Implementing TMB measurement in clinical practice: considerations on assay requirements

Clinical evidence demonstrates that treatment with immune checkpoint inhibitor immunotherapy agents can have considerable benefit across multiple tumours. However, there is a need for the development of predictive biomarkers that identify patients who are most likely to respond to immunotherapy. Comprehensive characterisation of tumours using genomic, transcriptomic, and proteomic approaches continues to lead the way in advancing precision medicine. Genetic correlates of response to therapy have been known for some time, but recent clinical evidence has strengthened the significance of high tumour mutational burden (TMB) as a biomarker of response and hence a rational target for immunotherapy. Concordantly, immune checkpoint inhibitors have changed clinical practice for lung cancer and melanoma, which are tumour types with some of the highest mutational burdens. TMB is an implementable approach for molecular biology and/or pathology laboratories that provides a quantitative measure of the total number of mutations in tumour tissue of patients and can be assessed by whole genome, whole exome, or large targeted gene panel sequencing of biopsied material. Currently, TMB assessment is not standardised across research and clinical studies. As a biomarker that affects treatment decisions, it is essential to unify TMB assessment approaches to allow for reliable, comparable results across studies. When implementing TMB measurement assays, it is important to consider factors that may impact the method workflow, the results of the assay, and the interpretation of the data. Such factors include biopsy sample type, sample quality and quantity, genome coverage, sequencing platform, bioinformatic pipeline, and the definitions of the final threshold that determines high TMB. This review outlines the factors for adoption of TMB measurement into clinical practice, providing an understanding of TMB assay considerations throughout the sample journey, and suggests principles to effectively implement TMB assays in a clinical setting to aid and optimise treatment decisions.

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

[2]  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.

[3]  J. Szustakowski,et al.  STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. , 2018, Cancer discovery.

[4]  Wendell D. Jones,et al.  Abstract LB-380: TMB calling in absence of matched normal: Learning germline behavior , 2018, Molecular and Cellular Biology / Genetics.

[5]  V. Weigman,et al.  Abstract LB-381: Effects of sequencing parameters and panel size on mutational burden calculations , 2018, Molecular and Cellular Biology / Genetics.

[6]  Wendell D. Jones,et al.  Abstract 2250: Tumor mutational burden: Guidelines for derivation and robustness of measurement , 2018, Bioinformatics and Systems Biology.

[7]  J. Szustakowski,et al.  Abstract 5528: Evaluation of tumor mutation burden as a biomarker for immune checkpoint inhibitor efficacy: A calibration study of whole exome sequencing with FoundationOne® , 2018, Clinical Research (Excluding Clinical Trials).

[8]  S. Bilke,et al.  Abstract 435: Accurate measurement of tumor mutation burden through tumor-only sequencing using a 500-gene panel , 2018, Molecular and Cellular Biology / Genetics.

[9]  Matthew D. Hellmann,et al.  Abstract CT078: Tumor mutational burden (TMB) as a biomarker for clinical benefit from dual immune checkpoint blockade with nivolumab (nivo) + ipilimumab (ipi) in first-line (1L) non-small cell lung cancer (NSCLC): identification of TMB cutoff from CheckMate 568 , 2018, Clinical Trials.

[10]  D. Lipson,et al.  Abstract 5706: A blood-based next-generation sequencing assay to determine tumor mutational burden (bTMB) is associated with benefit to an anti-PD-L1 inhibitor, atezolizumab , 2018, Immunology.

[11]  Edward S. Kim,et al.  Prospective clinical evaluation of blood-based tumor mutational burden (bTMB) as a predictive biomarker for atezolizumab (atezo) in 1L non-small cell lung cancer (NSCLC): Interim B-F1RST results. , 2018 .

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

[13]  P. Philip,et al.  Safety and Efficacy of Pembrolizumab Monotherapy in Patients With Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial , 2018, JAMA oncology.

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

[15]  Y. Shao,et al.  Clinicopathological characteristics of POLE mutation in patients with non-small-cell lung cancer. , 2018, Lung cancer.

[16]  Brooke L. Billman,et al.  Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[17]  J. Lunceford,et al.  Genomic biomarkers in relation to PD-1 checkpoint blockade response. , 2018 .

[18]  P. Hegde,et al.  Atezolizumab (atezo) vs. chemotherapy (chemo) in platinum-treated locally advanced or metastatic urothelial carcinoma (mUC): Immune biomarkers, tumor mutational burden (TMB), and clinical outcomes from the phase III IMvigor211 study. , 2018 .

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

[20]  Philip J. Stephens,et al.  A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal , 2018, PLoS Comput. Biol..

[21]  Ahmet Zehir,et al.  Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[23]  Larissa V Furtado,et al.  Feasibility of Endobronchial Ultrasound‐guided Transbronchial Needle Aspiration Cytology Specimens for Next Generation Sequencing in Non–small‐cell Lung Cancer , 2017, Clinical lung cancer.

[24]  S. Sleijfer,et al.  Molecular Tumor Boards: current practice and future needs , 2017, Annals of oncology : official journal of the European Society for Medical Oncology.

[25]  M. Ringnér,et al.  Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma , 2017, Nature Communications.

[26]  Etienne Rouleau,et al.  Whole exome sequencing for determination of tumor mutation load in liquid biopsy from advanced cancer patients , 2017, PloS one.

[27]  K. Cole,et al.  Comprehensive Analysis of Hypermutation in Human Cancer , 2017, Cell.

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

[29]  A. Levine,et al.  A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy , 2017, Nature.

[30]  I. Wistuba,et al.  Detection of somatic mutations in cell-free DNA in plasma and correlation with overall survival in patients with solid tumors , 2017, Oncotarget.

[31]  K. Bloom,et al.  Programmed Death-Ligand 1 Immunohistochemistry Testing: A Review of Analytical Assays and Clinical Implementation in Non-Small-Cell Lung Cancer. , 2017, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[32]  Russell Bonneville,et al.  Landscape of Microsatellite Instability Across 39 Cancer Types. , 2017, JCO precision oncology.

[33]  J. T. Jørgensen,et al.  Companion and Complementary Diagnostics–Focus on PD-L1 Expression Assays for PD-1/PD-L1 Checkpoint Inhibitors in Non–Small Cell Lung Cancer , 2017, Therapeutic drug monitoring.

[34]  M. Tiemann,et al.  Concordance between Comprehensive Cancer Genome Profiling in Plasma and Tumor Specimens , 2017, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[35]  Razelle Kurzrock,et al.  Hypermutated Circulating Tumor DNA: Correlation with Response to Checkpoint Inhibitor–Based Immunotherapy , 2017, Clinical Cancer Research.

[36]  L. Sholl Next-Generation Sequencing from Liquid Biopsies in Lung Cancer Patients: Advances in Comprehensive Biomarker Testing. , 2017, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[37]  B. Levy,et al.  Beyond PD-L1 testing-emerging biomarkers for immunotherapy in non-small cell lung cancer. , 2017, Annals of translational medicine.

[38]  L. Rubinstein,et al.  The Challenge for Development of Valuable Immuno-oncology Biomarkers , 2017, Clinical Cancer Research.

[39]  Matthew D. Galsky,et al.  848PDImpact of zumor mutation burden on nivolumab efficacy in second-line urothelial carcinoma patients: Exploratory analysis of the phase ii checkmate 275 study , 2017 .

[40]  Seung-Ho Shin,et al.  A Method to Evaluate the Quality of Clinical Gene-Panel Sequencing Data for Single-Nucleotide Variant Detection. , 2017, The Journal of molecular diagnostics : JMD.

[41]  D. Lipson,et al.  102PAnalytic validation of a next generation sequencing assay to identify tumor mutational burden from blood (bTMB) to support investigation of an anti-PD-L1 agent, atezolizumab, in a first line non-small cell lung cancer trial (BFAST) , 2017 .

[42]  Edward S. Kim,et al.  1383TiPBlood first line ready screening trial (B-F1RST) and blood first assay screening trial (BFAST) enable clinical development of novel blood-based biomarker assays for tumor mutational burden (TMB) and somatic mutations in 1L advanced or metastatic NSCLC , 2017 .

[43]  A. Abernethy,et al.  1139PDAnalyzing biomarkers of cancer immunotherapy (CIT) response using a real-world clinico-genomic database , 2017 .

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

[45]  Ying Sun,et al.  Genomic Analysis of Tumor Microenvironment Immune Types across 14 Solid Cancer Types: Immunotherapeutic Implications , 2017, Theranostics.

[46]  J. Taube,et al.  A Prospective, Multi-institutional, Pathologist-Based Assessment of 4 Immunohistochemistry Assays for PD-L1 Expression in Non–Small Cell Lung Cancer , 2017, JAMA oncology.

[47]  Kenji Suzuki,et al.  DNA mismatch repair deficiency in surgically resected lung adenocarcinoma: Microsatellite instability analysis using the Promega panel. , 2017, Lung cancer.

[48]  Ludmila V. Danilova,et al.  Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade , 2017, Science.

[49]  G. Curigliano,et al.  Mismatch Repair Deficiency as a Predictive Biomarker for Immunotherapy Efficacy , 2017, BioMed research international.

[50]  T. Stricker,et al.  Abstract LB-105: Characterization of total mutational burden in the GENIE cohort: Small and large panels can provide TMB information but to varying degrees , 2017 .

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

[52]  Ioana R. Bonta,et al.  Correlation between tumor mutation burden and response to immunotherapy. , 2017 .

[53]  B. Somer,et al.  Spectrum of tumor mutational load (TML) in genitourinary cancers (GU CA). , 2017 .

[54]  T. Taxter,et al.  Comparison of tumor mutational burden (TMB) across tumor tissue and circulating tumor DNA (ctDNA). , 2017 .

[55]  Ash A. Alizadeh,et al.  Analysis of circulating tumor DNA in localized lung cancer for detection of molecular residual disease and personalization of adjuvant strategies. , 2017 .

[56]  Y. Sasaki,et al.  Assessment of the quality of DNA from various formalin-fixed paraffin-embedded (FFPE) tissues and the use of this DNA for next-generation sequencing (NGS) with no artifactual mutation , 2017, PloS one.

[57]  Marina N Nikiforova,et al.  Guidelines for Validation of Next-Generation Sequencing-Based Oncology Panels: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists. , 2017, The Journal of molecular diagnostics : JMD.

[58]  M. Vignali,et al.  Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis , 2017, PLoS medicine.

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

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

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

[62]  Donavan T. Cheng,et al.  Mutational Landscape of Metastatic Cancer Revealed from Prospective Clinical Sequencing of 10,000 Patients , 2017, Nature Medicine.

[63]  J. Heymann,et al.  Next-generation sequencing of non-small cell lung cancer using a customized, targeted sequencing panel: Emphasis on small biopsy and cytology , 2017, CytoJournal.

[64]  P. A. Futreal,et al.  Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance , 2017, Science Translational Medicine.

[65]  N. Peled,et al.  Clinical Impact of Hybrid Capture–Based Next‐Generation Sequencing on Changes in Treatment Decisions in Lung Cancer , 2017, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[66]  Yan Guo,et al.  The Utilization of Formalin Fixed-Paraffin-Embedded Specimens in High Throughput Genomic Studies , 2017, International journal of genomics.

[67]  J. Sosman,et al.  Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma , 2017, Cell.

[68]  R. Bourgon,et al.  Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial , 2017, The Lancet.

[69]  Bernat Gel,et al.  Benchmarking of Whole Exome Sequencing and Ad Hoc Designed Panels for Genetic Testing of Hereditary Cancer , 2017, Scientific Reports.

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

[71]  Jacqueline A. Hall,et al.  Quality to rely on: meeting report of the 5th Meeting of External Quality Assessment, Naples 2016 , 2016, ESMO Open.

[72]  N. Jamieson,et al.  Gene expression profiling to predict responsiveness to immunotherapy , 2016, Cancer Gene Therapy.

[73]  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.

[74]  C. Horak,et al.  Baseline tumor T cell receptor (TcR) sequencing analysis and neo antigen load is associated with benefit in melanoma patients receiving sequential nivolumab and ipilimumab , 2016 .

[75]  D. Merico,et al.  Immune Checkpoint Inhibition for Hypermutant Glioblastoma Multiforme Resulting From Germline Biallelic Mismatch Repair Deficiency. , 2016, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[76]  Etienne Rouleau,et al.  Integration of next-generation sequencing in clinical diagnostic molecular pathology laboratories for analysis of solid tumours; an expert opinion on behalf of IQN Path ASBL , 2016, Virchows Archiv.

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

[78]  M. Dimopoulos,et al.  DNA damage, tumor mutational load and their impact on immune responses against cancer. , 2016, Annals of translational medicine.

[79]  Nikhil Wagle,et al.  The impact of tumor profiling approaches and genomic data strategies for cancer precision medicine , 2016, Genome Medicine.

[80]  C. Lefebvre,et al.  Mutational Landscape and Sensitivity to Immune Checkpoint Blockers , 2016, Clinical Cancer Research.

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

[82]  Daniel S. Chen,et al.  Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). , 2016, Annals of oncology : official journal of the European Society for Medical Oncology.

[83]  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 .

[84]  R. Bourgon,et al.  Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial , 2016, The Lancet.

[85]  Shivang Doshi,et al.  Genomic Sequencing Procedure Microcosting Analysis and Health Economic Cost-Impact Analysis , 2016, The Journal of molecular diagnostics : JMD.

[86]  M. Marton,et al.  Data Interoperability of Whole Exome Sequencing (WES) Based Mutational Burden Estimates from Different Laboratories , 2016, International journal of molecular sciences.

[87]  I. Wistuba,et al.  Optimizing the DNA yield for molecular analysis from cytologic preparations , 2016, Cancer cytopathology.

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

[89]  Samuel P. Strom Current practices and guidelines for clinical next-generation sequencing oncology testing , 2016, Cancer biology & medicine.

[90]  S. Nishizuka,et al.  Individualized Mutation Detection in Circulating Tumor DNA for Monitoring Colorectal Tumor Burden Using a Cancer-Associated Gene Sequencing Panel , 2016, PloS one.

[91]  A. Papavassiliou,et al.  The biological complexity of urothelial carcinoma: Insights into carcinogenesis, targets and biomarkers of response to therapeutic approaches. , 2015, Seminars in cancer biology.

[92]  Tudung T Nguyen,et al.  Identification of major factors associated with failed clinical molecular oncology testing performed by next generation sequencing (NGS) , 2015, Molecular oncology.

[93]  L. Moens,et al.  HaloPlex Targeted Resequencing for Mutation Detection in Clinical Formalin-Fixed, Paraffin-Embedded Tumor Samples. , 2015, The Journal of molecular diagnostics : JMD.

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

[95]  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.

[96]  R. Goswami,et al.  Analysis of Pre-Analytic Factors Affecting the Success of Clinical Next-Generation Sequencing of Solid Organ Malignancies , 2015, Cancers.

[97]  Bert Vogelstein,et al.  PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. , 2015, The New England journal of medicine.

[98]  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.

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

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

[101]  N. Hacohen,et al.  Molecular and Genetic Properties of Tumors Associated with Local Immune Cytolytic Activity , 2015, Cell.

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

[103]  William J. Howat,et al.  Tissue fixation and the effect of molecular fixatives on downstream staining procedures , 2014, Methods.

[104]  Etienne Rouleau,et al.  Guidance for laboratories performing molecular pathology for cancer patients , 2014, Journal of Clinical Pathology.

[105]  Russell Weiner,et al.  Navigating the Rapids: The Development of Regulated Next-Generation Sequencing-Based Clinical Trial Assays and Companion Diagnostics , 2014, Front. Oncol..

[106]  P. Coulie,et al.  Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy , 2014, Nature Reviews Cancer.

[107]  C. Ponting,et al.  Sequencing depth and coverage: key considerations in genomic analyses , 2014, Nature Reviews Genetics.

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

[109]  Benjamin J. Raphael,et al.  Mutational landscape and significance across 12 major cancer types , 2013, Nature.

[110]  P. Srivastava,et al.  Immune response to mutant neo-antigens , 2013, Oncoimmunology.

[111]  Rashmi Kanagal-Shamanna,et al.  Clinical validation of a next-generation sequencing screen for mutational hotspots in 46 cancer-related genes. , 2013, The Journal of molecular diagnostics : JMD.

[112]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[113]  A. Lièvre,et al.  Role of microsatellite instability in the management of colorectal cancers. , 2013, Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver.

[114]  Steven A. Roberts,et al.  Mutational heterogeneity in cancer and the search for new cancer genes , 2014 .

[115]  Pia Kvistborg,et al.  The cancer antigenome , 2012, The EMBO journal.

[116]  Martin Kircher,et al.  High‐throughput DNA sequencing – concepts and limitations , 2010, BioEssays : news and reviews in molecular, cellular and developmental biology.

[117]  M. Robson,et al.  American Society of Clinical Oncology Policy Statement Update: Genetic and Genomic Testing for Cancer Susceptibility. , 2010, Journal of Clinical Oncology.

[118]  Emily H Turner,et al.  Targeted Capture and Massively Parallel Sequencing of Twelve Human Exomes , 2009, Nature.

[119]  W. Grizzle,et al.  Special symposium: fixation and tissue processing models , 2009, Biotechnic & histochemistry : official publication of the Biological Stain Commission.

[120]  S. Jewell,et al.  Copyright © American Society for Investigative Pathology Review Effect of Fixatives and Tissue Processing on the Content and Integrity of Nucleic Acids , 2022 .

[121]  E. Gilboa The makings of a tumor rejection antigen. , 1999, Immunity.

[122]  Alexis B. Carter,et al.  Standards and Guidelines for Validating Next-Generation Sequencing Bioinformatics Pipelines: A Joint Recommendation of the Association for Molecular Pathology and the College of American Pathologists. , 2018, The Journal of molecular diagnostics : JMD.

[123]  G. Hampton,et al.  OA20.01 Tumor Mutation Burden (TMB) is Associated with Improved Efficacy of Atezolizumab in 1L and 2L+ NSCLC Patients , 2017 .

[124]  J. Crown,et al.  PUB053 Panel Based Hybrid Capture Sequencing Assay to Correlate Mutational Load with Response to Immunotherapy , 2017 .

[125]  Samuel P. Strom Current practices and guidelines for clinical next-generation sequencing oncology testing@@@Current practices and guidelines for clinical next-generation sequencing oncology testing , 2016 .

[126]  K. Offit,et al.  American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[127]  Sofamor Danek,et al.  SUMMARY OF SAFETY AND EFFECTIVENESS DATA (SSED) , 2004 .