Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition

Kevin Litchfield, James L Reading, Clare Puttick, Krupa Thakkar, Chris Abbosh, Robert Bentham, Thomas B K Watkins, Rachel Rosenthal, Dhruva Biswas, Andrew Rowan, Emilia Lim, Maise Al Bakir, Virginia Turati, José Afonso Guerra-Assunção, Lucia Conde, Andrew J S Furness, Sunil Kumar Saini, Sine R Hadrup, Javier Herrero, Se-Hoon Lee, Peter Van Loo, Tariq Enver, James Larkin, Matthew D Hellmann, Samra Turajlic, Sergio A Quezada, Nicholas McGranahan, Charles Swanton, Kevin Litchfield, James L Reading, Clare Puttick, Krupa Thakkar, Chris Abbosh, Robert Bentham, Thomas B K Watkins, Rachel Rosenthal, Dhruva Biswas, Andrew Rowan, Emilia Lim, Maise Al Bakir, Virginia Turati, José Afonso Guerra-Assunção, Lucia Conde, Andrew J S Furness, Sunil Kumar Saini, Sine R Hadrup, Javier Herrero, Se-Hoon Lee, Peter Van Loo, Tariq Enver, James Larkin, Matthew D Hellmann, Samra Turajlic, Sergio A Quezada, Nicholas McGranahan, Charles Swanton

Abstract

Checkpoint inhibitors (CPIs) augment adaptive immunity. Systematic pan-tumor analyses may reveal the relative importance of tumor-cell-intrinsic and microenvironmental features underpinning CPI sensitization. Here, we collated whole-exome and transcriptomic data for >1,000 CPI-treated patients across seven tumor types, utilizing standardized bioinformatics workflows and clinical outcome criteria to validate multivariable predictors of CPI sensitization. Clonal tumor mutation burden (TMB) was the strongest predictor of CPI response, followed by total TMB and CXCL9 expression. Subclonal TMB, somatic copy alteration burden, and histocompatibility leukocyte antigen (HLA) evolutionary divergence failed to attain pan-cancer significance. Dinucleotide variants were identified as a source of immunogenic epitopes associated with radical amino acid substitutions and enhanced peptide hydrophobicity/immunogenicity. Copy-number analysis revealed two additional determinants of CPI outcome supported by prior functional evidence: 9q34 (TRAF2) loss associated with response and CCND1 amplification associated with resistance. Finally, single-cell RNA sequencing (RNA-seq) of clonal neoantigen-reactive CD8 tumor-infiltrating lymphocytes (TILs), combined with bulk RNA-seq analysis of CPI-responding tumors, identified CCR5 and CXCL13 as T-cell-intrinsic markers of CPI sensitivity.

Keywords: CXCL9; biomarkers; checkpoint inhibitors; clonal TMB; immunogenicity; immunotherapy; meta-analysis; mutation; neoantigen.

Conflict of interest statement

Declaration of interests K.L. has a patent on indel burden and CPI response pending and outside of the submitted work, speaker fees from Roche tissue diagnostics, research funding from CRUK TDL/Ono/LifeArc alliance, and a consulting role with Monopteros Therapeutics. S.T. has received speaking fees from Roche, AstraZeneca, Novartis, and Ipsen. S.T. has the following patents filed: Indel mutations as a therapeutic target and predictive biomarker PCTGB2018/051892 and PCTGB2018/051893 and Clear Cell Renal Cell Carcinoma Biomarkers P113326GB. S.Q. reports personal fees and employment with Achilles Therapeutics (where he is CSO) outside of the submitted work. J.L.R. consults for Achilles Therapeutics. N.M. has received consultancy fees and has stock options in Achilles Therapeutics. N.M. holds European patents relating to targeting neoantigens (PCT/EP2016/ 059401), identifying patient response to immune checkpoint blockade (PCT/ EP2016/071471), determining HLA LOH (PCT/GB2018/052004), and predicting survival rates of patients with cancer (PCT/GB2020/050221). C.A. receives research salary from AstraZeneca and is an AstraZeneca Fellow and acting study physician on the MERMAID-1 study. C.A. holds pending patents in methods to detect tumor recurrence (PCT/GB2017/053289). C.A. and C.S. declare patent PCT/US2017/028013 for methods to detect lung cancer. C.A. has received speaker fees from Novartis, Roche Diagnostics, Bristol Myers Squibb, and AstraZeneca and was an advisory board member for AstraZeneca. M.D.H. has stock and other ownership interests in Shattuck Labs, Immunai, and Arcus Biosciences; reports honoraria from AstraZeneca and Bristol-Myers Squibb; has a consulting or advisory role with Bristol-Myers Squibb, Merck, Genentech/Roche, AstraZeneca, Nektar, Syndax, Mirati Therapeutics, Shattuck Labs, Immunai, Blueprint Medicines, Achilles Therapeutics, and Arcus Biosciences; receives research funding from Bristol-Myers Squibb (Inst); has patents, royalties, and other intellectual property (a patent has been filed by Memorial Sloan Kettering [PCT/US2015/062208] for the use of TMB for prediction of immunotherapy efficacy, which is licensed to Personal Genome Diagnostics); and receives travel and accommodation expense reimbursement from AstraZeneca, Bristol-Myers Squibb, and Eli Lilly. J.L. reports personal fees from Eisai, GlaxoSmithKline, Kymab, Roche/Genentech, Secarna, Pierre Fabre, and EUSA Pharma and grants and personal fees from Bristol-Myers Squibb, Merck Sharp & Dohme, Pfizer, and Novartis outside of the submitted work. C. Swanton acknowledges grant support from Pfizer, AstraZeneca, Bristol-Myers Squibb, Roche-Ventana, Boehringer-Ingelheim, Archer Dx (collaboration in minimal residual disease sequencing technologies), and Ono Pharmaceutical; is an AstraZeneca advisory board member and chief investigator for the MeRmaiD1 clinical trial; has consulted for Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol-Myers Squibb, Celgene, Amgen, AstraZeneca, Illumina, Genentech, Roche-Ventana, GRAIL, Medicxi, Bicycle Therapeutics, and the Sarah Cannon Research Institute; has stock options in Apogen Biotechnologies, Epic Bioscience, and GRAIL; and has stock options and is co-founder of Achilles Therapeutics. C.S. holds European patents relating to assay technology to detect tumor recurrence (PCT/GB2017/053289), targeting neoantigens (PCT/EP2016/059401), identifying patient response to immune checkpoint blockade (PCT/EP2016/071471), determining HLA LOH (PCT/GB2018/052004), predicting survival rates of patients with cancer (PCT/GB2020/050221), and identifying patients who respond to cancer treatment (PCT/GB2018/051912), as well as a US patent relating to detecting tumor mutations (PCT/US2017/28013) and both a European and US patent related to identifying insertion/deletion mutation targets (PCT/GB2018/051892). S.R.H. is co-founder of Tetramershop and PokeAcell. D.B. reports personal fees from NanoString, outside this work, and he has a patent PCT/GB2020/050221 issued on methods for cancer prognostication.

Copyright © 2021 Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Design of the meta-analysis study Input studies to the meta-analysis (Figure 2) results (top) and validation cohorts for the multivariable predictive modeling (Figure 3) (bottom).
Figure S1
Figure S1
Supplementary meta-analysis data, related to Figure 2 Panel A shows the correlation in biomarker effect sizes for radiological response and overall survival clinical endpoints (Spearman's correlation). Panel B shows response rate by number of NMD-escape mutations for all available samples. Panel C shows results from previously published histology specific biomarkers, or metrics that could not be calculated in > 75% of the cohort samples.
Figure 2
Figure 2
The biomarker landscape of CPI response (A) Previously published biomarkers are shown as rows and individual cohorts within the CPI1000+ cohort as columns. The heatmap indicates the effect size of each biomarker in each cohort, measured as the log2 odds ratio (OR) for response “CR/PR” versus no response “SD/PD/NE” derived from logistic regression. Blue denotes association with response, red association with no response. Drug class and cohort sizes are annotated, and the right-hand forest plot shows the overall effect size and significance of each biomarker in meta-analysis across all studies, based on effect sizes and standard errors from each individual cohort. p values are shown from meta-analysis (random effects, on account of the different tumor types), with the first set of p-values including all samples (p-meta all cohorts) and last set (p-meta validation cohorts) including validation cohorts only (i.e., when a biomarker was originally discovered in a cohort, this cohort was excluded from the meta-analysis). For clarity of plotting, outlier OR values were capped between OR = 0.1 and OR = 10 (all outlier values were nonsignificant results skewed by rare event counts, and raw (uncapped) values were still used in the meta-analysis). (B) The CPI1000+ cohort broken into cancer/drug subgroups for combinations with two or more independent cohorts. OR effect sizes are shown on the y axis, and biomarkers that are either significant in the pan-cancer 2A analysis or within an individual subgroup are shown. Colors are arbitrary and are used only to distinguish the groups. (C) Correlation between biomarkers that are measured on a continuous scale. (D) Proportion of variance explained for each category of biomarker, for each study, calculated using logistic regression pseudo-R2.
Figure S2
Figure S2
(A) shows significant histology or drug-specific biomarker interactions identified in the CPI1000+ cohort (using histology*biomarker and drug*biomarker interaction terms in logistic regression), and (B) shows dinucleotide variant associations with CPI response, related to Figures 2 and 4
Figure 3
Figure 3
A multivariable predictor of CPI response outperforms TMB (A) Feature importance scores from XGBoost for the multivariable model, corresponding to 1,000 Monte Carlo sampling rounds. (B) Design, samples included, and features utilized in the final model training. (C) The top five feature importance scores from the final pan-cancer model. (D) ROC curves and AUC values for the multivariable predictor benchmarked to TMB, as a univariable comparator, in the three independent test cohorts (not used in any of the model training steps). p values report the significance of improved performance for the multivariable versus TMB model using DeLong’s test.
Figure 4
Figure 4
Mutational processes associated with CPI response (A) Forest plot of each mutation signature and its association with CPI response, with odds ratio values shown on the y-axis, and p-values derived from meta-analysis (hence the results are not biased by mixing histology types). (B) Proportion of signature 7 (UV) mutations (left) and the number of dinucleotide variants (DNVs) per tumor (middle), split by histology type. The panel on the right shows the correlation between signature 7 proportion and DNV count. p value and correlation coefficient are from Spearman’s rank test. (C) Grid of substitutions from the reference amino acid (rows) to the mutated amino acid (columns). The heatmap is colored from low to high, based on the simple count of each observed ref > alt change in the cohort, shown on a log10 scale. The first grid (left) shows the data for SNVs, and the second grid (middle) shows data for DNVs. The first barplot (middle) then quantifies the number of unique changes observed for SNVs and DNVs, and the second barplot shows the proportion of amino acid changes resulting in a radical amino acid change (i.e., Grantham distance ≥ 100) compared to those resulting in a conservative change (Grantham distance 

Figure 5

Somatic copy-number alteration (SCNA) profile…

Figure 5

Somatic copy-number alteration (SCNA) profile of CPI responders versus nonresponders (A) Frequency of…

Figure 5
Somatic copy-number alteration (SCNA) profile of CPI responders versus nonresponders (A) Frequency of somatic copy-number gain (top) and loss (bottom) across the genome for CPI responders (“CR/PR”) versus nonresponders (“SD/PD”). (B) Cytobands with significantly different loss or gain frequencies in responders versus nonresponders,with p-value derived from Fisher's exact test, and q values from FDR correction.. (C) Fine mapping of the 9q34 locus to identify the peak of differential loss frequency between groups. (D) TRAF2-loss percentage frequencies for cohorts with a significant difference between responders and nonresponders, with p-value derived from Fisher's exact test. (E) Probability of haploinsufficiency (pLI) scores from the gnomAD/ExAC consortium data (n = 125,748 germline human samples).

Figure S3

9q34 (TRAF2) analysis and immune…

Figure S3

9q34 (TRAF2) analysis and immune evasion data, related to Figure 5 Panel A…

Figure S3
9q34 (TRAF2) analysis and immune evasion data, related to Figure 5 Panel A shows drug sensitivity screening data for two compounds, for TRAF2 heterozygously mutated versus TRAF2 wild-type cell lines. Panel B shows immune evasion analysis, measuring as the % of patients with an antigen presentation pathway defect between tumors with 9q34 wild-type (i.e., no loss) compared to 9q34 loss tumors. The left barplot includes either a somatic copy number loss, or a non-synonymous mutation, in an antigen presentation pathway gene. The right plot includes non-synonymous mutations only. Antigen presentation pathway genes were defined as per (Rosenthal et al., 2019), also see methods. Panel C shows the frequency of whole chromosome loss in TCGA for the set of cancer types included in the CPI1000+ study.

Figure S4

Cytobands with significantly different copy-number…

Figure S4

Cytobands with significantly different copy-number loss or gain frequencies in responders versus nonresponders,…

Figure S4
Cytobands with significantly different copy-number loss or gain frequencies in responders versus nonresponders, related to Figure 5 Analysis is split by 4 tumor/drug types.

Figure 6

Focal amplification and deletion profile…

Figure 6

Focal amplification and deletion profile of CPI responders versus nonresponders (A) CPI response…

Figure 6
Focal amplification and deletion profile of CPI responders versus nonresponders (A) CPI response rate (% “CR/PR”) in patients with focal amplification (defined as copy number ≥ 5) or homozygous deletion (copy number = 0) compared to wild-type (nonamplified/deleted) tumors. The analysis was conducted for all oncogenes/tumor suppressor genes with greater than 5% amplification/deletion frequency, and p-values were derived from Fisher's exact test. (B) Counts of CCND1 amplification by histology. (C) mRNA expression for CCND1 in responders versus nonresponders from the Mariathasan et al. urothelial cancer cohort, with p-value derived from Mann-Whitney U test. (D) Overall survival analysis in MSK-IMPACT urothelial cancer CPI-treated patients for CCND1-amplified versus wild-type tumor groups. (E) Overall survival analysis in MSK-IMPACT urothelial cancer non-CPI-treated patients for CCND1-amplified versus wild-type tumor groups. (F) Overall survival analysis in MSK-IMPACT pan-cancer CPI-treated patients for CCND1-amplified versus wild-type tumor groups. (G) Overall survival analysis in MSK-IMPACT pan-cancer non-CPI-treated patients for CCND1-amplified versus wild-type tumor groups.

Figure 7

CD8 + neoantigen-reactive single-cell RNA-seq…

Figure 7

CD8 + neoantigen-reactive single-cell RNA-seq and CPI1000+ cohort analysis (A) Single-cell RNA sequencing…

Figure 7
CD8+ neoantigen-reactive single-cell RNA-seq and CPI1000+ cohort analysis (A) Single-cell RNA sequencing (RNA-seq) data from neoantigen multimer negative versus positive CD8+ TILs. The top plot shows the sorting of multimer positive versus negative T cells, and the bottom plot shows differential gene expression analysis between multimer-positive versus multimer-negative cells, with log2 fold change (FC) shown on the x axis and −log10 value on the y axis. Significant genes with > 2 FC upregulation (log2(FC) > 1) and false discovery rate (FDR) < 0.05 are shown blue. (B) The same FC upregulation values from (A) on the y axis and then overlaid on the x axis is upregulation scores from the CPI1000+ cohort (log2[FC] values for responders “PR/CR” versus nonresponders “SD/PD”). The panel only shows genes significantly upregulated in both experiments. (C) Patient-level mRNA data for the two most strongly unregulated genes (CXCL13 and CCR5) from (B) from the CPI1000+ cohort, with p-value derived from Mann-Whitney U test..

Figure S5

Clustering by common germline SNP…

Figure S5

Clustering by common germline SNP panel to ensure no duplicate participants were recorded…

Figure S5
Clustering by common germline SNP panel to ensure no duplicate participants were recorded in the CPI1000+ cohort, related to STAR methods Columns are patients, rows are SNPs.

Figure S6

Purity, sequencing coverage, and choice…

Figure S6

Purity, sequencing coverage, and choice of exome capture kits do not correlate with…

Figure S6
Purity, sequencing coverage, and choice of exome capture kits do not correlate with TMB scores in the CPI1000+ cohort, related to STAR methods

Figure S7

Purity vs TMB correlations by…

Figure S7

Purity vs TMB correlations by study, related to STAR methods

Figure S7
Purity vs TMB correlations by study, related to STAR methods
All figures (15)
Figure 5
Figure 5
Somatic copy-number alteration (SCNA) profile of CPI responders versus nonresponders (A) Frequency of somatic copy-number gain (top) and loss (bottom) across the genome for CPI responders (“CR/PR”) versus nonresponders (“SD/PD”). (B) Cytobands with significantly different loss or gain frequencies in responders versus nonresponders,with p-value derived from Fisher's exact test, and q values from FDR correction.. (C) Fine mapping of the 9q34 locus to identify the peak of differential loss frequency between groups. (D) TRAF2-loss percentage frequencies for cohorts with a significant difference between responders and nonresponders, with p-value derived from Fisher's exact test. (E) Probability of haploinsufficiency (pLI) scores from the gnomAD/ExAC consortium data (n = 125,748 germline human samples).
Figure S3
Figure S3
9q34 (TRAF2) analysis and immune evasion data, related to Figure 5 Panel A shows drug sensitivity screening data for two compounds, for TRAF2 heterozygously mutated versus TRAF2 wild-type cell lines. Panel B shows immune evasion analysis, measuring as the % of patients with an antigen presentation pathway defect between tumors with 9q34 wild-type (i.e., no loss) compared to 9q34 loss tumors. The left barplot includes either a somatic copy number loss, or a non-synonymous mutation, in an antigen presentation pathway gene. The right plot includes non-synonymous mutations only. Antigen presentation pathway genes were defined as per (Rosenthal et al., 2019), also see methods. Panel C shows the frequency of whole chromosome loss in TCGA for the set of cancer types included in the CPI1000+ study.
Figure S4
Figure S4
Cytobands with significantly different copy-number loss or gain frequencies in responders versus nonresponders, related to Figure 5 Analysis is split by 4 tumor/drug types.
Figure 6
Figure 6
Focal amplification and deletion profile of CPI responders versus nonresponders (A) CPI response rate (% “CR/PR”) in patients with focal amplification (defined as copy number ≥ 5) or homozygous deletion (copy number = 0) compared to wild-type (nonamplified/deleted) tumors. The analysis was conducted for all oncogenes/tumor suppressor genes with greater than 5% amplification/deletion frequency, and p-values were derived from Fisher's exact test. (B) Counts of CCND1 amplification by histology. (C) mRNA expression for CCND1 in responders versus nonresponders from the Mariathasan et al. urothelial cancer cohort, with p-value derived from Mann-Whitney U test. (D) Overall survival analysis in MSK-IMPACT urothelial cancer CPI-treated patients for CCND1-amplified versus wild-type tumor groups. (E) Overall survival analysis in MSK-IMPACT urothelial cancer non-CPI-treated patients for CCND1-amplified versus wild-type tumor groups. (F) Overall survival analysis in MSK-IMPACT pan-cancer CPI-treated patients for CCND1-amplified versus wild-type tumor groups. (G) Overall survival analysis in MSK-IMPACT pan-cancer non-CPI-treated patients for CCND1-amplified versus wild-type tumor groups.
Figure 7
Figure 7
CD8+ neoantigen-reactive single-cell RNA-seq and CPI1000+ cohort analysis (A) Single-cell RNA sequencing (RNA-seq) data from neoantigen multimer negative versus positive CD8+ TILs. The top plot shows the sorting of multimer positive versus negative T cells, and the bottom plot shows differential gene expression analysis between multimer-positive versus multimer-negative cells, with log2 fold change (FC) shown on the x axis and −log10 value on the y axis. Significant genes with > 2 FC upregulation (log2(FC) > 1) and false discovery rate (FDR) < 0.05 are shown blue. (B) The same FC upregulation values from (A) on the y axis and then overlaid on the x axis is upregulation scores from the CPI1000+ cohort (log2[FC] values for responders “PR/CR” versus nonresponders “SD/PD”). The panel only shows genes significantly upregulated in both experiments. (C) Patient-level mRNA data for the two most strongly unregulated genes (CXCL13 and CCR5) from (B) from the CPI1000+ cohort, with p-value derived from Mann-Whitney U test..
Figure S5
Figure S5
Clustering by common germline SNP panel to ensure no duplicate participants were recorded in the CPI1000+ cohort, related to STAR methods Columns are patients, rows are SNPs.
Figure S6
Figure S6
Purity, sequencing coverage, and choice of exome capture kits do not correlate with TMB scores in the CPI1000+ cohort, related to STAR methods
Figure S7
Figure S7
Purity vs TMB correlations by study, related to STAR methods

References

    1. Alexandrov L.B., Jones P.H., Wedge D.C., Sale J.E., Campbell P.J., Nik-Zainal S., Stratton M.R. Clock-like mutational processes in human somatic cells. Nat. Genet. 2015;47:1402–1407.
    1. Anagnostou V., Niknafs N., Marrone K., Bruhm D.C., White J.R., Naidoo J., Hummelink K., Monkhorst K., Lalezari F., Lanis M. Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat. Can. 2020;1:99–111.
    1. Andreatta M., Nielsen M. Gapped sequence alignment using artificial neural networks: application to the MHC class I system. Bioinformatics. 2016;32:511–517.
    1. Aredo J.V., Padda S.K., Kunder C.A., Han S.S., Neal J.W., Shrager J.B., Wakelee H.A. Impact of KRAS mutation subtype and concurrent pathogenic mutations on non-small cell lung cancer outcomes. Lung Cancer. 2019;133:144–150.
    1. Auslander N., Zhang G., Lee J.S., Frederick D.T., Miao B., Moll T., Tian T., Wei Z., Madan S., Sullivan R.J. Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat. Med. 2018;24:1545–1549.
    1. Ayers M., Lunceford J., Nebozhyn M., Murphy E., Loboda A., Kaufman D.R., Albright A., Cheng J.D., Kang S.P., Shankaran V. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 2017;127:2930–2940.
    1. Balachandran V.P., Łuksza M., Zhao J.N., Makarov V., Moral J.A., Remark R., Herbst B., Askan G., Bhanot U., Senbabaoglu Y., Australian Pancreatic Cancer Genome Initiative. Garvan Institute of Medical Research. Prince of Wales Hospital. Royal North Shore Hospital. University of Glasgow. St Vincent’s Hospital. QIMR Berghofer Medical Research Institute. University of Melbourne, Centre for Cancer Research. University of Queensland, Institute for Molecular Bioscience. Bankstown Hospital. Liverpool Hospital. Royal Prince Alfred Hospital, Chris O’Brien Lifehouse. Westmead Hospital. Fremantle Hospital. St John of God Healthcare. Royal Adelaide Hospital. Flinders Medical Centre. Envoi Pathology. Princess Alexandria Hospital. Austin Hospital. Johns Hopkins Medical Institutes. ARC-Net Centre for Applied Research on Cancer Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature. 2017;551:512–516.
    1. Bielski C.M., Zehir A., Penson A.V., Donoghue M.T.A., Chatila W., Armenia J., Chang M.T., Schram A.M., Jonsson P., Bandlamudi C. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat. Genet. 2018;50:1189–1195.
    1. Brash D.E. UV signature mutations. Photochem. Photobiol. 2015;91:15–26.
    1. Braun D.A., Hou Y., Bakouny Z., Ficial M., Sant’ Angelo M., Forman J., Ross-Macdonald P., Berger A.C., Jegede O.A., Elagina L. Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat. Med. 2020;26:909–918.
    1. Bravata D.M., Olkin I. Simple pooling versus combining in meta-analysis. Eval. Health Prof. 2001;24:218–230.
    1. Cha E., Klinger M., Hou Y., Cummings C., Ribas A., Faham M., Fong L. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl. Med. 2014;6:238ra70.
    1. Chapuy B., Stewart C., Dunford A.J., Kim J., Wienand K., Kamburov A., Griffin G.K., Chen P.H., Lako A., Redd R.A. Genomic analyses of PMBL reveal new drivers and mechanisms of sensitivity to PD-1 blockade. Blood. 2019;134:2369–2382.
    1. Chen L., Diao L., Yang Y., Yi X., Rodriguez B.L., Li Y., Villalobos P.A., Cascone T., Liu X., Tan L. CD38-mediated immunosuppression as a mechanism of tumor cell escape from PD-1/PD-L1 blockade. Cancer Discov. 2018;8:1156–1175.
    1. Cheng J., Vanneste E., Konings P., Voet T., Vermeesch J.R., Moreau Y. Single-cell copy number variation detection. Genome Biol. 2011;12:R80.
    1. Chow M.T., Ozga A.J., Servis R.L., Frederick D.T., Lo J.A., Fisher D.E., Freeman G.J., Boland G.M., Luster A.D. Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity. 2019;50:1498–1512.e5.
    1. Chowell D., Krishna S., Becker P.D., Cocita C., Shu J., Tan X., Greenberg P.D., Klavinskis L.S., Blattman J.N., Anderson K.S. TCR contact residue hydrophobicity is a hallmark of immunogenic CD8+ T cell epitopes. Proc. Natl. Acad. Sci. USA. 2015;112:E1754–E1762.
    1. Chowell D., Morris L.G.T., Grigg C.M., Weber J.K., Samstein R.M., Makarov V., Kuo F., Kendall S.M., Requena D., Riaz N. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359:582–587.
    1. Chowell D., Krishna C., Pierini F., Makarov V., Rizvi N.A., Kuo F., Morris L.G.T., Riaz N., Lenz T.L., Chan T.A. Evolutionary divergence of HLA class I genotype impacts efficacy of cancer immunotherapy. Nat. Med. 2019;25:1715–1720.
    1. Chudley L., McCann K.J., Coleman A., Cazaly A.M., Bidmon N., Britten C.M., van der Burg S.H., Gouttefangeas C., Jandus C., Laske K. Harmonisation of short-term in vitro culture for the expansion of antigen-specific CD8(+) T cells with detection by ELISPOT and HLA-multimer staining. Cancer Immunol. Immunother. 2014;63:1199–1211.
    1. Cibulskis K., Lawrence M.S., Carter S.L., Sivachenko A., Jaffe D., Sougnez C., Gabriel S., Meyerson M., Lander E.S., Getz G. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219.
    1. Conforti F., Pala L., Bagnardi V., De Pas T., Martinetti M., Viale G., Gelber R.D., Goldhirsch A. Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta-analysis. Lancet Oncol. 2018;19:737–746.
    1. Consortium A.P.G., AACR Project GENIE Consortium AACR Project GENIE: powering precision medicine through an international consortium. Cancer Discov. 2017;7:818–831.
    1. Conway J.R., Kofman E., Mo S.S., Elmarakeby H., Van Allen E. Genomics of response to immune checkpoint therapies for cancer: implications for precision medicine. Genome Med. 2018;10:93.
    1. Cristescu R., Mogg R., Ayers M., Albright A., Murphy E., Yearley J., Sher X., Liu X.Q., Lu H., Nebozhyn M. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science. 2018;362:362.
    1. Dagan T., Talmor Y., Graur D. Ratios of radical to conservative amino acid replacement are affected by mutational and compositional factors and may not be indicative of positive Darwinian selection. Mol. Biol. Evol. 2002;19:1022–1025.
    1. Davoli T., Uno H., Wooten E.C., Elledge S.J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355:eaaf8399.
    1. Dobin A., Davis C.A., Schlesinger F., Drenkow J., Zaleski C., Jha S., Batut P., Chaisson M., Gingeras T.R. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
    1. Endesfelder D., Burrell R., Kanu N., McGranahan N., Howell M., Parker P.J., Downward J., Swanton C., Kschischo M. Chromosomal instability selects gene copy-number variants encoding core regulators of proliferation in ER+ breast cancer. Cancer Res. 2014;74:4853–4863.
    1. Favero F., Joshi T., Marquard A.M., Birkbak N.J., Krzystanek M., Li Q., Szallasi Z., Eklund A.C. Sequenza: allele-specific copy number and mutation profiles from tumor sequencing data. Ann. Oncol. 2015;26:64–70.
    1. Fehrenbacher L., Spira A., Ballinger M., Kowanetz M., Vansteenkiste J., Mazieres J., Park K., Smith D., Artal-Cortes A., Lewanski C., POPLAR Study Group Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387:1837–1846.
    1. Fontanilles M., Marguet F., Ruminy P., Basset C., Noel A., Beaussire L., Viennot M., Viailly P.J., Cassinari K., Chambon P. Simultaneous detection of EGFR amplification and EGFRvIII variant using digital PCR-based method in glioblastoma. Acta Neuropathol. Commun. 2020;8:52.
    1. Gettinger S., Choi J., Hastings K., Truini A., Datar I., Sowell R., Wurtz A., Dong W., Cai G., Melnick M.A. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017;7:1420–1435.
    1. Ghorani E., Rosenthal R., McGranahan N., Reading J.L., Lynch M., Peggs K.S., Swanton C., Quezada S.A. Differential binding affinity of mutated peptides for MHC class I is a predictor of survival in advanced lung cancer and melanoma. Ann. Oncol. 2018;29:271–279.
    1. Ghorani E., Reading J.L., Henry J.Y., de Massy M.R., Rosenthal R., Turati V., Joshi K., Furness A.J.S., Aissa A.B., Saini S.K. The T cell differentiation landscape is shaped by tumour mutations in lung cancer. Nat. Can. 2020;1:546–561.
    1. Gibney G.T., Weiner L.M., Atkins M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016;17:e542–e551.
    1. Gorbachev A.V., Kobayashi H., Kudo D., Tannenbaum C.S., Finke J.H., Shu S., Farber J.M., Fairchild R.L. CXC chemokine ligand 9/monokine induced by IFN-gamma production by tumor cells is critical for T cell-mediated suppression of cutaneous tumors. J. Immunol. 2007;178:2278–2286.
    1. Grantham R. Amino acid difference formula to help explain protein evolution. Science. 1974;185:862–864.
    1. Gros A., Parkhurst M.R., Tran E., Pasetto A., Robbins P.F., Ilyas S., Prickett T.D., Gartner J.J., Crystal J.S., Roberts I.M. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016;22:433–438.
    1. Havel J.J., Chowell D., Chan T.A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer. 2019;19:133–150.
    1. Helmink B.A., Reddy S.M., Gao J., Zhang S., Basar R., Thakur R., Yizhak K., Sade-Feldman M., Blando J., Han G. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549–555.
    1. House I.G., Savas P., Lai J., Chen A.X.Y., Oliver A.J., Teo Z.L., Todd K.L., Henderson M.A., Giuffrida L., Petley E.V. Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin. Cancer Res. 2020;26:487–504.
    1. Huang L., Malu S., McKenzie J.A., Andrews M.C., Talukder A.H., Tieu T., Karpinets T., Haymaker C., Forget M.A., Williams L.J. The RNA-binding protein MEX3B mediates resistance to cancer immunotherapy by downregulating HLA-A expression. Clin. Cancer Res. 2018;24:3366–3376.
    1. Hugo W., Zaretsky J.M., Sun L., Song C., Moreno B.H., Hu-Lieskovan S., Berent-Maoz B., Pang J., Chmielowski B., Cherry G. Genomic and transcriptomic features of response to Anti-PD-1 therapy in metastatic melanoma. Cell. 2016;165:35–44.
    1. Jamal-Hanjani M., Wilson G.A., McGranahan N., Birkbak N.J., Watkins T.B.K., Veeriah S., Shafi S., Johnson D.H., Mitter R., Rosenthal R., TRACERx Consortium Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2109–2121.
    1. Karczewski K.J., Francioli L.C., Tiao G., Cummings B.B., Alföldi J., Wang Q., Collins R.L., Laricchia K.M., Ganna A., Birnbaum D.P., Genome Aggregation Database Consortium The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020;581:434–443.
    1. Karin N., Wildbaum G., Thelen M. Biased signaling pathways via CXCR3 control the development and function of CD4+ T cell subsets. J. Leukoc. Biol. 2016;99:857–862.
    1. Knepper T.C., Montesion M., Russell J.S., Sokol E.S., Frampton G.M., Miller V.A., Albacker L.A., McLeod H.L., Eroglu Z., Khushalani N.I. The genomic landscape of Merkel cell carcinoma and clinicogenomic biomarkers of response to immune checkpoint inhibitor therapy. Clin. Cancer Res. 2019;25:5961–5971.
    1. Koboldt D.C., Zhang Q., Larson D.E., Shen D., McLellan M.D., Lin L., Miller C.A., Mardis E.R., Ding L., Wilson R.K. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576.
    1. Koyama S., Akbay E.A., Li Y.Y., Herter-Sprie G.S., Buczkowski K.A., Richards W.G., Gandhi L., Redig A.J., Rodig S.J., Asahina H. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 2016;7:10501.
    1. Krijgsman O., Carvalho B., Meijer G.A., Steenbergen R.D., Ylstra B. Focal chromosomal copy number aberrations in cancer-Needles in a genome haystack. Biochim. Biophys. Acta. 2014;1843:2698–2704.
    1. Kyte J., Doolittle R.F. A simple method for displaying the hydropathic character of a protein. J. Mol. Biol. 1982;157:105–132.
    1. Le D.T., Uram J.N., Wang H., Bartlett B.R., Kemberling H., Eyring A.D., Skora A.D., Luber B.S., Azad N.S., Laheru D. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015;372:2509–2520.
    1. Li B., Dewey C.N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323.
    1. Li H., Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Li Z., Liu J., Zhang X., Fang L., Zhang C., Zhang Z., Yan L., Tang Y., Fan Y. Prognostic significance of cyclin D1 expression in renal cell carcinoma: a systematic review and meta-analysis. Pathol. Oncol. Res. 2020;26:1401–1409.
    1. Lindeboom R.G.H., Vermeulen M., Lehner B., Supek F. The impact of nonsense-mediated mRNA decay on genetic disease, gene editing and cancer immunotherapy. Nat. Genet. 2019;51:1645–1651.
    1. Liu D., Schilling B., Liu D., Sucker A., Livingstone E., Jerby-Arnon L., Zimmer L., Gutzmer R., Satzger I., Loquai C. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 2019;25:1916–1927.
    1. Łuksza M., Riaz N., Makarov V., Balachandran V.P., Hellmann M.D., Solovyov A., Rizvi N.A., Merghoub T., Levine A.J., Chan T.A. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature. 2017;551:517–520.
    1. Mahdey H.M., Ramanathan A., Ismail S.M., Abraham M.T., Jamaluddin M., Zain R.B. Cyclin D1 amplification in tongue and cheek squamous cell carcinoma. Asian Pac. J. Cancer Prev. 2011;12:2199–2204.
    1. Mariathasan S., Turley S.J., Nickles D., Castiglioni A., Yuen K., Wang Y., Kadel E.E., III, Koeppen H., Astarita J.L., Cubas R. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–548.
    1. McDermott D.F., Huseni M.A., Atkins M.B., Motzer R.J., Rini B.I., Escudier B., Fong L., Joseph R.W., Pal S.K., Reeves J.A. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat. Med. 2018;24:749–757.
    1. McGranahan N., Furness A.J., Rosenthal R., Ramskov S., Lyngaa R., Saini S.K., Jamal-Hanjani M., Wilson G.A., Birkbak N.J., Hiley C.T. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351:1463–1469.
    1. McGranahan N., Rosenthal R., Hiley C.T., Rowan A.J., Watkins T.B.K., Wilson G.A., Birkbak N.J., Veeriah S., Van Loo P., Herrero J., Swanton C., TRACERx Consortium Allele-specific HLA loss and immune escape in lung cancer evolution. Cell. 2017;171:1259–1271.e11.
    1. McKenna A., Hanna M., Banks E., Sivachenko A., Cibulskis K., Kernytsky A., Garimella K., Altshuler D., Gabriel S., Daly M., DePristo M.A. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303.
    1. Merino D.M., McShane L.M., Fabrizio D., Funari V., Chen S.J., White J.R., Wenz P., Baden J., Barrett J.C., Chaudhary R., TMB Harmonization Consortium Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project. J. Immunother. Cancer. 2020;8:e000147.
    1. Messina J.L., Fenstermacher D.A., Eschrich S., Qu X., Berglund A.E., Lloyd M.C., Schell M.J., Sondak V.K., Weber J.S., Mulé J.J. 12-Chemokine gene signature identifies lymph node-like structures in melanoma: potential for patient selection for immunotherapy? Sci. Rep. 2012;2:765.
    1. Miao D., Margolis C.A., Vokes N.I., Liu D., Taylor-Weiner A., Wankowicz S.M., Adeegbe D., Keliher D., Schilling B., Tracy A. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 2018;50:1271–1281.
    1. Nilsen G., Liestøl K., Van Loo P., Moen Vollan H.K., Eide M.B., Rueda O.M., Chin S.F., Russell R., Baumbusch L.O., Caldas C. Copynumber: efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics. 2012;13:591.
    1. Pabla S., Conroy J.M., Nesline M.K., Glenn S.T., Papanicolau-Sengos A., Burgher B., Hagen J., Giamo V., Andreas J., Lenzo F.L. Proliferative potential and resistance to immune checkpoint blockade in lung cancer patients. J. Immunother. Cancer. 2019;7:27.
    1. Panda A., de Cubas A.A., Stein M., Riedlinger G., Kra J., Mayer T., Smith C.C., Vincent B.G., Serody J.S., Beckermann K.E. Endogenous retrovirus expression is associated with response to immune checkpoint blockade in clear cell renal cell carcinoma. JCI Insight. 2018;3:e121522.
    1. Peng W., Chen J.Q., Liu C., Malu S., Creasy C., Tetzlaff M.T., Xu C., McKenzie J.A., Zhang C., Liang X. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 2016;6:202–216.
    1. Pierini F., Lenz T.L. Divergent allele advantage at human MHC genes: signatures of past and ongoing selection. Mol. Biol. Evol. 2018;35:2145–2158.
    1. Ren B., Li W., Yang Y., Wu S. The impact of cyclin D1 overexpression on the prognosis of bladder cancer: a meta-analysis. World J. Surg. Oncol. 2014;12:55.
    1. Riaz N., Havel J.J., Kendall S.M., Makarov V., Walsh L.A., Desrichard A., Weinhold N., Chan T.A. Recurrent SERPINB3 and SERPINB4 mutations in patients who respond to anti-CTLA4 immunotherapy. Nat. Genet. 2016;48:1327–1329.
    1. Riaz N., Havel J.J., Makarov V., Desrichard A., Urba W.J., Sims J.S., Hodi F.S., Martín-Algarra S., Mandal R., Sharfman W.H. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171:934–949.e16.
    1. Rimmer A., Phan H., Mathieson I., Iqbal Z., Twigg S.R.F., Wilkie A.O.M., McVean G., Lunter G., WGS500 Consortium Integrating mapping-, assembly- and haplotype-based approaches for calling variants in clinical sequencing applications. Nat. Genet. 2014;46:912–918.
    1. Rizvi N.A., Hellmann M.D., Snyder A., Kvistborg P., Makarov V., Havel J.J., Lee W., Yuan J., Wong P., Ho T.S. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128.
    1. Robinson J., Soormally A.R., Hayhurst J.D., Marsh S.G.E. The IPD-IMGT/HLA database: new developments in reporting HLA variation. Hum. Immunol. 2016;77:233–237.
    1. Roh W., Chen P.L., Reuben A., Spencer C.N., Prieto P.A., Miller J.P., Gopalakrishnan V., Wang F., Cooper Z.A., Reddy S.M. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 2017;9:eaah3560.
    1. Rooney M.S., Shukla S.A., Wu C.J., Getz G., Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.
    1. Rosenthal R., McGranahan N., Herrero J., Taylor B.S., Swanton C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 2016;17:31.
    1. Rosenthal R., Cadieux E.L., Salgado R., Bakir M.A., Moore D.A., Hiley C.T., Lund T., Tanić M., Reading J.L., Joshi K., TRACERx consortium Neoantigen-directed immune escape in lung cancer evolution. Nature. 2019;567:479–485.
    1. Roth A., Khattra J., Yap D., Wan A., Laks E., Biele J., Ha G., Aparicio S., Bouchard-Côté A., Shah S.P. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods. 2014;11:396–398.
    1. Samstein R.M., Lee C.H., Shoushtari A.N., Hellmann M.D., Shen R., Janjigian Y.Y., Barron D.A., Zehir A., Jordan E.J., Omuro A. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 2019;51:202–206.
    1. Seiler R., Thalmann G.N., Rotzer D., Perren A., Fleischmann A. CCND1/CyclinD1 status in metastasizing bladder cancer: a prognosticator and predictor of chemotherapeutic response. Mod. Pathol. 2014;27:87–95.
    1. Shim J.H., Kim H.S., Cha H., Kim S., Kim T.M., Anagnostou V., Choi Y.L., Jung H.A., Sun J.M., Ahn J.S. HLA-corrected tumor mutation burden and homologous recombination deficiency for the prediction of response to PD-(L)1 blockade in advanced non-small-cell lung cancer patients. Ann. Oncol. 2020;31:902–911.
    1. Shin D.S., Zaretsky J.M., Escuin-Ordinas H., Garcia-Diaz A., Hu-Lieskovan S., Kalbasi A., Grasso C.S., Hugo W., Sandoval S., Torrejon D.Y. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7:188–201.
    1. Shrestha R., Nabavi N., Lin Y.Y., Mo F., Anderson S., Volik S., Adomat H.H., Lin D., Xue H., Dong X. BAP1 haploinsufficiency predicts a distinct immunogenic class of malignant peritoneal mesothelioma. Genome Med. 2019;11:8.
    1. Shukla S.A., Rooney M.S., Rajasagi M., Tiao G., Dixon P.M., Lawrence M.S., Stevens J., Lane W.J., Dellagatta J.L., Steelman S. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 2015;33:1152–1158.
    1. Smith J.C., Sheltzer J.M. Systematic identification of mutations and copy number alterations associated with cancer patient prognosis. eLife. 2018;7:e39217.
    1. Snyder A., Makarov V., Merghoub T., Yuan J., Zaretsky J.M., Desrichard A., Walsh L.A., Postow M.A., Wong P., Ho T.S. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014;371:2189–2199.
    1. Snyder A., Nathanson T., Funt S.A., Ahuja A., Buros Novik J., Hellmann M.D., Chang E., Aksoy B.A., Al-Ahmadie H., Yusko E. Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: an exploratory multi-omic analysis. PLoS Med. 2017;14:e1002309.
    1. Thommen D.S., Koelzer V.H., Herzig P., Roller A., Trefny M., Dimeloe S., Kiialainen A., Hanhart J., Schill C., Hess C. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 2018;24:994–1004.
    1. Tokunaga R., Nakagawa S., Sakamoto Y., Nakamura K., Naseem M., Izumi D., Kosumi K., Taki K., Higashi T., Miyata T. 12-Chemokine signature, a predictor of tumor recurrence in colorectal cancer. International Journal of Cancer. 2020;147:532–541.
    1. Tran E., Ahmadzadeh M., Lu Y.C., Gros A., Turcotte S., Robbins P.F., Gartner J.J., Zheng Z., Li Y.F., Ray S. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science. 2015;350:1387–1390.
    1. Trefny M.P., Rothschild S.I., Uhlenbrock F., Rieder D., Kasenda B., Stanczak M.A., Berner F., Kashyap A.S., Kaiser M., Herzig P. A variant of a killer cell immunoglobulin-like receptor is associated with resistance to PD-1 blockade in lung cancer. Clin. Cancer Res. 2019;25:3026–3034.
    1. Trucco L.D., Mundra P.A., Hogan K., Garcia-Martinez P., Viros A., Mandal A.K., Macagno N., Gaudy-Marqueste C., Allan D., Baenke F. Ultraviolet radiation-induced DNA damage is prognostic for outcome in melanoma. Nat. Med. 2019;25:221–224.
    1. Tumeh P.C., Harview C.L., Yearley J.H., Shintaku I.P., Taylor E.J., Robert L., Chmielowski B., Spasic M., Henry G., Ciobanu V. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571.
    1. Turajlic S., Litchfield K., Xu H., Rosenthal R., McGranahan N., Reading J.L., Wong Y.N.S., Rowan A., Kanu N., Al Bakir M. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 2017;18:1009–1021.
    1. Turajlic S., Xu H., Litchfield K., Rowan A., Chambers T., Lopez J.I., Nicol D., O’Brien T., Larkin J., Horswell S., PEACE. TRACERx Renal Consortium Tracking cancer evolution reveals constrained routes to metastases: TRACERx Renal. Cell. 2018;173:581–594.e12.
    1. Van Allen E.M., Miao D., Schilling B., Shukla S.A., Blank C., Zimmer L., Sucker A., Hillen U., Foppen M.H.G., Goldinger S.M. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207–211.
    1. Van Loo P., Nordgard S.H., Lingjærde O.C., Russnes H.G., Rye I.H., Sun W., Weigman V.J., Marynen P., Zetterberg A., Naume B. Allele-specific copy number analysis of tumors. Proc. Natl. Acad. Sci. USA. 2010;107:16910–16915.
    1. Vízkeleti L., Ecsedi S., Rákosy Z., Orosz A., Lázár V., Emri G., Koroknai V., Kiss T., Ádány R., Balázs M. The role of CCND1 alterations during the progression of cutaneous malignant melanoma. Tumour Biol. 2012;33:2189–2199.
    1. Vokes N.I., Liu D., Ricciuti B., Jimenez-Aguilar E., Rizvi H., Dietlein F., He M.X., Margolis C.A., Elmarakeby H.A., Girshman J. Harmonization of tumor mutational burden quantification and association with response to immune checkpoint blockade in non-small-cell lung cancer. JCO Precis. Oncol. 2019;3 PO.19.00171.
    1. Vredevoogd D.W., Kuilman T., Ligtenberg M.A., Boshuizen J., Stecker K.E., de Bruijn B., Krijgsman O., Huang X., Kenski J.C.N., Lacroix R. Augmenting immunotherapy impact by lowering tumor TNF cytotoxicity threshold. Cell. 2019;178:585–599.e15.
    1. Wang K., Li M., Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164.
    1. Wang M.T., Chen G., An S.J., Chen Z.H., Huang Z.M., Xiao P., Ben X.S., Xie Z., Chen S.L., Luo D.L. Prognostic significance of cyclinD1 amplification and the co-alteration of cyclinD1/pRb/ppRb in patients with esophageal squamous cell carcinoma. Dis. Esophagus. 2012;25:664–670.
    1. Wang L., Saci A., Szabo P.M., Chasalow S.D., Castillo-Martin M., Domingo-Domenech J., Siefker-Radtke A., Sharma P., Sfakianos J.P., Gong Y. EMT- and stroma-related gene expression and resistance to PD-1 blockade in urothelial cancer. Nat. Commun. 2018;9:3503.
    1. Wasserstein R.L., Schirm A.L., Lazar N.A. Moving to a world beyond “p < 0.05”. Am. Stat. 2019;73:1–19.
    1. Watkins T.B.K., Lim E.L., Petkovic M., Elizalde S., Birkbak N.J., Wilson G.A., Moore D.A., Grönroos E., Rowan A., Dewhurst S.M. Pervasive chromosomal instability and karyotype order in tumour evolution. Nature. 2020;587:126–132.
    1. Wolf Y., Bartok O., Patkar S., Eli G.B., Cohen S., Litchfield K., Levy R., Jiménez-Sánchez A., Trabish S., Lee J.S. UVB-induced tumor heterogeneity diminishes immune response in melanoma. Cell. 2019;179:219–235.e21.
    1. Yang W., Soares J., Greninger P., Edelman E.J., Lightfoot H., Forbes S., Bindal N., Beare D., Smith J.A., Thompson I.R. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955–D961.
    1. Zhang J., Bu X., Wang H., Zhu Y., Geng Y., Nihira N.T., Tan Y., Ci Y., Wu F., Dai X. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature. 2018;553:91–95.
    1. Zhao B., Sedlak J.C., Srinivas R., Creixell P., Pritchard J.R., Tidor B., Lauffenburger D.A., Hemann M.T. Exploiting temporal collateral sensitivity in tumor clonal evolution. Cell. 2016;165:234–246.

Source: PubMed

3
購読する