A Blood-based Assay for Assessment of Tumor Mutational Burden in First-line Metastatic NSCLC Treatment: Results from the MYSTIC Study

Purpose: Tumor mutational burden (TMB) has been shown to be predictive of survival benefit in patients with non–small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors. Measuring TMB in the blood (bTMB) using circulating cell-free tumor DNA (ctDNA) offers practical advantages compared with TMB measurement in tissue (tTMB); however, there is a need for validated assays and identification of optimal cutoffs. We describe the analytic validation of a new bTMB algorithm and its clinical utility using data from the phase III MYSTIC trial. Patients and Methods: The dataset used for the clinical validation was from MYSTIC, which evaluated first-line durvalumab (anti–PD-L1 antibody) ± tremelimumab (anticytotoxic T-lymphocyte-associated antigen-4 antibody) or chemotherapy for metastatic NSCLC. bTMB and tTMB were evaluated using the GuardantOMNI and FoundationOne CDx assays, respectively. A Cox proportional hazards model and minimal P value cross-validation approach were used to identify the optimal bTMB cutoff. Results: In MYSTIC, somatic mutations could be detected in ctDNA extracted from plasma samples in a majority of patients, allowing subsequent calculation of bTMB. The success rate for obtaining valid TMB scores was higher for bTMB (809/1,001; 81%) than for tTMB (460/735; 63%). Minimal P value cross-validation analysis confirmed the selection of bTMB ≥20 mutations per megabase (mut/Mb) as the optimal cutoff for clinical benefit with durvalumab + tremelimumab. Conclusions: Our study demonstrates the feasibility, accuracy, and reproducibility of the GuardantOMNI ctDNA platform for quantifying bTMB from plasma samples. Using the new bTMB algorithm and an optimal bTMB cutoff of ≥20 mut/Mb, high bTMB was predictive of clinical benefit with durvalumab + tremelimumab versus chemotherapy.

[1]  G. Giaccone,et al.  Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. , 2020, The New England journal of medicine.

[2]  S. Baylin,et al.  Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer , 2020, Nature Cancer.

[3]  C. Langer,et al.  MA25.04 Blood-Based Tumor Mutation Burden as a Predictive Biomarker for Outcomes After Pembrolizumab Based First Line Therapy in Metastatic NSCLC , 2019, Journal of Thoracic Oncology.

[4]  M. Socinski,et al.  Final efficacy results from B-F1RST, a prospective phase II trial evaluating blood-based tumour mutational burden (bTMB) as a predictive biomarker for atezolizumab (atezo) in 1L non-small cell lung cancer (NSCLC) , 2019, Annals of Oncology.

[5]  E. Helman,et al.  Abstract 3404: Landscape and genomic correlates of ctDNA-based tumor mutational burden across six solid tumor types , 2019, Molecular and Cellular Biology / Genetics.

[6]  Catalin C. Barbacioru,et al.  Abstract 1675: Analytical validation of MSI High detection with GuardantOMNI , 2019, Bioinformatics, Convergence Science, and Systems Biology.

[7]  Jeffrey H. Chuang,et al.  Mutations in DNA repair genes are associated with increased neoantigen burden and a distinct immunophenotype in lung squamous cell carcinoma , 2019, Scientific Reports.

[8]  Matthew D. Hellmann,et al.  First-Line Nivolumab Plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer (CheckMate 568): Outcomes by Programmed Death Ligand 1 and Tumor Mutational Burden as Biomarkers , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[9]  F. López-Ríos,et al.  Implementing TMB measurement in clinical practice: considerations on assay requirements , 2019, ESMO Open.

[10]  Jing Zhao,et al.  Development and analytical validation of a plasma-based tumor mutational burden (TMB) score from next-generation sequencing panels. , 2018, Annals of oncology : official journal of the European Society for Medical Oncology.

[11]  Jacob Silterra,et al.  Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab , 2018, Nature Medicine.

[12]  R. Motzer,et al.  Immune Checkpoint Blockade in Advanced Renal-Cell Carcinoma. , 2018, The New England journal of medicine.

[13]  E. Helman,et al.  Abstract 4272: A novel approach to differentiation of somatic vs. germline variants in liquid biopsies using a betabinomial model , 2018, Bioinformatics and Systems Biology.

[14]  Shahrooz Rabizadeh,et al.  Three-fold overestimation of tumor mutation burden using 248 gene panel versus whole exome. , 2018 .

[15]  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.

[16]  G. Mills,et al.  ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade , 2018, Nature Medicine.

[17]  C. Paweletz,et al.  Validation of a Plasma-Based Comprehensive Cancer Genotyping Assay Utilizing Orthogonal Tissue- and Plasma-Based Methodologies , 2018, Clinical Cancer Research.

[18]  S. Novello,et al.  Pembrolizumab plus Chemotherapy in Metastatic Non–Small‐Cell Lung Cancer , 2018, The New England journal of medicine.

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

[20]  Arnaud Boyer,et al.  Durvalumab after chemoradiotherapy in stage III non-small cell lung cancer. , 2018, Journal of thoracic disease.

[21]  Maximilian Diehn,et al.  Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. , 2018, Archives of pathology & laboratory medicine.

[22]  P. Hegde,et al.  Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial , 2018, The Lancet.

[23]  H. Groen,et al.  Mutations in EMT-Related Genes in ALK Positive Crizotinib Resistant Non-Small Cell Lung Cancers , 2018, Cancers.

[24]  E. Jaffee,et al.  Tumor Mutational Burden and Response Rate to PD-1 Inhibition. , 2017, The New England journal of medicine.

[25]  S. Mortimer,et al.  The Landscape of Actionable Genomic Alterations in Cell-Free Circulating Tumor DNA from 21,807 Advanced Cancer Patients , 2017, Clinical Cancer Research.

[26]  D. Carbone,et al.  First-Line Nivolumab in Stage IV or Recurrent Non-Small Cell Lung Cancer , 2017 .

[27]  Nicolai J. Birkbak,et al.  Tracking the Evolution of Non‐Small‐Cell Lung Cancer , 2017, The New England journal of medicine.

[28]  Ashwini Naik,et al.  Phylogenetic ctDNA analysis depicts early stage lung cancer evolution , 2017, Nature.

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

[30]  R. Pe Benito,et al.  Genomic Profiling of Advanced Non–Small Cell Lung Cancer in Community Settings: Gaps and Opportunities , 2017, Clinical lung cancer.

[31]  E. Jaffee,et al.  Targeting neoantigens to augment antitumour immunity , 2017, Nature Reviews Cancer.

[32]  Anil Vachani,et al.  Detection of Therapeutically Targetable Driver and Resistance Mutations in Lung Cancer Patients by Next-Generation Sequencing of Cell-Free Circulating Tumor DNA , 2016, Clinical Cancer Research.

[33]  B. Kermani,et al.  Analytical and Clinical Validation of a Digital Sequencing Panel for Quantitative, Highly Accurate Evaluation of Cell-Free Circulating Tumor DNA , 2015, PloS one.

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

[35]  Min Zhao,et al.  dbEMT: an epithelial-mesenchymal transition associated gene resource , 2015, Scientific Reports.

[36]  B. Vogelstein,et al.  PD-1 blockade in tumors with mismatch repair deficiency. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[37]  T. Schumacher,et al.  Neoantigens in cancer immunotherapy , 2015, Science.

[38]  M. Millenson,et al.  PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. , 2015, The New England journal of medicine.

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

[40]  Z. Szallasi,et al.  Spatial and temporal diversity in genomic instability processes defines lung cancer evolution , 2014, Science.

[41]  Alex M. Fichtenholtz,et al.  Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing , 2013, Nature Biotechnology.

[42]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

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

[44]  Steven A. Rosenberg,et al.  Adoptive immunotherapy for cancer: harnessing the T cell response , 2012, Nature Reviews Immunology.

[45]  P. A. Futreal,et al.  Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. , 2012, The New England journal of medicine.

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

[47]  W. Sauerbrei,et al.  Dangers of using "optimal" cutpoints in the evaluation of prognostic factors. , 1994, Journal of the National Cancer Institute.

[48]  Andrew Dunford,et al.  Genomic Heterogeneity as a Barrier to Precision Medicine in Gastroesophageal Adenocarcinoma. , 2018, Cancer discovery.

[49]  Richard B. Lanman,et al.  Costs of Diagnostic Assessment for Lung Cancer: A Medicare Claims Analysis , 2017, Clinical lung cancer.