevated expression of the CTL exhaustion markers PDCDl
`and CTL“. Mutational epitopes Were very scarce in tumors without evidence of CT1. infiltration. These findings suggest
`that the abundance of predicted immunogenic mutations may be useful for identifying patients likely to benefit from
`checkpoint blockade and related immunotherapies.
`
`{Supplemental material is available for this article]
`
`The accumulation of somatic mutations underlies the initiation
`
`and progression of most cancers by conferring upon tumor cells
`unrestricted proliferative capacity (Hanahan and Weinberg 2011).
`The analysis of cancer genomes has revealed that tumor muta-
`tional landscapes (Vogelstein et al. 2013) are extremely variable
`among patients, among different tumors from the same patient,
`and even among the different regions of a single tumor (Gerlinger
`et al. 2012). There is a need for personalized strategies for cancer
`therapy that are compatible with mutational heterogeneity, and in
`this regard, immune interventions that aim to initiate or enhance
`anti-tumor immune responses hold much promise. Therapeutic
`antibodies and chimeric antigen receptor (CAR) technologies have
`shown anti-cancer efficacy (Fox et al. 2011), but such antibody-
`based approaches are limited to cell surface target antigens (Slamon
`et a1. 2001: Coiffier et a1. 2002: Yang et a1. 2003; Cunningham
`et al. 2004: Kalos et al. 2011). In contrast, most tumor mutations
`are point mutations in genes encoding intracellular proteins. Short
`peptide fragments of these proteins, after intracellular processing
`and presentation at the cell surface as MHC ligands, can elicit Tcell
`immunoreactivity. Further,
`the presence of tumor infiltrating
`lymphocytes (TIL). in particular, CD8’ T cells, has been associated
`with increased survival (Sato et al. 2005: Nelson 2008; Obie et al.
`2009; Yamada et al. 2010; Gooden et al. 2011: Hwang et al. 2012),
`suggesting that the adaptive immune system can mount protective
`anti-tumor responses in many cancer patients (Kim et al. 2007; Fox
`
`at al. 2011). The antigen specificities of tumor~infiltrating T cells
`remain almost completely undefined (Andersen el al. 2012), but
`there are numerous examples of cytotoxic T cells recognizing sin-
`gle amino acid coding changes originating from somatic tumor
`mutations (Lennerz et al. 2005: Matsushita et al. 2012; Heemsketk
`et a1. 2013; Lu et al. 2013; Robbins et al. 2013; van Rooii et a1. 2013:
`Wick et al. 2014). Thus, the notion that tumor mutations are reser-
`voirs of exploitable hen-antigens remains compelling (Heemskerk
`et a1. 2013). For a mutation to be recognized by CD8' T cells, the
`mutant peptide must be presented by MHC I molecules on the
`surface of the tumor cell. The ability of a peptide to bind a given
`MHC l molecule with sufficient affinity for the peptide-MHC
`complex to be stabilized at the cell surface is the single most lim-
`iting step in antigen presentation and T cell activation (Yewdell
`and Bennink 1999]. Recently, several algorithms have been de—
`veloped that can predict which peptides will bind to given MHC
`molecules (Nielsen et al. 2003,- Bui et a1. 2005; Peters and Sette
`2005; Vita et al. 2010,- Lundegaard ct al. 201 1}, thereby providing
`guidance into which mutations are immunogenic.
`The Cancer Genome Atlas (TCGA) (http:/‘lcancergenomenih.
`govl) is an initiative of the National Institutes of Health that has
`created a comprehensive catalog of somatic tumor mutations
`identified using deep sequencing. As a member of The Cancer
`Genome Atlas Research Network, our center has generated exten-
`sive tumor RNA-seq data. Here, we have used public TCGA RNA-seq
`data to explore the T cell immunoreactivity of somatic missense
`
`’Comspondlng author
`E-rnall rholtflbcgse.“
`© 2014 Brown et al. This article, published in Genome Research, is available
`Article published online before print. Article, supplemental material, and publi-
`under a Creative Commons License (Attribution 4.0 international), as described
`cation date are athttp://www.genome.orglcgi/doi/'l0.110119r.155985.113.
`Freely available online through the Genome Research Open Access option,
`at httpzllcreativecornmons.otgl|icensesibyi4.0.
`
`
`24:743—750 Published by Cold Spring Harbor Laboratory Press;
`
`lSSN 1088-90‘ill14: wwwgennmeorg
`
`Genome Research
`wwwgeoomeorg
`
`743
`
`

`

`Brown et al.
`
`mutations across six tumor sites. This type of analysis is challenged
`not only by large numbers of mutations unique to individual pa-
`tients, but also by the complexity of personalized antigen pre-
`sentation by MHC arising from the extreme HLA allelic diversity in
`the outbred human population. Previous studies have explored the
`potential immunogenicity of tumor mutations (Segal et al. 2008;
`Warren and Holt 2010; Khalili et al. 2012}, but these have been
`hampered by small sample size and the inability to specify autoi-
`ogous HLA restriction. Recently, we described a method of HLA
`calling from RNA-seq data that shows high sensitivity and speci-
`ficity (Warren et al. 2012). Here, we have obtained matched tumor
`mutational profiles and HIA-A genotypes from TCGA subjects and
`used these data to predict patient-specific mutational epitope
`profiles. The evaluation of these data together with RNA-seq-de-
`rived markers of T cell infiltration and overall patient survival
`provides the first comprehensive view of the landscape of poten-
`tially immunogenic mutations in solid tumors.
`
`Results
`
`Summary of available data
`
`Raw TCGA RNA-seq data plus clinical metadata and complete
`profiles of sequence-verified missense mutations were obtained
`with permission from the Cancer Genomics Hub (https:I/cghub.
`ucsc.edu). Our analysis covers six tumor sites, including colon and
`rectum (combined as colorectal), ovary, breast, brain, kidney, and
`lung. These were the only tumor sites with complete and non-
`embargoed data at the time of this study. The RNA-seq data were
`first processed using HLAminer (Warren et al. 2012) to predict, at
`four-digit resolution, the two HLA-A alleles carried by each subject.
`Data from 515 patients with unambiguous HLA-A calls were pro-
`cessed further. The distribution of missense mutation counts
`
`across patients with different tumor types is shown in Figure 1. For
`each of the 22,758 total missense mutations, we evaluated binding
`
`
`
`Numberofmutationsperpatient
`
`100150200250
`
`50
`
`
`
`of all possible 8— to Il-mer mutant peptide variants to autologous
`HLA-A encoded MHC proteins using the Immune Epitope Database
`(IEDB) T Cell Epitope-MHC Binding Prediction Tool (Vita et al.
`2010) (http:/lwwaedborgl). We EOCused our analysis on HlA-A
`alleles because (1) MHC I proteins (encoded by HLA.A, -B, and -C
`genes) present antigens to CD8+ cytotoxic T cells, which are the
`subset of T cells most strongly linked to patient survival, and (2)
`HLA-A alleles of MHC I yield the most accurate peptide binding
`affinity predictions by IEDB and most other algorithms due to the
`abundance of HLA-A-specific training data (Hoof et al. 2009). All
`mutational data, RNA-seq derived HlA—A calls, IEDB epitope pre-
`dictions, RNA-seq-derived' gene expression values, and clinical
`metadata were compiled in a MySQL database for analysis.
`
`CDEA expression is associated with survival
`
`We first asked if we could reproduce the known association be-
`tween increased numbers of tumor-infiltrating CD8‘ T cells and
`increased overall survival (Sato et al. 2005; Nelson 2008; Obie et al.
`2009; Yamada et al. 2010; Gooden et al. 2011; Hwang et al. 2012).
`CD8‘ TIL levels are usually measured by immunohistologica]
`staining. To interrogate RNA—seq data, we used the expression of
`CDSA (one component of the C08 dimer) as a surrogate for CD8*
`TIL levels. We observed significantly higher overall survival for pa-
`tients with high CD8A expression than for those patients with low
`CD8A expression (HR: 0.71, 95% CI = 0.53 to 0.94.1”: 1.7 x 10—2)
`(Fig. 2A). Likewise, the data recapitulated the known association
`between high HLA-A expression and improved overall survival
`(HR = 0.59, 95% CI = 0.44 to 0.81, P = 8.6 X 10’ 4‘) (Fig. 28; Concha
`et al. 1991; Ogino et al. 2006: Kitamura et al. 2007; Han et al. 2008;
`Biien et al. 2010). Based on these positive findings with established
`T cell and MHC markers, we proceeded to evaluate candidate
`peptide epitopes, which represent the third molecular compo-
`nent required forTcell recognition and destruction of target cells.
`
`The abundance of tumor missense mutations is not associated
`with suthrai
`
`Initially, we asked if there is a relationship between overall muta-
`tion count and CBS“ TIL. Ranking patients by decreasing CDSA
`expression and displaying the mutation count for each patient’s
`tumor revealed a skewed distribution whereby tumors with low
`CDSA expression had sparse mutations and tumors with high mu-
`tation counts were among those with elevated CDBA expression
`(Fig. 3A). Tumors with above median CDBA expression contained
`73.6% of the total mutations (P = 2.0 X 10"6 by iterative randomi-
`zation and resampling as described in Methods). However, there was
`no association between total mutation count and overall survival
`
`(HR = 0.91, 95% CI: 0.68 to 1.23, P: 5.5 x 10") (Fig. 33).
`
`Tumor missense mutations that have predicted
`immunoreactivity are associated with increased survival
`
`Lung
`I1 = 34
`
`Ovary
`n = 218
`
`Breast
`n = 24
`
`Colorectal
`n = 170
`
`Brain
`n = 16
`
`ifldney
`n = 53
`
`We reasoned that missense mutations yielding peptides with poor
`MHC l binding would be immunologically silent and hence likely
`to obscure any association between missense mutations, anti-tu-
`Flgure 1. Boxpiots showing the number of mutations per patient for
`mor immunoreactivity, and survival. To address this, we repeated
`each cancer type. The y—axis is cut off at 250 mutations for better visual.
`the above analysis focusing on those mutations that were most
`ization of the majority of the data. The dark horizontal bar shows the
`median, whereas the box encompasses the interquartile range (middle
`likely to be immunogenic by several criteria, including (1) the
`50% of the data). Whiskers reach the farthest data point that is within
`expression of the gene in the tumor bearing the mutation was
`1.5x the interquartiie range from the nearest box edge (quartile). Box
`above the median expression level of that same gene in all tumors,
`width is proportional to the sample size (lung: 34, ovary: 218, breast: 24,
`colorectal: 170, brain: 16, kidney: 53).
`(2) HlA-A expression in the tumor bearing the mutation was above
`
`
`744 Genome Research
`wwwgenomeorg
`
`

`

`lmmunogenic tumor mutations correlate with survival
`
`
`
`
`
`SurvivalProbability
`
`
`
`
`
`
`
`
`
`SurvivalProbability
`
`0
`
`1000
`
`2000
`
`3000
`
`Days
`
`4000
`
`5000
`
`0
`
`"300
`
`2000
`
`3000
`
`4000
`
`5000
`
`Figure 2. Overall survival for patients based on CDEA or HLA-A expression. Kaplan-Meier curves were constructed to look at the difference in survival of patients
`(n = 512) with low and high expression levels of (IDEA (A) or HLA-A (3). Patients were split into two groups based on the median expression value. Patients with
`high expression showed increased survival compared to those with low expression of either (A) CDBA (HR 2 0.71, 95% CI = 0.53 to 0.94, P = 1.7 X 104) or
`(B) HLA-A (HR = 0.59, 95% CI = 0.4-4 to 0.81, P = 8.6 X 10"). Tick marks on the gmph denote the last time survival status was known for living patients.
`
`the HiA—A expression requirement from the definition of a predicted
`immunogenic mutation, we fit a model including all prognostic
`factors to the subset of patients with high (above median) tumor
`HLA-A expression. Within this subset of patients, we observed that
`patients with at least one predicted immunogenic mutation had
`a significantly lower relative risk of death than those without (HR =
`0.44, 95% CI = 0.22 to 0.88, P: 2.0 X 10’2). Evaluating the reciprocal
`group of patients with low (below median) HLA-A expression, where
`the potential of immunogenic mutations to elicit bona fide anti-
`tumor responses is expected to be curtailed, there was no significant
`association between the presence of predicted immunogenic muta-
`tions and survival (HR = 1.30, 95% CI = 0.83 to 2.04, P: 2.6 X 10").
`The results from all survival analyses are summarized in Table 1.
`
`the median expression of HLA-A in all tumors, and (3) the pre—
`dicted autologous HLA-A binding affinity of the best scoring pep-
`tide containing a given mutation had an leo value of 500 nM or
`less. This value has been estimated, experimentally, to be the af-
`finity necessary for an epitope to elicit an immune response (Sette
`et al. 1994). Applying these filters, the predicted immunogenic
`mutation count was zero in 334 patients. The remaining 181 pa-
`tients had predicted immunogenic mutation counts ranging from
`1 to 147, with a median of 3. The predicted immunogenic mutation
`count showed a strong relationship with tumor CDSA expression,
`where tumors with higher numbers of such mutations had higher
`CDSA expression (Fig. 3C). Of all predicted immunogenic mutations,
`84. 7% were in tumors with above median CDBA expression (P = 1.0 X
`10*). We did not see any relationship between predicted immuno-
`genic mutation count and CD4 expression by tumors (P = 6.9 x 10’ 1)
`(Supplemental Fig. 1), consistent with the fact that we had assessed
`epitopes presented by MHC class I, which is recognized exclusively
`by CD8+ T cells. Interestingly, patients with tumors containing at
`least one predicted immunogenic mutation showed markedly in-
`anti-tumor T cell responses (Schneider et at. 2006; Blank and
`creased overall survival compared to those without predicted im-
`Mackensen 2007). Blockade of these inhibitory receptors by tar-
`munogenic mutations (HR = 0.53, 95% CI = 0.36 to 0.80, P = 2.1 X
`geted monoclonal antibodies can disinhibit anti-tumor immunity
`10—3) (Fig. 3D). To further examine this association, we fit a model
`and improve clinical outcomes (Hodi et al. 2003, 2008, 2010,-
`including all available prognostic factors (age, gender, cancer type,
`Hamanishi et al. 2007; Mansh 201 1; Brahmer et al. 2012; Topalian
`and tumor stage), as well as predicted immunogenic mutations. This
`et al. 2012; ). Given that many patients in the current study had
`model also showed significantly improved overall survival for patients
`clinically significant cancer despite having predicted immuno-
`with predicted inununogenic mutations relative to those without
`genic mutations and CD8+ TIL, we asked if there was an association
`(HR = 0.50, 95% CI = 0.31 to 0.80, P: 3.9 x 10“), indicating that the
`between immunogenic mutation load and expression of PDCDI or
`effect of predicted immunogenic mutations was independent of the
`CTLA4. We found that patients with higher numbers of predicted
`other prognostic factors. Fitting a model which contained an in-
`immunogenic mutations had increased expression of not only CD821
`teraction between cancer type and predicted immunogenic mutations
`but also PDCDi and C1144. Displaying these values in a three-way
`did not yield a significant result (P = 9.2 X 10”), indicating that the
`hive plot (Krzywinski et al. 2012) highlights the association between
`prognostic effect is not limited to a specific cancer diagnosis.
`these T cell markers and immunogenic mutation load (Fig. 4).
`Given that tumor HLA-A expression alone is a known in-
`Significance was assessed by iterative randomization and resam-
`dicator of favorable patient survival
`(Fig. 23), we asked if the
`pling (as described in Methods). Of all tumors with predicted im-
`number of predicted immunogenic mutations provides additional
`munogenic mutations, 45.9% had above median expression of all
`three of PDCDI, CTLA4, and CD8A (P = 1.0 x 10-6).
`predictive value independent of HLA-A expression. After removing
`
`
`Predicted immunogenic mutation counts correlate
`with the expression of T cell exhaustion markers
`PDCDl and CTLA4 are T cell surface molecules that can inhibit
`
`Genome Research
`www.genome.org
`
`745
`
`

`

`Brown et al.
`
`
`
`
`
`
`
`
`
`100—i
`
`> C
`
`
`
`DflAFPKM
`
`
`
`TotalMutationCounl
`
`O C
`
`
`
`UBAFPKM
`
`
`
`
`
`lmmunogenicMutationCount
`
`Survival
`
`Probability — High Mutation Count
`
`0.0
`
`
`
`. Low Mutation Count
`.
`—.—-——1—‘—'—T—'—'—‘——l——
`0
`1000
`2000
`3000
`4000
`5000
`
`DIYS
`
`
`
`
`
`._
`H.
`i
`
`.
`
`‘i.
`
`gE
`.EE
`Em
`.2
`
`.3
`
`—-.
`V‘.
`° ‘
`
`9| co
`
`a
`CS
`
`\
`I
`
`D
`
`77*
`
`1
`ztimmunogenkMutatlons
`fllmmunngenchulabom .
`
`r
`
`T000
`
`2000
`
`3000
`
`4000
`
`5000
`
`ISO-J
`Flgure 3. The total number of mutations in tumors is not associated with survival, while the number of predicted immunogenic mutations is associated
`with survival. (AC) A "skew plot" was made for all patients (n = 515), ordering patients along the x—axis according to their CDSA expression. Each patient's
`C081! expression was plotted above the x»axis, and total mutation count (A) or predicted immunogenic mutation count (C) was plotted below the x—axis.
`73.5% of the total mutation count belonged topatients with above median CDSA expression (P = 2.0 x 10“), and 84.7% of the total predicted
`immunogenic mutation count belonged to patients with above median (DEA expression (P: l .0 x 10"). (3,0) Kapian-Meier curves were constructed to
`look at the difference in survival between patients with low versus high numbers of mutations. Patients (n = 468) were split into two groups based on the
`median mutation count. There was no difference in survival between the two groups when stratifying on total mutation count (3) (HR = 0.91, 95% CI =
`0.68 to 1.23, P = 5.5 x 10"), but there was a statistically significant difference between the two groups when stratifying on predicted immunogenic
`mutation count (D) (HR = 0.53, 95% CI = 0.36 to 0.80, P = 2.1 x 10’3). Tick marks on the Kaplan-Meier graphs denote the last time survival status was
`known for living patients.
`
`Discussion
`
`the most frequent type of oncogenic mutation, which raises the
`question of whether missense mutations also underlie tumor im—
`munoreactivity. Exome analysis in mice has revealed specific
`The adaptive immune system opposes tumor development, and
`missense mutations that encode MHC class I presented mutational
`the elicitation of immunogenic cell death is a key component of
`epitopes that are capable of eliciting T cell-mediated tumor re-
`both targeted immunotherapies and conventional treatment mo-
`jection (Castle et a1. 2012,- Matsushita et a]. 2012). Moreover, hu-
`dalities including radiation and chemotherapy (Kroerner et a1.
`man tumor exome sequencing studies have identified mutational
`2013). There is a robust association between T ceil infiltration of
`epitopes recognized by autologous CD8+ TIL (Heemskerk et al.
`solid tumors and favorable patient outcomes. Missense variants are
`
`
`746 Genome Research
`www.genome.org
`
`

`

`lmmunogenic tumor mutations correlate with survival
`
`Table 1. Summary of survival analysis
` Predictor HR 95% Cl P—value
`
`
`
`
`CDEA expression
`HUI-A expression
`Total mutations
`Predicted immunogenic
`mutationsa
`Predicted immunogenic
`mutations, low HlJi-J‘ia
`2.0 x 1072'
`0.22—0.88
`0.44
`Predicted immunogenic,
`high HLA-A‘
`
`0.5 3—0.94
`0.44—0.81
`0.68—1.23
`0.31 —0.80
`
`1.7 X 10 ' 2*
`8.6 X 10' 4“
`5.5 x 10"
`3.9 X 10’3“
`
`0.83—2.04
`
`2.6 x 10"
`
`0.71
`0.59
`0.91
`0.50
`
`1.30
`
`(‘) P-values < 0.05. (") P—values < 0.005.
`“Analysis that accounted for variation from known prognostic factors,
`
`infiltration by activated CD8” T cells (Dolcetti et al. 1999}: thus,
`a mutator tumor phenotype may, in general, enhance immuno—
`reactivity. Other classes of potentially immunogenic mutations
`require exploration, such as gene fusions resulting from genomic
`rearrangements. Instances of tumors with high CD8+ TIL but few
`immunogenic mutations may also be due to immune editing
`(Matsushita et al. 2012; Vesely and Schreiber 2013). Specifically,
`tumor cells bearing highly immunogenic mutations may have been
`selectively eliminated by T cells, resulting in accumulation of CD8+
`TIL but fewer irnmunogenic mutations remaining to be detected.
`The results of the present study have clinical implications. We
`have shown that patients with tumors bearing missense mutations
`predicted to be immunogenic have a survival advantage (Fig. 3D).
`These tumors also show evidence of higher CD8+ TIL, which sug-
`gests that a number of these mutations might be immunoreactive.
`The existence of these mutations is encouraging because, in prin-
`ciple. they could be leveraged by persona

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