Mutation load estimation model as a predictor of the response to cancer immunotherapy

The determination of the mutation load, a total number of nonsynonymous point mutations, by whole-exome sequencing was shown to be useful in predicting the treatment responses to cancer immunotherapy. However, this technique is expensive and time-consuming, which hampers its application in clinical practice. Therefore, the objective of this study was to construct a mutation load estimation model for lung adenocarcinoma, using a small set of genes, as a predictor of the immunotherapy treatment response. Using the somatic mutation data downloaded from The Cancer Genome Atlas (TCGA) database, a computational framework was developed. The estimation model consisted of only 24 genes, used to estimate the mutation load in the independent validation cohort precisely (R2 = 0.7626). Additionally, the estimated mutation load can be used to identify the patients with durable clinical benefits, with 85% sensitivity, 93% specificity, and 89% accuracy, indicating that the model can serve as a predictive biomarker for cancer immunotherapy treatment response. Furthermore, our analyses demonstrated the necessity of the cancer-specific models by the constructed melanoma and colorectal models. Since most genes in the lung adenocarcinoma model are not currently included in the sequencing panels, a customized targeted sequencing panel can be designed with the selected model genes to assess the mutation load, instead of whole-exome sequencing or the currently used panel-based methods. Consequently, the cost and time required for the assessment of mutation load may be considerably decreased, which indicates that the presented model is a more cost-effective approach to cancer immunotherapy response prediction in clinical practice.Cancer genetics: Predicting patient response to immunotherapyEstimating patients’ mutation load from a small set of genes can accurately predict their response to cancer immunotherapy. Harnessing patients’ immune response to target tumor cells is an effective treatment approach in some cases but not others. A patient’s number of deleterious genetic mutations across all their protein-coding genes has been shown to correlate with their responsiveness to immunotherapy. However, whole-exome sequencing is time-consuming and costly. Yu-Chao Wang at the National Yang-Ming University, Taiwan, and colleagues have developed cancer-specific mutation load estimation models for adenocarcinoma, melanoma and colorectal cancer that require sequencing only a small number of genes. They show that the mutation load in lung adenocarcinoma patients can be estimated from 24 genes and that they can predict immunotherapy responsiveness with similar accuracy to that obtained using whole-exome sequencing.

[1]  S. Rosenberg,et al.  CTLA-4 Blockade with Ipilimumab: Long-term Follow-up of 177 Patients with Metastatic Melanoma , 2012, Clinical Cancer Research.

[2]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[3]  Yuan Qi,et al.  Clinical actionability enhanced through deep targeted sequencing of solid tumors. , 2015, Clinical chemistry.

[4]  Steven J. M. Jones,et al.  Comprehensive molecular characterization of human colon and rectal cancer , 2012, Nature.

[5]  Gert Jan van der Wilt,et al.  Is the $1000 Genome as Near as We Think? A Cost Analysis of Next-Generation Sequencing. , 2016, Clinical chemistry.

[6]  Pornpimol Charoentong,et al.  Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade , 2016, bioRxiv.

[7]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[8]  Steven J. M. Jones,et al.  Genomic Classification of Cutaneous Melanoma , 2015, Cell.

[9]  J. Taube,et al.  Antagonists of PD-1 and PD-L1 in Cancer Treatment. , 2015, Seminars in oncology.

[10]  J. Wolchok,et al.  Genetic basis for clinical response to CTLA-4 blockade in melanoma. , 2014, The New England journal of medicine.

[11]  Angela N. Brooks,et al.  Mapping the Hallmarks of Lung Adenocarcinoma with Massively Parallel Sequencing , 2012, Cell.

[12]  International Human Genome Sequencing Consortium Finishing the euchromatic sequence of the human genome , 2004 .

[13]  A. Ribas,et al.  Anti-programmed cell death protein-1/ligand-1 therapy in different cancers , 2015, British Journal of Cancer.

[14]  Syed Haider,et al.  Ensembl BioMarts: a hub for data retrieval across taxonomic space , 2011, Database J. Biol. Databases Curation.

[15]  C. Drake,et al.  Immune checkpoint blockade: a common denominator approach to cancer therapy. , 2015, Cancer cell.

[16]  Eric S. Lander,et al.  Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma , 2016, Cell reports.

[17]  J. Taube,et al.  Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy , 2016, Nature Reviews Cancer.

[18]  Jerald B. Johnson,et al.  Model selection in ecology and evolution. , 2004, Trends in ecology & evolution.

[19]  Melanie A. Huntley,et al.  Recurrent R-spondin fusions in colon cancer , 2012, Nature.

[20]  Jorge Sabbaga,et al.  Comprehensive cancer-gene panels can be used to estimate mutational load and predict clinical benefit to PD-1 blockade in clinical practice , 2015, Oncotarget.

[21]  Nicolai J. Birkbak,et al.  Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade , 2016, Science.

[22]  Levi Garraway,et al.  Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden , 2017, Genome Medicine.

[23]  Steven J. M. Jones,et al.  Comprehensive molecular profiling of lung adenocarcinoma , 2014, Nature.

[24]  James R. Eshleman,et al.  Microsatellite Instability as a Biomarker for PD-1 Blockade , 2016, Clinical Cancer Research.

[25]  Tim Hubbard Finishing the euchromatic sequence of the human genome , 2004 .

[26]  M. Atkins,et al.  Predictive biomarkers for checkpoint inhibitor-based immunotherapy. , 2016, The Lancet. Oncology.

[27]  Donavan T. Cheng,et al.  Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology. , 2015, The Journal of molecular diagnostics : JMD.

[28]  Maxim N. Artyomov,et al.  Checkpoint Blockade Cancer Immunotherapy Targets Tumour-Specific Mutant Antigens , 2014, Nature.

[29]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[30]  J. Wolchok,et al.  Immune Checkpoint Blockade in Cancer Therapy. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[31]  J. Sosman,et al.  Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma , 2016, Cell.

[32]  D. Schadendorf,et al.  Improved survival with ipilimumab in patients with metastatic melanoma. , 2010, The New England journal of medicine.

[33]  S. Gabriel,et al.  Genomic correlates of response to CTLA-4 blockade in metastatic melanoma , 2015, Science.

[34]  B. Morrow,et al.  Development of a Targeted Multi-Disorder High-Throughput Sequencing Assay for the Effective Identification of Disease-Causing Variants , 2015, PLoS ONE.

[35]  Yu Shyr,et al.  Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade , 2016, Cancer Immunology Research.

[36]  P. A. Futreal,et al.  Novel algorithmic approach predicts tumor mutation load and correlates with immunotherapy clinical outcomes using a defined gene mutation set , 2016, BMC Medicine.