What makes TMB an ambivalent biomarker for immunotherapy? A subtle mismatch between the sample-based design of variant callers and real clinical cohort

Tumor mutation burden (TMB) is a widely recognized biomarker for predicting the efficacy of immunotherapy. However, its use still remains highly controversial. In this study, we examine the underlying causes of this controversy based on clinical needs. By tracing the source of the TMB errors and analyzing the design philosophy behind variant callers, we identify the conflict between the incompleteness of biostatistics rules and the variety of clinical samples as the critical issue that renders TMB an ambivalent biomarker. A series of experiments were conducted to illustrate the challenges of mutation detection in clinical practice. Additionally, we also discuss potential strategies for overcoming these conflict issues to enable the application of TMB in guiding decision-making in real clinical settings.

[1]  Jiayin Wang,et al.  Is an SV caller compatible with sequencing data? An online recommendation tool to automatically recommend the optimal caller based on data features , 2023, Frontiers in Genetics.

[2]  Jiayin Wang,et al.  TMBcat: A multi-endpoint p-value criterion on different discrepancy metrics for superiorly inferring tumor mutation burden thresholds , 2022, Frontiers in Immunology.

[3]  Jiayin Wang,et al.  PEcnv: accurate and efficient detection of copy number variations of various lengths , 2022, Briefings Bioinform..

[4]  Yangguang Shi,et al.  Abstract LB512: RATIONALE-304: The association of tumor mutational burden (TMB) with clinical outcomes of tislelizumab (TIS) + chemotherapy (chemo) versus chemo alone as first-line treatment for advanced non-squamous non-small cell lung cancer (nsq-NSCLC) , 2022, Cancer Research.

[5]  Jiaxuan Li,et al.  Ensemble of ML-KNN for classification algorithm recommendation , 2021, Knowl. Based Syst..

[6]  A. Jewett,et al.  Immunotherapy of Glioblastoma: Current Strategies and Challenges in Tumor Model Development , 2021, Cells.

[7]  Manasi Sheth,et al.  Exploring the relationship between Overall Survival (OS), Progression Free Survival (PFS) and Objective Response Rate (ORR) in patients with advanced melanoma. , 2020, Cancer treatment and research communications.

[8]  T. Chan,et al.  The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: a decision centered on empowering patients and their physicians. , 2020, Annals of oncology : official journal of the European Society for Medical Oncology.

[9]  Y. Bang,et al.  The association of tissue tumor mutational burden (tTMB) using the Foundation Medicine genomic platform with efficacy of pembrolizumab versus paclitaxel in patients (pts) with gastric cancer (GC) from KEYNOTE-061. , 2020 .

[10]  S. Nomura,et al.  TAS-116, an oral HSP90 inhibitor, in combination with nivolumab in patients with colorectal cancer and other solid tumors: An open-label, dose-finding, and expansion phase Ib trial (EPOC1704). , 2020 .

[11]  A. Matakidou,et al.  Molecular biomarkers to identify patients (pts) who may benefit from durvalumab (D; anti-PD-L1) ± tremelimumab (T; anti-CTLA-4) in recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) from HAWK and CONDOR studies. , 2020 .

[12]  R. Ferris,et al.  Plasma-based tumor mutational burden (bTMB) as predictor for survival in phase III EAGLE study: Durvalumab (D) ± tremelimumab (T) versus chemotherapy (CT) in recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC) after platinum failure. , 2020 .

[13]  Y. Bang,et al.  The association of molecular biomarkers with efficacy of pembrolizumab versus paclitaxel in patients with gastric cancer (GC) from KEYNOTE-061. , 2020 .

[14]  K. Bensalah,et al.  Immunotherapy in Renal Cell Carcinoma: The Future Is Now , 2020, International journal of molecular sciences.

[15]  M. Narukawa,et al.  Magnitude of advantage in tumor response contributes to a better correlation between treatment effects on overall survival and progression-free survival: a literature-based meta-analysis of clinical trials in patients with metastatic colorectal cancer , 2020, International Journal of Clinical Oncology.

[16]  L. Diaz,et al.  Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[17]  Tae-Min Kim,et al.  False-negative errors in next-generation sequencing contribute substantially to inconsistency of mutation databases , 2019, PloS one.

[18]  G. Colloca,et al.  Analysis of response-related endpoints in trials of first-line medical treatment of metastatic colorectal cancer , 2019, International Journal of Clinical Oncology.

[19]  Julianne K. David,et al.  Burden of tumor mutations, neoepitopes, and other variants are weak predictors of cancer immunotherapy response and overall survival , 2019, Genome Medicine.

[20]  Y. Kamatani,et al.  Comprehensive evaluation of structural variation detection algorithms for whole genome sequencing , 2019, Genome Biology.

[21]  C. Brennan,et al.  Tumor mutational load predicts survival after immunotherapy across multiple cancer types , 2019, Nature Genetics.

[22]  J. Lunceford,et al.  Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy , 2018, Science.

[23]  Thomas Colthurst,et al.  A universal SNP and small-indel variant caller using deep neural networks , 2018, Nature Biotechnology.

[24]  Martin Posch,et al.  Methods for the analysis of multiple endpoints in small populations: A review , 2018, Journal of biopharmaceutical statistics.

[25]  R. Bourgon,et al.  Association of high tissue TMB and atezolizumab efficacy across multiple tumor types. , 2018 .

[26]  D. Schadendorf,et al.  Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors , 2018, Nature Genetics.

[27]  G. Getz,et al.  Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors , 2018, Nature Genetics.

[28]  Arun Ahuja,et al.  Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer , 2018, Cancer cell.

[29]  Paolo A Ascierto,et al.  Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. , 2018, Cancer cell.

[30]  J. Szustakowski,et al.  Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden , 2018, The New England journal of medicine.

[31]  L. Macconaill,et al.  Frameshift events predict anti-PD-1/L1 response in head and neck cancer. , 2018, JCI insight.

[32]  Mark W. Ball,et al.  Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma , 2018, Science.

[33]  Chang Xu,et al.  A review of somatic single nucleotide variant calling algorithms for next-generation sequencing data , 2018, Computational and structural biotechnology journal.

[34]  Renke Pan,et al.  TNscope: Accurate Detection of Somatic Mutations with Haplotype-based Variant Candidate Detection and Machine Learning Filtering , 2018, bioRxiv.

[35]  T. Chan,et al.  Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab , 2017, Cell.

[36]  P. Stephens,et al.  Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers , 2017, Molecular Cancer Therapeutics.

[37]  M. Socinski,et al.  First‐Line Nivolumab in Stage IV or Recurrent Non–Small‐Cell Lung Cancer , 2017, The New England journal of medicine.

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

[39]  Daniel Sinnett,et al.  SNooPer: a machine learning-based method for somatic variant identification from low-pass next-generation sequencing , 2016, BMC Genomics.

[40]  S. Chanock,et al.  Burden of Nonsynonymous Mutations among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. , 2016, Cancer research.

[41]  Philip J. Stephens,et al.  Total mutation burden (TMB) in lung cancer (LC) and relationship with response to PD-1/PD-L1 targeted therapies. , 2016 .

[42]  Emmanuel Barillot,et al.  SV-Bay: structural variant detection in cancer genomes using a Bayesian approach with correction for GC-content and read mappability , 2016, Bioinform..

[43]  Michael C. Heinold,et al.  A comprehensive assessment of somatic mutation detection in cancer using whole-genome sequencing , 2015, Nature Communications.

[44]  Jatinder Singh International conference on harmonization of technical requirements for registration of pharmaceuticals for human use , 2015, Journal of pharmacology & pharmacotherapeutics.

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

[46]  P. Sharma,et al.  Immune Checkpoint Targeting in Cancer Therapy: Toward Combination Strategies with Curative Potential , 2015, Cell.

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

[48]  R. Satya,et al.  Comparison of somatic mutation calling methods in amplicon and whole exome sequence data , 2014, BMC Genomics.

[49]  Ryan M. Layer,et al.  LUMPY: a probabilistic framework for structural variant discovery , 2012, Genome Biology.

[50]  V. Beneš,et al.  DELLY: structural variant discovery by integrated paired-end and split-read analysis , 2012, Bioinform..

[51]  George Coukos,et al.  Cancer immunotherapy comes of age , 2011, Nature.

[52]  Gholamreza Haffari,et al.  Feature-based classifiers for somatic mutation detection in tumour–normal paired sequencing data , 2011, Bioinform..

[53]  R. Wilson,et al.  BreakDancer: An algorithm for high resolution mapping of genomic structural variation , 2009, Nature Methods.

[54]  Raymond J. Carroll,et al.  Measurement error in nonlinear models: a modern perspective , 2006 .

[55]  Leslie G. Valiant,et al.  Computational limitations on learning from examples , 1988, JACM.

[56]  M. Postma,et al.  Do Surrogate Endpoints Better Correlate with Overall Survival in Studies That Did Not Allow for Crossover or Reported Balanced Postprogression Treatments? An Application in Advanced Non-Small Cell Lung Cancer. , 2018, Value in health : the journal of the International Society for Pharmacoeconomics and Outcomes Research.

[57]  Leslie G. Valiant,et al.  A theory of the learnable , 1984, CACM.