1
|
Clements A, Enserro D, Strickland KC, Previs R, Matei D, Mutch D, Powell M, Klopp A, Miller DS, Small W, DiSilvestro P, Spirtos N, Cosgrove C, Sfakianos G, Liu JR, Vargas R, Shahin M, Corr B, Dessources K, Ueland F, Warshal D, Gillen J, Secord AA. Molecular classification of endometrial cancers (EC) and association with relapse-free survival (RFS) and overall survival (OS) outcomes: Ancillary analysis of GOG-0258. Gynecol Oncol 2025; 193:119-129. [PMID: 39854806 PMCID: PMC11929956 DOI: 10.1016/j.ygyno.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE Determine if molecular classification using mismatch repair (MMR) and p53 protein expression predicts recurrence-free survival (RFS) and overall survival (OS) in endometrial cancer (EC) patients treated with chemotherapy and radiation (CRT) versus chemotherapy (CT). METHODS GOG-0258, a phase III randomized trial (NCT00942357), compared CRT to CT. Immunohistochemistry assessed MMR and p53 status. Kaplan-Meier curves and adjusted Cox models analyzed survival outcomes by molecular subtype. RESULTS ECs classified as deficient MMR (dMMR) (27 %), p53 abnormal (p53abn) (24 %), and p53 wild type (p53wt) (49 %). p53abn were more frequent in patients that were older, Black, and had serous histology (p < 0.001). Median follow up was 113 months. Five-year RFS and OS were worse with p53abn (29 % [Hazard Ratio (HR) = 3.39 (95 % Confidence Interval (CI): 2.34-4.91)] and 39 % [HR = 4.64 (95 % CI: 3.16-6.79)] compared to those with p53wt (referent) (p < 0.001). The five-year RFS and OS for dMMR cancers were (58 % [HR = 1.30 (95 % CI: 0.85-1.97)] and 77 % [HR = 1.53 (95 % CI: 0.99-2.36)] compared to those with p53wt (69 % and 85 %). After adjusting for age, gross residual disease, and treatment, p53wt improved RFS with CRT compared to CT in an exploratory analysis (77 % vs 60 %; HR = 0.54 (95 % CI: 0.32-0.94). The 5-year and 10-year OS rates were similar in CRT compared to CT in all subgroups. CONCLUSION Molecular classification appears to be predictive and prognostic, with worse survival in those with p53abn tumors. In an exploratory analysis, p53wt appears to predict improved RFS, favoring CRT over CT. There was no difference in treatment efficacy based on molecular subtype for OS.
Collapse
Affiliation(s)
| | - Danielle Enserro
- Roswell Park, Clinical Trials Development, Buffalo, NY, United States of America.
| | | | - Rebecca Previs
- Labcorp, Medical Affairs, Chapel Hill, NC, United States of America.
| | - Daniela Matei
- Feinberg School of Medicine, Northwestern, Chicago, IL, United States of America.
| | - David Mutch
- Washington University in St Louis, St Louis, MO, United States of America.
| | - Matthew Powell
- Washington University in St Louis, St Louis, MO, United States of America.
| | - Ann Klopp
- MD Anderson Cancer Center, Houston, TX, United States of America.
| | - David Scott Miller
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America.
| | - William Small
- Loyola Medicine, Maywood, IL, United States of America.
| | | | - Nick Spirtos
- Women's Cancer Center of Nevada, Las Vegas, NV, United States of America.
| | - Casey Cosgrove
- The Ohio State University, Columbus, OH, United States of America.
| | - Greg Sfakianos
- Piedmont Columbus Midtown JBACC Medical Oncology, Columbus, GA, United States of America.
| | - J Rebecca Liu
- Cancer Research Consortium of Michigan, Ann Arbor, MI, United States of America.
| | - Roberto Vargas
- The Cleveland Clinic, Cleveland, OH, United States of America.
| | - Mark Shahin
- Hanjani Institute for Gynecologic Oncology Abington Memorial Hospital, Abington, PA, United States of America.
| | - Bradley Corr
- University of Colorado School of Medicine, Aurora, CO, United States of America.
| | - Kimberly Dessources
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America.
| | - Frederick Ueland
- University of Kentucky Markey Cancer Center, Lexington, KY, United States of America.
| | - David Warshal
- Cooper Hospital University Medical Center, Camden, NJ, United States of America.
| | - Jessica Gillen
- Cancer Research for the Ozarks, Springfield, MO, United States of America.
| | | |
Collapse
|
2
|
Chowdhury D, Das A, Mishra M, Khutere T, Bodakhe SH. Physiological markers for immunotherapeutics: a review. J Chemother 2024:1-24. [PMID: 39711144 DOI: 10.1080/1120009x.2024.2443701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Immunotherapy has been advanced through multiple approaches, including immunogenic cytokines, monoclonal antibodies, therapeutic vaccinations, adoptive cell transfer, stem cell transplantation, and oncolytic viruses. This review analyses various strategies in genomics, transcriptomics, single-cell techniques, computational analysis, big data, and imaging technologies for the identification of tumour microbiota and microenvironments. Immunotherapy is becoming acknowledged as a feasible cancer treatment method, facilitating innovative cancer medicines and personalized medicine techniques.
Collapse
Affiliation(s)
- Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Mrityunjay Mishra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Trinkal Khutere
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
3
|
Mordaunt DA, Stark Z, Santos Gonzalez F, Dalziel K, Goranitis I. Development of a microcosting protocol to determine the economic cost of diagnostic genomic testing for rare diseases in Australia. BMJ Open 2023; 13:e069441. [PMID: 38030253 PMCID: PMC10689401 DOI: 10.1136/bmjopen-2022-069441] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
INTRODUCTION Genomic testing is a relatively new, disruptive and complex health technology with multiple clinical applications in rare diseases, cancer and infection control. Genomic testing is increasingly being implemented into clinical practice, following regulatory approval, funding and adoption in models of care, particularly in the area of rare disease diagnosis. A significant barrier to the adoption and implementation of genomic testing is funding. What remains unclear is what the cost of genomic testing is, what the underlying drivers of cost are and whether policy differences contribute to cost variance in different jurisdictions, such as the requirement to have staff with a medical license involved in testing. This costing study will be useful in future economic evaluations and health technology assessments to inform optimal levels of reimbursement and to support comprehensive and comparable assessment of healthcare resource utilisation in the delivery of genomic testing globally. METHODS A framework is presented that focuses on uncovering the process of genomic testing for any given laboratory, evaluating its utilisation and unit costs, and modelling the cost drivers and overall expenses associated with delivering genomic testing. The goal is to aid in refining and implementing policies regarding both the regulation and funding of genomic testing. A process-focused (activity-based) methodology is outlined, which encompasses resources, assesses individual cost components through a combination of bottom-up and top-down microcosting techniques and allows disaggregation of resource type and process step. ETHICS AND DISSEMINATION The outputs of the study will be reported at relevant regional genetics and health economics conferences, as well as submitted to a peer-reviewed journal focusing on genomics. Human research ethics committee approval is not required for this microcosting study. This study does not involve research on human subjects, and all data used in the analysis are either publicly available.
Collapse
Affiliation(s)
- Dylan A Mordaunt
- Health Economics Unit, Centre for Health Policy, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Women's and Children's Division, Southern Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Zornitza Stark
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Australian Genomics Health Alliance, Australian Genomics Health Alliance, Australia, UK
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Francisco Santos Gonzalez
- Health Economics Unit, Centre for Health Policy, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kim Dalziel
- Health Economics Unit, Centre for Health Policy, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ilias Goranitis
- Health Economics Unit, Centre for Health Policy, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Saldivar JS, Harris J, Ayash E, Hong M, Tandon P, Sinha S, Hebron PM, Houghton EE, Thorne K, Goodman LJ, Li C, Marfatia TR, Anderson J, Morra M, Lyle J, Bartha G, Chen R. Analytic validation of NeXT Dx™, a comprehensive genomic profiling assay. Oncotarget 2023; 14:789-806. [PMID: 37646774 PMCID: PMC10467627 DOI: 10.18632/oncotarget.28490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023] Open
Abstract
We describe the analytic validation of NeXT Dx, a comprehensive genomic profiling assay to aid therapy and clinical trial selection for patients diagnosed with solid tumor cancers. Proprietary methods were utilized to perform whole exome and whole transcriptome sequencing for detection of single nucleotide variants (SNVs), insertions/deletions (indels), copy number alterations (CNAs), and gene fusions, and determination of tumor mutation burden and microsatellite instability. Variant calling is enhanced by sequencing a patient-specific normal sample from, for example, a blood specimen. This provides highly accurate somatic variant calls as well as the incidental reporting of pathogenic and likely pathogenic germline alterations. Fusion detection via RNA sequencing provides more extensive and accurate fusion calling compared to DNA-based tests. NeXT Dx features the proprietary Accuracy and Content Enhanced technology, developed to optimize sequencing and provide more uniform coverage across the exome. The exome was validated at a median sequencing depth of >500x. While variants from 401 cancer-associated genes are currently reported from the assay, the exome/transcriptome assay is broadly validated to enable reporting of additional variants as they become clinically relevant. NeXT Dx demonstrated analytic sensitivities as follows: SNVs (99.4%), indels (98.2%), CNAs (98.0%), and fusions (95.8%). The overall analytic specificity was >99.0%.
Collapse
Affiliation(s)
| | - Jason Harris
- Personalis, Inc., Fremont, CA 94555, USA
- These authors contributed equally to this work
| | - Erin Ayash
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | | | | | | | | | - Conan Li
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | - John Lyle
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | |
Collapse
|
5
|
Tripathi R, Larson K, Fowler G, Han D, Vetto JT, Bordeaux JS, Yu WY. A Clinical Decision Tool to Calculate Pretest Probability of Sentinel Lymph Node Metastasis in Primary Cutaneous Melanoma. Ann Surg Oncol 2023; 30:4321-4328. [PMID: 36840860 PMCID: PMC9961302 DOI: 10.1245/s10434-023-13220-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/24/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND Although sentinel lymph node biopsy (SLNB) status is a strong prognostic indicator for cutaneous melanoma, unnecessary SLNBs have substantial cost and morbidity burden. OBJECTIVE This study was designed to develop, validate, and present a personalized, clinical, decision-making tool using nationally representative data with clinically actionable probability thresholds (Expected Lymphatic Metastasis Outcome [ELMO]). METHODS Data from the Surveillance, Epidemiology, and End Results (SEER) Registry from 2000 to 2017 and the National Cancer Database (NCDB) from 2004 to 2015 were used to develop and internally validate a logistic ridge regression predictive model for SLNB positivity. External validation was done with 1568 patients at a large tertiary referral center. RESULTS The development cohort included 134,809 patients, and the internal validation cohort included 38,518 patients. ELMO (AUC 0.85) resulted in a 29.54% SLNB reduction rate and greater sensitivity in predicting SLNB status for T1b, T2a, and T2b tumors than previous models. In external validation, ELMO had an accuracy of 0.7586 and AUC of 0.7218. Limitations of this study are potential miscoding, unaccounted confounders, and effect modification. CONCLUSIONS ELMO ( https://melanoma-sentinel.herokuapp.com/ ) has been developed and validated (internally and externally) by using the largest publicly available dataset of melanoma patients and was found to have high accuracy compared with other published models and gene expression tests. Individualized risk estimates for SLNB positivity are critical in facilitating thorough decision-making for healthcare providers and patients with melanoma.
Collapse
Affiliation(s)
- Raghav Tripathi
- Department of Dermatology, Johns Hopkins Medicine, Baltimore, MD, USA.
| | | | - Graham Fowler
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Dale Han
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - John T Vetto
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Jeremy S Bordeaux
- Department of Dermatology, University Hospitals Cleveland Medical Center/Case Western Reserve University, Cleveland, OH, USA
| | - Wesley Y Yu
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
6
|
Tripathi R, Larson K, Fowler G, Han D, Vetto JT, Bordeaux JS, Yu WY. ASO Author Reflections: Using Big Data to Bring Precision Medicine to Melanoma Management. Ann Surg Oncol 2023; 30:4329-4330. [PMID: 36820933 DOI: 10.1245/s10434-023-13268-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023]
Affiliation(s)
- Raghav Tripathi
- Department of Dermatology, Johns Hopkins Medicine, Baltimore, MD, USA.
| | | | - Graham Fowler
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Dale Han
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - John T Vetto
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Jeremy S Bordeaux
- Department of Dermatology, Case Comprehensive Cancer Center, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Wesley Y Yu
- Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
7
|
Hernandez-Pacheco N, Melén E. Unraveling the genetic architecture of asthma. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1257. [PMID: 36618807 PMCID: PMC9816849 DOI: 10.21037/atm-22-5375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022]
Affiliation(s)
- Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden;,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden;,Sachs’ Children’s Hospital, South General Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Bosserman LD, Mambetsariev I, Ladbury C, Barzi A, Johnson D, Morse D, Deaville D, Smith W, Rajurkar S, Merla A, Hajjar G, Kim D, Fricke J, Trisal V, Salgia R. Pyramidal Decision Support Framework Leverages Subspecialty Expertise across Enterprise to Achieve Superior Cancer Outcomes and Personalized, Precision Care Plans. J Clin Med 2022; 11:jcm11226738. [PMID: 36431215 PMCID: PMC9697355 DOI: 10.3390/jcm11226738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
The complexity of cancer care requires integrated and continuous support to deliver appropriate care. An expert network with complementary expertise and the capability of multidisciplinary care is an integral part of contemporary oncology care. Appropriate infrastructure is necessary to empower this network to deliver personalized precision care to their patients. Providing decision support as cancer care becomes exponentially more complex with new diagnostic and therapeutic choices remains challenging. City of Hope has developed a Pyramidal Decision Support Framework to address these challenges, which were exacerbated by the COVID pandemic, health plan restrictions, and growing geographic site diversity. Optimizing efficient and targeted decision support backed by multidisciplinary cancer expertise can improve individual patient treatment plans to achieve improved care and survival wherever patients are treated.
Collapse
Affiliation(s)
- Linda D. Bosserman
- Department of Medical Oncology and Therapeutics Research, City of Hope, Irwindale, CA 91706, USA
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Colton Ladbury
- Department of Radiation Oncology, City of Hope, Duarte, CA 91010, USA
| | - Afsaneh Barzi
- Department of Medical Oncology and Therapeutics Research, City of Hope, Irwindale, CA 91706, USA
| | - Deron Johnson
- Department of Clinical Informatics, City of Hope, Duarte, CA 91010, USA
| | - Denise Morse
- Department of Quality, Risk and Regulatory Management, City of Hope, Duarte, CA 91010, USA
| | - Debbie Deaville
- Department of Enterprise Business Intelligence, City of Hope, Irwindale, CA 91706, USA
| | - Wade Smith
- Department of Medical Oncology and Therapeutics Research, City of Hope, Newport Beach, CA 92660, USA
| | - Swapnil Rajurkar
- Department of Medical Oncology and Therapeutics Research, City of Hope, Upland, CA 91784, USA
| | - Amartej Merla
- Department of Medical Oncology and Therapeutics Research, City of Hope, Antelope Valley, CA 93534, USA
| | - George Hajjar
- Department of Medical Oncology and Therapeutics Research, City of Hope, Mission Hills, CA 91345, USA
| | - Daniel Kim
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Vijay Trisal
- Department of Medicine, City of Hope, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
- Correspondence:
| |
Collapse
|
9
|
Bafiti V, Katsila T. Pharmacometabolomics-Based Translational Biomarkers: How to Navigate the Data Ocean. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:542-551. [PMID: 36149303 DOI: 10.1089/omi.2022.0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Metabolome is the end point of the genome-environment interplay, and enables an important holistic overview of individual adaptability and host responses to environmental, ecological, as well as endogenous changes such as disease. Pharmacometabolomics is the application of metabolome knowledge to decipher the mechanisms of interindividual and intraindividual variations in drug efficacy and safety. Pharmacometabolomics also contributes to prediction of drug treatment outcomes on the basis of baseline (predose) and postdose metabotypes through mathematical modeling. Thus, pharmacometabolomics is a strong asset for a diverse community of stakeholders interested in theory and practice of evidence-based and precision/personalized medicine: academic researchers, public health scholars, health professionals, pharmaceutical, diagnostics, and biotechnology industries, among others. In this expert review, we discuss pharmacometabolomics in four contexts: (1) an interdisciplinary omics tool and field to map the mechanisms and scale of interindividual variability in drug effects, (2) discovery and development of translational biomarkers, (3) advance digital biomarkers, and (4) empower drug repurposing, a field that is increasingly proving useful in the current era of Covid-19. As the applications of pharmacometabolomics are growing rapidly in the current postgenome era, next-generation proteomics and metabolomics follow the example of next-generation sequencing analyses. Pharmacometabolomics can also empower data reliability and reproducibility through multiomics integration strategies, which use each data layer to correct, connect with, and inform each other. Finally, we underscore here that contextual data remain crucial for precision medicine and drug development that stand the test of time and clinical relevance.
Collapse
Affiliation(s)
- Vivi Bafiti
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| |
Collapse
|
10
|
Loddo M, Hardisty KM, Llewelyn A, Haddow T, Thatcher R, Williams G. Utilisation of semiconductor sequencing for detection of actionable fusions in solid tumours. PLoS One 2022; 17:e0246778. [PMID: 35984852 PMCID: PMC9390944 DOI: 10.1371/journal.pone.0246778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 07/22/2022] [Indexed: 11/19/2022] Open
Abstract
Oncogenic fusions represent compelling druggable targets in solid tumours highlighted by the recent site agnostic FDA approval of larotrectinib for NTRK rearrangements. However screening for fusions in routinely processed tissue samples is constrained due to degradation of nucleic acid as a result of formalin fixation., To investigate the clinical utility of semiconductor sequencing optimised for detection of actionable fusion transcripts in formalin fixed samples, we have undertaken an analysis of test trending data generated by a clinically validated next generation sequencing platform designed to capture 867 of the most clinically relevant druggable driver-partner oncogenic fusions. Here we show across a real-life cohort of 1112 patients with solid tumours that actionable fusions occur at high frequency (7.4%) with linkage to a wide range of targeted therapy protocols including seven fusion-drug matches with FDA/EMA approval and/or NCCN/ESMO recommendations and 80 clinical trials. The more prevalent actionable fusions identified were independent of tumour type in keeping with signalling via evolutionary conserved RAS/RAF/MEK/ERK, PI3K/AKT/MTOR, PLCy/PKC and JAK/STAT pathways. Taken together our data indicates that semiconductor sequencing for detection of actionable fusions can be integrated into routine diagnostic pathology workflows enabling the identification of personalised treatment options that have potential to improve clinical cancer management across many tumour types.
Collapse
Affiliation(s)
- Marco Loddo
- Oncologica UK Ltd, Cambridge, United Kingdom
- * E-mail: (ML); (GW)
| | | | | | | | | | - Gareth Williams
- Oncologica UK Ltd, Cambridge, United Kingdom
- * E-mail: (ML); (GW)
| |
Collapse
|
11
|
Shukla N, Levine MF, Gundem G, Domenico D, Spitzer B, Bouvier N, Arango-Ossa JE, Glodzik D, Medina-Martínez JS, Bhanot U, Gutiérrez-Abril J, Zhou Y, Fiala E, Stockfisch E, Li S, Rodriguez-Sanchez MI, O'Donohue T, Cobbs C, Roehrl MHA, Benhamida J, Iglesias Cardenas F, Ortiz M, Kinnaman M, Roberts S, Ladanyi M, Modak S, Farouk-Sait S, Slotkin E, Karajannis MA, Dela Cruz F, Glade Bender J, Zehir A, Viale A, Walsh MF, Kung AL, Papaemmanuil E. Feasibility of whole genome and transcriptome profiling in pediatric and young adult cancers. Nat Commun 2022; 13:2485. [PMID: 35585047 PMCID: PMC9117241 DOI: 10.1038/s41467-022-30233-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 04/21/2022] [Indexed: 02/07/2023] Open
Abstract
The utility of cancer whole genome and transcriptome sequencing (cWGTS) in oncology is increasingly recognized. However, implementation of cWGTS is challenged by the need to deliver results within clinically relevant timeframes, concerns about assay sensitivity, reporting and prioritization of findings. In a prospective research study we develop a workflow that reports comprehensive cWGTS results in 9 days. Comparison of cWGTS to diagnostic panel assays demonstrates the potential of cWGTS to capture all clinically reported mutations with comparable sensitivity in a single workflow. Benchmarking identifies a minimum of 80× as optimal depth for clinical WGS sequencing. Integration of germline, somatic DNA and RNA-seq data enable data-driven variant prioritization and reporting, with oncogenic findings reported in 54% more patients than standard of care. These results establish key technical considerations for the implementation of cWGTS as an integrated test in clinical oncology. Cancer whole-genome and transcriptome sequencing (cWGTS) has been challenging to implement in clinical settings. Here, the authors develop a workflow to deliver robust cWGTS analyses and reports within clinically-relevant timeframes for paediatric, adolescent and young adult solid tumour patients.
Collapse
Affiliation(s)
- N Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M F Levine
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - G Gundem
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - D Domenico
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - B Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - N Bouvier
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J E Arango-Ossa
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - D Glodzik
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J S Medina-Martínez
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - U Bhanot
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Precision Pathology Biobanking Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J Gutiérrez-Abril
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Y Zhou
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Fiala
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Stockfisch
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Li
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - T O'Donohue
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - C Cobbs
- Integrated Genomics Operation Core, Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M H A Roehrl
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Precision Pathology Biobanking Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J Benhamida
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - F Iglesias Cardenas
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Kinnaman
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - S Farouk-Sait
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Slotkin
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M A Karajannis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - F Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - J Glade Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Viale
- Integrated Genomics Operation Core, Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - M F Walsh
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A L Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - E Papaemmanuil
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
12
|
Orellana TJ, Kim H, Beriwal S, Bhargava R, Berger J, Buckanovich RJ, Coffman LG, Courtney-Brooks M, Mahdi H, Olawaiye AB, Sukumvanich P, Taylor SE, Smith KJ, Lesnock JL. Cost-effectiveness analysis of tumor molecular classification in high-risk early-stage endometrial cancer. Gynecol Oncol 2021; 164:129-135. [PMID: 34740462 DOI: 10.1016/j.ygyno.2021.10.071] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/04/2021] [Accepted: 10/10/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Tumor molecular analyses in endometrial cancer (EC) includes 4 distinct subtypes: (1) POLE-mutated, (2) mismatch repair protein (MMR) deficient, (3) p53 mutant, and (4) no specific molecular profile. Recently, a sub-analysis of PORTEC-3 demonstrated notable differences in treatment response between molecular classification (MC) groups. Cost of testing is one barrier to widespread adoption of MC. Therefore, we sought to determine the cost-effectiveness of MC in patients with stage I and II high-risk EC. METHODS A Markov decision model was developed to compare tumor molecular classification (TMC) vs. no testing (NT). A healthcare payor's perspective and 5-year time horizon were used. Base case data were abstracted from PORTEC-3 and the molecular sub-analysis. Cost and utility data were derived from public databases, peer-reviewed literature, and expert input. Strategies were compared using the incremental cost-effectiveness ratio (ICER) with effectiveness in quality-adjusted life years (QALYs) and evaluated with a willingness-to-pay threshold of $100,000 per QALY gained. Sensitivity analyses were performed to test model robustness. RESULTS When compared to NT, TMC was cost effective with an ICER of $25,578 per QALY gained; incremental cost was $1780 and incremental effectiveness was 0.070 QALYs. In one-way sensitivity analyses, results were most sensitive to the cost of POLE testing, but TMC remained cost-effective over all parameter ranges. CONCLUSIONS TMC in early-stage high-risk EC is cost-effective, and the model results were robust over a range of parameters. Given that MC can be used to guide adjuvant treatment decisions, these findings support adoption of TMC into routine practice.
Collapse
Affiliation(s)
- T J Orellana
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States.
| | - H Kim
- Department of Radiation Oncology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA 15232, United States
| | - S Beriwal
- Department of Radiation Oncology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA 15232, United States
| | - R Bhargava
- Department of Pathology, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Pittsburgh, PA 15213, United States
| | - J Berger
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| | - R J Buckanovich
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States; Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, United States
| | - L G Coffman
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States; Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, United States
| | - M Courtney-Brooks
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| | - H Mahdi
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| | - A B Olawaiye
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| | - P Sukumvanich
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| | - S E Taylor
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| | - K J Smith
- Center for Research on Health Care, Department of Medicine, University of Pittsburgh School of Medicine, 200 Meyran Ave., Suite 200, Pittsburgh, PA 15213, United States
| | - J L Lesnock
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens' Hospital of the University of Pittsburgh Medical Center, 300 Halket Street, Suite 1750, Pittsburgh, PA 15213, United States
| |
Collapse
|
13
|
LeVee A, Lin CY, Posadas E, Figlin R, Bhowmick NA, Di Vizio D, Ellis L, Rosser CJ, Freeman MR, Theodorescu D, Freedland SJ, Gong J. Clinical Utility of Olaparib in the Treatment of Metastatic Castration-Resistant Prostate Cancer: A Review of Current Evidence and Patient Selection. Onco Targets Ther 2021; 14:4819-4832. [PMID: 34552338 PMCID: PMC8450162 DOI: 10.2147/ott.s315170] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/03/2021] [Indexed: 11/23/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is an aggressive and fatal disease with a median survival of 36 months. With the advent of genetic sequencing to identify individual genomic profiles and acquired tumor-specific pathways, targeted therapies have revolutionized cancer treatment, including the treatment strategy in mCRPC. Poly(adenosine 5'-diphosphate) ribose polymerase inhibitors (PARPi) are oral drugs that target mutations in the homologous recombination repair (HRR) pathway, which are found in approximately 27% of prostate cancer patients. In May 2020, the first PARP inhibitor, olaparib, was approved by the US Food and Drug Administration for men with mCRPC with HHR gene mutations based on the findings of the Phase III PROfound trial that showed improved overall survival in men with mCRPC who received olaparib and whose disease had progressed on a novel hormonal agent. This review summarizes the current evidence and clinical utility of olaparib as treatment in men with mCRPC. We describe the mechanism of action of PARPi, key clinical trials of olaparib in men with mCRPC, and ongoing Phase II and III clinical trials investigating olaparib in combination therapy and as front-line therapy in mCRPC.
Collapse
Affiliation(s)
- Alexis LeVee
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ching Ying Lin
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Edwin Posadas
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert Figlin
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Neil A Bhowmick
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and Therapeutics, Biomedical Sciences, and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Leigh Ellis
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Charlos J Rosser
- Department of Surgery, Division of Urology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael R Freeman
- Department of Surgery, Division of Cancer Biology and Therapeutics, Biomedical Sciences, and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Department of Surgery, Division of Cancer Biology and Therapeutics, Biomedical Sciences, and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Freedland
- Department of Surgery, Division of Urology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Section of Urology, Durham VA Medical Center, Durham, NC, USA
| | - Jun Gong
- Department of Medicine, Division of Hematology and Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
14
|
van de Ven M, Simons MJHG, Koffijberg H, Joore MA, IJzerman MJ, Retèl VP, van Harten WH. Whole genome sequencing in oncology: using scenario drafting to explore future developments. BMC Cancer 2021; 21:488. [PMID: 33933021 PMCID: PMC8088550 DOI: 10.1186/s12885-021-08214-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background In oncology, Whole Genome Sequencing (WGS) is not yet widely implemented due to uncertainties such as the required infrastructure and expertise, costs and reimbursements, and unknown pan-cancer clinical utility. Therefore, this study aimed to investigate possible future developments facilitating or impeding the use of WGS as a molecular diagnostic in oncology through scenario drafting. Methods A four-step process was adopted for scenario drafting. First, the literature was searched for barriers and facilitators related to the implementation of WGS. Second, they were prioritized by international experts, and third, combined into coherent scenarios. Fourth, the scenarios were implemented in an online survey and their likelihood of taking place within 5 years was elicited from another group of experts. Based on the minimum, maximum, and most likely (mode) parameters, individual Program Evaluation and Review Technique (PERT) probability density functions were determined. Subsequently, individual opinions were aggregated by performing unweighted linear pooling, from which summary statistics were extracted and reported. Results Sixty-two unique barriers and facilitators were extracted from 70 articles. Price, clinical utility, and turnaround time of WGS were ranked as the most important aspects. Nine scenarios were developed and scored on likelihood by 18 experts. The scenario about introducing WGS as a clinical diagnostic with a lower price, shorter turnaround time, and improved degree of actionability, scored the highest likelihood (median: 68.3%). Scenarios with low likelihoods and strong consensus were about better treatment responses to more actionable targets (26.1%), and the effect of centralizing WGS (24.1%). Conclusions Based on current expert opinions, the implementation of WGS as a clinical diagnostic in oncology is heavily dependent on the price, clinical utility (both in terms of identifying actionable targets as in adding sufficient value in subsequent treatment), and turnaround time. These aspects and the optimal way of service provision are the main drivers for the implementation of WGS and should be focused on in further research. More knowledge regarding these factors is needed to inform strategic decision making regarding the implementation of WGS, which warrants support from all relevant stakeholders. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08214-8.
Collapse
Affiliation(s)
- Michiel van de Ven
- Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Martijn J H G Simons
- Maastricht University Medical Center, Maastricht, The Netherlands.,Maastricht University, Care and Public Health Research Institute (CAPHRI), Maastricht, The Netherlands
| | - Hendrik Koffijberg
- Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Manuela A Joore
- Maastricht University Medical Center, Maastricht, The Netherlands.,Maastricht University, Care and Public Health Research Institute (CAPHRI), Maastricht, The Netherlands
| | - Maarten J IJzerman
- Technical Medical Centre, University of Twente, Enschede, The Netherlands.,University of Melbourne Centre for Cancer Research, University of Melbourne, Melbourne, Australia.,Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Valesca P Retèl
- Technical Medical Centre, University of Twente, Enschede, The Netherlands. .,Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands.
| | - Wim H van Harten
- Technical Medical Centre, University of Twente, Enschede, The Netherlands.,Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands.,Rijnstate General Hospital, Arnhem, The Netherlands
| |
Collapse
|
15
|
White T, Szelinger S, LoBello J, King A, Aldrich J, Garinger N, Halbert M, Richholt RF, Mastrian SD, Babb C, Ozols AA, Goodman LJ, Basu GD, Royce T. Analytic validation and clinical utilization of the comprehensive genomic profiling test, GEM ExTra ®. Oncotarget 2021; 12:726-739. [PMID: 33889297 PMCID: PMC8057276 DOI: 10.18632/oncotarget.27945] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 03/28/2021] [Indexed: 12/20/2022] Open
Abstract
We developed and analytically validated a comprehensive genomic profiling (CGP) assay, GEM ExTra, for patients with advanced solid tumors that uses Next Generation Sequencing (NGS) to characterize whole exomes employing a paired tumor-normal subtraction methodology. The assay detects single nucleotide variants (SNV), indels, focal copy number alterations (CNA), TERT promoter region, as well as tumor mutation burden (TMB) and microsatellite instability (MSI) status. Additionally, the assay incorporates whole transcriptome sequencing of the tumor sample that allows for the detection of gene fusions and select special transcripts, including AR-V7, EGFR vIII, EGFRvIV, and MET exon 14 skipping events. The assay has a mean target coverage of 180X for the normal (germline) and 400X for tumor DNA including enhanced probe design to facilitate the sequencing of difficult regions. Proprietary bioinformatics, paired with comprehensive clinical curation results in reporting that defines clinically actionable, FDA-approved, and clinical trial drug options for the management of the patient's cancer. GEM ExTra demonstrated analytic specificity (PPV) of > 99.9% and analytic sensitivity of 98.8%. Application of GEM ExTra to 1,435 patient samples revealed clinically actionable alterations in 83.9% of reports, including 31 (2.5%) where therapeutic recommendations were based on RNA fusion findings only.
Collapse
Affiliation(s)
- Tracey White
- Ashion Analytics, LLC, Phoenix, Arizona 85004, USA
- These authors contributed equally to this work
| | - Szabolcs Szelinger
- Ashion Analytics, LLC, Phoenix, Arizona 85004, USA
- These authors contributed equally to this work
| | | | - Amy King
- Ashion Analytics, LLC, Phoenix, Arizona 85004, USA
| | | | | | | | | | | | - Cody Babb
- Ashion Analytics, LLC, Phoenix, Arizona 85004, USA
| | | | | | | | - Thomas Royce
- Ashion Analytics, LLC, Phoenix, Arizona 85004, USA
| |
Collapse
|
16
|
Su D, Wu B, Shi L. Cost-Effectiveness of Genomic Test-Directed Olaparib for Metastatic Castration-Resistant Prostate Cancer. Front Pharmacol 2021; 11:610601. [PMID: 33574757 PMCID: PMC7870786 DOI: 10.3389/fphar.2020.610601] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose: The effectiveness of poly (adenosine diphosphate–ribose) polymerase (PARP) inhibitor olaparib for metastatic castration-resistant prostate cancer (MCRPC) with multiple loss-of-function alterations in genes that are involved in DNA repair has been demonstrated. We aimed to evaluate the cost-effectiveness of genomic test-directed olaparib on MCRPC from the US payer perspective. Methods: A partitioned survival model was adopted to project the disease course of MCRPC had at least one gene alteration in BRCA1, BRCA2 and ATM (Scenario A) and has alterations in any of all 15 prespecified genes (Scenario B) after next-generation sequencing test. The efficacy and toxicity data were gathered from the PROfound trial. Clinical probabilities related to survival were estimated from the reported survival probabilities in each PROfound group. Cost and health preference data were derived from the literature. The incremental cost-effectiveness ratio (ICER) was measured. Subgroup analysis and sensitivity analysis were performed for exploring the model uncertainties. Results: Olaparib yielded an additional 0.063 and 0.068 of quality-adjusted life year (QALY) with the augmented cost of $7,382 and saved the cost of $ 1,980 compared to standard care in scenario A and B, respectively, which yielded an ICER of $116,903/QALY and a cost-saving option. The lower weekly cost related to olaparib treatment led to the dominant findings in scenario B. The varied results between scenario A and B could be partly explained by different the number need to screen for identifying eligible patients who could be administered with olaparib, which sharply augmented the costs of the olaparib arm in scenario A. Subgroup analysis and sensitivity analysis revealed the results were generally robust in both of two scenarios. Conclusion: The genomic test-directed olaparib is a preferred option compared with standard care strategy for men with MCRPC who had any of all 15 prespecified genes.
Collapse
Affiliation(s)
- Dan Su
- Department of Pharmacy, the First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Bin Wu
- Medical Decision and Economic Group, Ren Ji Hospital, South Campus, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lizheng Shi
- Department of Global Health Management and Policy, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
17
|
Palmero R, Taus A, Viteri S, Majem M, Carcereny E, Garde-Noguera J, Felip E, Nadal E, Malfettone A, Sampayo M, Riva F, Nagy RJ, Lanman RB, Faull I, Dix D, Karachaliou N, Rosell R. Biomarker Discovery and Outcomes for Comprehensive Cell-Free Circulating Tumor DNA Versus Standard-of-Care Tissue Testing in Advanced Non–Small-Cell Lung Cancer. JCO Precis Oncol 2021; 5:93-102. [DOI: 10.1200/po.20.00241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Treatment guidelines for advanced non–small-cell lung cancer (aNSCLC) recommend broad molecular profiling for targeted therapy selection. This study prospectively assessed comprehensive next-generation sequencing (NGS) of cell-free circulating tumor DNA (cfDNA) compared with standard-of-care (SOC) tissue-based testing to identify guideline-recommended alterations in aNSCLC. PATIENTS AND METHODS Patients with treatment-naïve aNSCLC were tested using a well-validated NGS cfDNA panel, and results were compared with SOC tissue testing. The primary objective was noninferiority of cfDNA vs. tissue analysis for the detection of two guideline-recommended biomarkers ( EGFR and ALK) and an additional six actionable biomarkers. Secondary analyses included tissue versus cfDNA biomarker discovery, overall response rate (ORR), progression-free survival (PFS) to targeted therapy, and positive predictive value (PPV) of cfDNA. RESULTS The primary objective was met with cfDNA identifying actionable mutations in 46 patients versus 48 by tissue ( P < .05). In total, 0/186 patients were genotyped for all eight biomarkers with tissue, compared with 90.8% using cfDNA. Targetable alterations or KRAS were identified in 80.7% when cfDNA was used first versus 57.1% when tissue was used first. PPV for cfDNA-detected EGFR was 100.0% (25/25). ORR and PFS in patients receiving targeted therapy based on tissue or cfDNA were similar to those previously reported. Conclusion This prospective study confirms a previous report that comprehensive cfDNA testing is noninferior to SOC tissue testing in detecting aNSCLC-recommended biomarkers. Furthermore, cfDNA-based first-line therapy produced outcomes similar to tissue-based testing, demonstrating the clinical utility of comprehensive cfDNA genotyping as the initial genotyping modality in patients with treatment-naïve aNSCLC when tissue is insufficient or when all actionable biomarkers cannot be rapidly assessed.
Collapse
Affiliation(s)
| | - Alvaro Taus
- Hospital del Mar, Barcelona, Spain
- Universidad Autónoma de Barcelona (UAB), Barcelona, Spain
| | - Santiago Viteri
- Quirón Salud-Dexeus University Institute, IOR, Medical Oncology Department, Barcelona, Spain
| | | | - Enric Carcereny
- Institut Català d'Oncologia Badalona-Hospital Germans Trias i Pujol B-ARGO, Badalona, Spain
| | | | | | | | - Andrea Malfettone
- Medica Scientia Innovation Research—MEDSIR, Barcelona, Spain and Ridgewood, NJ
| | - Miguel Sampayo
- Medica Scientia Innovation Research—MEDSIR, Barcelona, Spain and Ridgewood, NJ
| | - François Riva
- Medica Scientia Innovation Research—MEDSIR, Barcelona, Spain and Ridgewood, NJ
| | | | | | - Iris Faull
- Guardant Health, South San Francisco, CA
| | - Daniel Dix
- Guardant Health, South San Francisco, CA
| | - Niki Karachaliou
- Catalan Institute of Oncology, Hospital Germans Trias I Pujol, Badalona, Spain
| | - Rafael Rosell
- Catalan Institute of Oncology, Hospital Germans Trias I Pujol, Badalona, Spain
| |
Collapse
|
18
|
Cai B, Zhou ZY, Xue W, Hazra NC, Singh M, Mishra D, Brixner D, Oderda G, Biskupiak J. Budget impact of capmatinib for adults with metastatic non-small cell lung cancer harboring a MET exon 14 skipping mutation in the United States. J Med Econ 2021; 24:131-139. [PMID: 33397178 DOI: 10.1080/13696998.2020.1867470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
AIMS To estimate the budget impact of adding capmatinib, the first FDA approved MET inhibitor, to a US commercial or Medicare health plan for patients with metastatic non-small cell lung cancer (mNSCLC) whose tumors have a mutation that leads to MET exon 14 (METex14) skipping. METHODS Target population size was estimated using published epidemiology data. Clinical data were obtained from the GEOMETRY mono-1 capmatinib trial and published trials. Treatments in the market mix included crizotinib, pembrolizumab, ramucirumab, and chemotherapy. Uptake of capmatinib and testing rates were based on market research. All costs (drug acquisition and administration, pre-progression, progression, terminal care, adverse event, and testing) were estimated based on public sources (2020 USD). RESULTS The number of patients eligible for capmatinib in the first three years was estimated to be 2-3 in a hypothetical 1 million member commercial plan and 34-44 in a hypothetical 1 million member Medicare plan each year. The estimated total budget impact ranged from $9,695 to $67,725 for a commercial plan and $141,350 to $985,695 for Medicare. With capmatinib included, a marginal per member per month budget impact was estimated (commercial: $0.0008 to $0.0056; Medicare: $0.0118 to $0.0821). Capmatinib inclusion resulted in lower medical costs (commercial: -$0.0003 to -$0.0007; Medicare: -$0.0037 to -$0.0106), partially offsetting increased drug costs ($0.0011 to $0.0064; $0.0154 to $0.0928, respectively), and were primarily driven by reductions in progression and terminal care costs (-$0.0003 to -$0.0009; -$0.0037 to -$0.0125, respectively). The results were most sensitive to capmatinib market share, capmatinib price, and treatment duration. LIMITATIONS Certain assumptions were applied to the model to account for inputs with limited evidence. CONCLUSIONS The estimated budget impact of including capmatinib for mNSCLC with a METex14 skipping mutation is minimal, and the increased drug costs were partially offset by savings in AEs, and progression-related and terminal care costs.
Collapse
Affiliation(s)
- Beilei Cai
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | | | | | | | | - Diana Brixner
- Department of Pharmacotherapy, University of Utah College of Pharmacy, Salt Lake City, UT, USA
| | - Gary Oderda
- Department of Pharmacotherapy, University of Utah College of Pharmacy, Salt Lake City, UT, USA
| | - Joseph Biskupiak
- Department of Pharmacotherapy, University of Utah College of Pharmacy, Salt Lake City, UT, USA
| |
Collapse
|
19
|
Seoighe C, Bracken AP, Buckley P, Doran P, Green R, Healy S, Kavanagh D, Kenny E, Lawler M, Lowery M, Morris D, Morrissey D, O'Byrne JJ, Shields D, Smith O, Steward CA, Sweeney B, Kolch W. The future of genomics in Ireland - focus on genomics for health. HRB Open Res 2020; 3:89. [PMID: 33855271 PMCID: PMC7993626 DOI: 10.12688/hrbopenres.13187.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
Genomics is revolutionizing biomedical research, medicine and healthcare globally in academic, public and industry sectors alike. Concrete examples around the world show that huge benefits for patients, society and economy can be accrued through effective and responsible genomic research and clinical applications. Unfortunately, Ireland has fallen behind and needs to act now in order to catch up. Here, we identify key issues that have resulted in Ireland lagging behind, describe how genomics can benefit Ireland and its people and outline the measures needed to make genomics work for Ireland and Irish patients. There is now an urgent need for a national genomics strategy that enables an effective, collaborative, responsible, well-regulated, and patient centred environment where genome research and clinical genomics can thrive. We present eight recommendations that could be the pillars of a national genomics health strategy.
Collapse
Affiliation(s)
- Cathal Seoighe
- National University of Ireland Galway, Galway, H91 TK33, Ireland
| | | | | | - Peter Doran
- University College Dublin, Dublin, 4, Ireland
- Mater Misericordiae University Hospital, Dublin, 7, Ireland
| | - Robert Green
- Brigham Health, Broad Institute, Ariadne Labs, Harvard Medical School, Boston, MA, 02115, USA
| | - Sandra Healy
- National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - David Kavanagh
- Genuity Science (Ireland) Ltd., Dublin, D18 K7W4, Ireland
| | - Elaine Kenny
- Trinity College Dublin, Dublin, 2, Ireland
- ELDA Biotech, Trinity Translational Medicine Institute, St James's Hospital, Dublin, D08 W9RT, Ireland
| | - Mark Lawler
- Queen's University Belfast, Belfast, Northern Ireland, BT7 1NN, Ireland
| | - Maeve Lowery
- Trinity College Dublin, Dublin, 2, Ireland
- Saint James' Hospital, Dublin, D08 NHY1, Ireland
| | - Derek Morris
- National University of Ireland Galway, Galway, H91 TK33, Ireland
| | - Darrin Morrissey
- National Institute for Bioprocessing Research and Training, Blackrock, A94 X099, Ireland
| | | | | | - Owen Smith
- University College Dublin, Dublin, 4, Ireland
- Children’s Health Ireland, Crumlin, Dublin, D12 N512, Ireland
| | | | | | - Walter Kolch
- National University of Ireland Galway, Galway, H91 TK33, Ireland
| |
Collapse
|
20
|
Salgia R, Mambetsariev I, Pharaon R, Fricke J, Baroz AR, Hozo I, Chen C, Koczywas M, Massarelli E, Reckamp K, Djulbegovic B. Evaluation of Omics-Based Strategies for the Management of Advanced Lung Cancer. JCO Oncol Pract 2020; 17:e257-e265. [PMID: 32639928 DOI: 10.1200/op.20.00117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Omic-informed therapy is being used more frequently for patients with non-small-cell lung cancer (NSCLC) being treated on the basis of evidence-based decision-making. However, there is a lack of a standardized framework to evaluate those decisions and understand the association between omics-based management strategies and survival among patients. Therefore, we compared outcomes between patients with lung adenocarcinoma who received omics-driven targeted therapy versus patients who received standard therapeutic options. PATIENTS AND METHODS This was a retrospective study of patients with advanced NSCLC adenocarcinoma (N = 798) at City of Hope who received genomic sequencing at the behest of their treating oncologists. A thoracic oncology registry was used as a clinicogenomic database to track patient outcomes. RESULTS Of 798 individuals with advanced NSCLC (median age, 65 years [range, 22-99 years]; 60% white; 50% with a history of smoking), 662 patients (83%) had molecular testing and 439 (55%) received targeted therapy on the basis of the omic-data. A fast-and-frugal decision tree (FFT) model was developed to evaluate the impact of omics-based strategy on decision-making, progression-free survival (PFS), and overall survival (OS). We calculated that the overall positive predictive value of the entire FFT strategy for predicting decisions regarding the use of tyrosine kinase inhibitor-based targeted therapy was 88% and the negative predictive value was 96%. In an adjusted Cox regression analysis, there was a significant correlation with survival benefit with the FFT omics-driven therapeutic strategy for both PFS (hazard ratio [HR], 0.56; 95% CI, 0.42 to 0.74; P < .001) and OS (HR, 0.51; 95% CI, 0.36 to 0.71; P < .001) as compared with standard therapeutic options. CONCLUSION Among patients with advanced NSCLC who received care in the academic oncology setting, omics-driven therapy decisions directly informed treatment in patients and was correlated with better OS and PFS.
Collapse
Affiliation(s)
- Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Rebecca Pharaon
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Angel Ray Baroz
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Iztok Hozo
- Department of Mathematics, Indiana University Northwest, Gary, IN
| | - Chen Chen
- Applied AI and Data Science, City of Hope, Duarte, CA
| | - Marianna Koczywas
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Erminia Massarelli
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Karen Reckamp
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA.,Division of Medical Oncology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Benjamin Djulbegovic
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| |
Collapse
|
21
|
Rajurkar S, Mambetsariev I, Pharaon R, Leach B, Tan T, Kulkarni P, Salgia R. Non-Small Cell Lung Cancer from Genomics to Therapeutics: A Framework for Community Practice Integration to Arrive at Personalized Therapy Strategies. J Clin Med 2020; 9:E1870. [PMID: 32549358 PMCID: PMC7356243 DOI: 10.3390/jcm9061870] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/25/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a heterogeneous disease, and therapeutic management has advanced with the identification of various key oncogenic mutations that promote lung cancer tumorigenesis. Subsequent studies have developed targeted therapies against these oncogenes in the hope of personalizing therapy based on the molecular genomics of the tumor. This review presents approved treatments against actionable mutations in NSCLC as well as promising targets and therapies. We also discuss the current status of molecular testing practices in community oncology sites that would help to direct oncologists in lung cancer decision-making. We propose a collaborative framework between community practice and academic sites that can help improve the utilization of personalized strategies in the community, through incorporation of increased testing rates, virtual molecular tumor boards, vendor-based oncology clinical pathways, and an academic-type singular electronic health record system.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (S.R.); (I.M.); (R.P.); (B.L.); (T.T.); (P.K.)
| |
Collapse
|
22
|
Chang H, Sasson A, Srinivasan S, Golhar R, Greenawalt DM, Geese WJ, Green G, Zerba K, Kirov S, Szustakowski J. Bioinformatic Methods and Bridging of Assay Results for Reliable Tumor Mutational Burden Assessment in Non-Small-Cell Lung Cancer. Mol Diagn Ther 2020; 23:507-520. [PMID: 31250328 PMCID: PMC6675777 DOI: 10.1007/s40291-019-00408-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction Tumor mutational burden (TMB) has emerged as a clinically relevant biomarker that may be associated with immune checkpoint inhibitor efficacy. Standardization of TMB measurement is essential for implementing diagnostic tools to guide treatment. Objective Here we describe the in-depth evaluation of bioinformatic TMB analysis by whole exome sequencing (WES) in formalin-fixed, paraffin-embedded samples from a phase III clinical trial. Methods In the CheckMate 026 clinical trial, TMB was retrospectively assessed in 312 patients with non-small-cell lung cancer (58% of the intent-to-treat population) who received first-line nivolumab treatment or standard-of-care chemotherapy. We examined the sensitivity of TMB assessment to bioinformatic filtering methods and assessed concordance between TMB data derived by WES and the FoundationOne® CDx assay. Results TMB scores comprising synonymous, indel, frameshift, and nonsense mutations (all mutations) were 3.1-fold higher than data including missense mutations only, but values were highly correlated (Spearman’s r = 0.99). Scores from CheckMate 026 samples including missense mutations only were similar to those generated from data in The Cancer Genome Atlas, but those including all mutations were generally higher. Using databases for germline subtraction (instead of matched controls) showed a trend for race-dependent increases in TMB scores. WES and FoundationOne CDx outputs were highly correlated (Spearman’s r = 0.90). Conclusions Parameter variation can impact TMB calculations, highlighting the need for standardization. Encouragingly, differences between assays could be accounted for by empirical calibration, suggesting that reliable TMB assessment across assays, platforms, and centers is achievable.
Collapse
Affiliation(s)
- Han Chang
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | - Ariella Sasson
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | - Sujaya Srinivasan
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | - Ryan Golhar
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | | | - William J Geese
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | - George Green
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | - Kim Zerba
- Global Biometric Sciences, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Stefan Kirov
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, 08648, USA
| | | |
Collapse
|
23
|
TMB: a promising immune-response biomarker, and potential spearhead in advancing targeted therapy trials. Cancer Gene Ther 2020; 27:841-853. [PMID: 32341410 DOI: 10.1038/s41417-020-0174-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 01/07/2023]
Abstract
Immune checkpoint inhibition (ICI) has revolutionized cancer treatment, and produced durable responses in many cancer types. However, there remains a subset of patients that do not respond despite their tumors exhibiting PD-L1 expression, which highlights the need for additional biomarkers relevant to response. Here, we review checkpoint inhibitor signal pathways, resistance and sensitivity mechanisms, as well as response rates. We also investigate the correlation and response to ICI with BRCA1/2 mutation status and homologous recombination deficient tumors. Collectively we show that the use of tumor mutational burden may be effective as an emerging biomarker.
Collapse
|
24
|
Lee M, Eng G, Barbari SR, Deshpande V, Shcherbakova PV, Gala MK. Homologous Recombination Repair Truncations Predict Hypermutation in Microsatellite Stable Colorectal and Endometrial Tumors. Clin Transl Gastroenterol 2020; 11:e00149. [PMID: 32352724 PMCID: PMC7145036 DOI: 10.14309/ctg.0000000000000149] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/13/2020] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Somatic mutations in BRCA1/2 and other homologous recombination repair (HRR) genes have been associated with sensitivity to PARP inhibitors and/or platinum agents in several cancers, whereas hypermutant tumors caused by alterations in POLE or mismatch repair genes have demonstrated robust responses to immunotherapy. We investigated the relationship between somatic truncations in HRR genes and hypermutation in colorectal cancer (CRC) and endometrial cancer (EC). METHODS We analyzed the mutational spectra associated with somatic BRCA1/2 truncations in multiple genomic cohorts (N = 2,335). From these results, we devised a classifier incorporating HRR genes to predict hypermutator status among microsatellite stable (MSS) tumors. Using additional genomic cohorts (N = 1,439) and functional in vivo assays, we tested the classifier to disambiguate POLE variants of unknown significance and identify MSS hypermutators without somatic POLE exonuclease domain mutations. RESULTS Hypermutator phenotypes were prevalent among CRCs with somatic BRCA1/2 truncations (50/62, 80.6%) and ECs with such mutations (44/47, 93.6%). The classifier predicted MSS hypermutators with a cumulative true-positive rate of 100% in CRC and 98.0% in EC and a false-positive rate of 0.07% and 0.63%. Validated by signature analyses of tumor exomes and in vivo assays, the classifier accurately reassigned multiple POLE variants of unknown significance as pathogenic and identified MSS hypermutant samples without POLE exonuclease domain mutations. DISCUSSION Somatic truncations in HRR can accurately fingerprint MSS hypermutators with or without known pathogenic exonuclease domain mutations in POLE and may serve as a low-cost biomarker for immunotherapy decisions in MSS CRC and EC.
Collapse
Affiliation(s)
- Minyi Lee
- Gastrointestinal Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - George Eng
- Gastrointestinal Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephanie R. Barbari
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vikram Deshpande
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Polina V. Shcherbakova
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Manish K. Gala
- Gastrointestinal Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
25
|
McCord M, Steffens A, Javier R, Kam KL, McCortney K, Horbinski C. The efficacy of DNA mismatch repair enzyme immunohistochemistry as a screening test for hypermutated gliomas. Acta Neuropathol Commun 2020; 8:15. [PMID: 32051040 PMCID: PMC7017562 DOI: 10.1186/s40478-020-0892-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
A subset of gliomas has DNA repair defects that lead to hypermutated genomes. While such tumors are resistant to alkylating chemotherapies, they may also express more mutant neoantigens on their cell surfaces, and thus be more responsive to immunotherapies. A fast, inexpensive method of screening for hypermutated gliomas would therefore be of great clinical value. Since immunohistochemistry (IHC) for the DNA mismatch repair (MMR) proteins Msh2, Msh6, Mlh1, and Pms2 is already used to screen for hypermutated colorectal cancers, we sought to determine whether that panel might have similar utility in gliomas. MMR IHC was scored in 100 WHO grade I-IV gliomas (from 96 patients) with known tumor mutation burden (TMB), while blinded to TMB data. Cases included 70 grade IV GBMs, 13 grade III astrocytomas, 4 grade II astrocytomas (3 diffuse astrocytomas and 1 pleomorphic xanthoastrocytoma), 1 grade I pilocytic astrocytoma, 2 grade III oligodendrogliomas, 7 grade II oligodendrogliomas, and 3 grade I glioneuronal tumors. Eight of 100 tumors showed loss of one or more MMR proteins by IHC, and all 8 were hypermutated. Among the remaining 92 gliomas with intact MMR IHC, only one was hypermutated; that tumor had an inactivating mutation in another DNA repair gene, ATM. Overall accuracy, sensitivity, and specificity for DNA MMR IHC compared to the gold standard of TMB were 99, 89, and 100%, respectively. The strongest correlates with hypermutation were prior TMZ treatment, MGMT promoter methylation, and IDH1 mutation. Among the 8 MMR-deficient hypermutated gliomas, 4 (50%) contained both MMR-lost and MMR-retained tumor cells. Together, these data suggest that MMR IHC could be a viable front-line screening test for gliomas in which immunotherapy is being considered. They also suggest that not all cells in a hypermutated glioma may actually be MMR-deficient, a finding that might need to be considered when treating such tumors with immunotherapies.
Collapse
|
26
|
Mambetsariev I, Wang Y, Chen C, Nadaf S, Pharaon R, Fricke J, Amanam I, Amini A, Bild A, Chu P, Erhunmwunsee L, Kim J, Munu J, Pillai R, Raz D, Sampath S, Vora L, Qiu F, Smith L, Batra SK, Massarelli E, Koczywas M, Reckamp K, Salgia R. Precision medicine and actionable alterations in lung cancer: A single institution experience. PLoS One 2020; 15:e0228188. [PMID: 32045431 PMCID: PMC7012442 DOI: 10.1371/journal.pone.0228188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Oncology has become more reliant on new testing methods and a greater use of electronic medical records, which provide a plethora of information available to physicians and researchers. However, to take advantage of vital clinical and research data for precision medicine, we must initially make an effort to create an infrastructure for the collection, storage, and utilization of this information with uniquely designed disease-specific registries that could support the collection of a large number of patients. MATERIALS AND METHODS In this study, we perform an in-depth analysis of a series of lung adenocarcinoma patients (n = 415) with genomic and clinical data in a recently created thoracic patient registry. RESULTS Of the 415 patients with lung adenocarcinoma, 59% (n = 245) were female; the median age was 64 (range, 22-92) years with a median OS of 33.29 months (95% CI, 29.77-39.48). The most common actionable alterations were identified in EGFR (n = 177/415 [42.7%]), ALK (n = 28/377 [7.4%]), and BRAF V600E (n = 7/288 [2.4%]). There was also a discernible difference in survival for 222 patients, who had an actionable alteration, with a median OS of 39.8 months as compared to 193 wild-type patients with a median OS of 26.0 months (P<0.001). We identified an unprecedented number of actionable alterations [53.5% (222/415)], including distinct individual alteration rates, as compared with 15.0% and 22.3% in TCGA and GENIE respectively. CONCLUSION The use of patient registries, focused genomic panels and the appropriate use of clinical guidelines in community and academic settings may influence cohort selection for clinical trials and improve survival outcomes.
Collapse
Affiliation(s)
- Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Yingyu Wang
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Chen Chen
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Sorena Nadaf
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Rebecca Pharaon
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Idoroenyi Amanam
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Arya Amini
- Department of Radiation Oncology, City of Hope, Duarte, California, United States of America
| | - Andrea Bild
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Peiguo Chu
- Department of Pathology, City of Hope, Duarte, California, United States of America
| | - Loretta Erhunmwunsee
- Department of Thoracic Surgery, City of Hope, Duarte, California, United States of America
| | - Jae Kim
- Department of Thoracic Surgery, City of Hope, Duarte, California, United States of America
| | - Janet Munu
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Raju Pillai
- Department of Pathology, City of Hope, Duarte, California, United States of America
| | - Dan Raz
- Department of Thoracic Surgery, City of Hope, Duarte, California, United States of America
| | - Sagus Sampath
- Department of Radiation Oncology, City of Hope, Duarte, California, United States of America
| | - Lalit Vora
- Department of Diagnostic Radiology, City of Hope, Duarte, California, United States of America
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Lynette Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Erminia Massarelli
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Marianna Koczywas
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Karen Reckamp
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| |
Collapse
|
27
|
Noskova H, Kyr M, Pal K, Merta T, Mudry P, Polaskova K, Ivkovic TC, Adamcova S, Hornakova T, Jezova M, Kren L, Sterba J, Slaby O. Assessment of Tumor Mutational Burden in Pediatric Tumors by Real-Life Whole-Exome Sequencing and In Silico Simulation of Targeted Gene Panels: How the Choice of Method Could Affect the Clinical Decision? Cancers (Basel) 2020; 12:cancers12010230. [PMID: 31963488 PMCID: PMC7016876 DOI: 10.3390/cancers12010230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Tumor mutational burden (TMB) is an emerging genomic biomarker in cancer that has been associated with improved response to immune checkpoint inhibitors (ICIs) in adult cancers. It was described that variability in TMB assessment is introduced by different laboratory techniques and various settings of bioinformatic pipelines. In pediatric oncology, no study has been published describing this variability so far. Methods: In our study, we performed whole exome sequencing (WES, both germline and somatic) and calculated TMB in 106 patients with high-risk/recurrent pediatric solid tumors of 28 distinct cancer types. Subsequently, we used WES data for TMB calculation using an in silico approach simulating two The Food and Drug Administration (FDA)-approved/authorized comprehensive genomic panels for cancer. Results: We describe a strong correlation between WES-based and panel-based TMBs; however, we show that this high correlation is significantly affected by inclusion of only a few hypermutated cases. In the series of nine cases, we determined TMB in two sequentially collected tumor tissue specimens and observed an increase in TMB along with tumor progression. Furthermore, we evaluated the extent to which potential ICI indication could be affected by variability in techniques and bioinformatic pipelines used for TMB assessment. We confirmed that this technological variability could significantly affect ICI indication in pediatric cancer patients; however, this significance decreases with the increasing cut-off values. Conclusions: For the first time in pediatric oncology, we assessed the reliability of TMB estimation across multiple pediatric cancer types using real-life WES and in silico analysis of two major targeted gene panels and confirmed a significant technological variability to be introduced by different laboratory techniques and various settings of bioinformatic pipelines.
Collapse
Affiliation(s)
- Hana Noskova
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic; (H.N.); (K.P.); (T.C.I.); (S.A.); (T.H.)
- Department of Pediatric Oncology, University Hospital Brno, 613 00 Brno, Czech Republic; (M.K.); (T.M.); (P.M.); (K.P.)
| | - Michal Kyr
- Department of Pediatric Oncology, University Hospital Brno, 613 00 Brno, Czech Republic; (M.K.); (T.M.); (P.M.); (K.P.)
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Karol Pal
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic; (H.N.); (K.P.); (T.C.I.); (S.A.); (T.H.)
- Department of Hematology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Tomas Merta
- Department of Pediatric Oncology, University Hospital Brno, 613 00 Brno, Czech Republic; (M.K.); (T.M.); (P.M.); (K.P.)
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno, 613 00 Brno, Czech Republic; (M.K.); (T.M.); (P.M.); (K.P.)
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Kristyna Polaskova
- Department of Pediatric Oncology, University Hospital Brno, 613 00 Brno, Czech Republic; (M.K.); (T.M.); (P.M.); (K.P.)
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Tina Catela Ivkovic
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic; (H.N.); (K.P.); (T.C.I.); (S.A.); (T.H.)
| | - Sona Adamcova
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic; (H.N.); (K.P.); (T.C.I.); (S.A.); (T.H.)
| | - Tekla Hornakova
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic; (H.N.); (K.P.); (T.C.I.); (S.A.); (T.H.)
| | - Marta Jezova
- Department of Pathology, University Hospital Brno, 62500 Brno, Czech Republic; (M.J.); (L.K.)
| | - Leos Kren
- Department of Pathology, University Hospital Brno, 62500 Brno, Czech Republic; (M.J.); (L.K.)
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno, 613 00 Brno, Czech Republic; (M.K.); (T.M.); (P.M.); (K.P.)
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 60200 Brno, Czech Republic
- Correspondence: (J.S.); (O.S.)
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic; (H.N.); (K.P.); (T.C.I.); (S.A.); (T.H.)
- Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
- Department of Pathology, University Hospital Brno, 62500 Brno, Czech Republic; (M.J.); (L.K.)
- Correspondence: (J.S.); (O.S.)
| |
Collapse
|
28
|
Gutierrez ME, Price KS, Lanman RB, Nagy RJ, Shah I, Mathura S, Mulcahy M, Norden AD, Goldberg SL. Genomic Profiling for KRAS, NRAS, BRAF, Microsatellite Instability, and Mismatch Repair Deficiency Among Patients With Metastatic Colon Cancer. JCO Precis Oncol 2019; 3:PO.19.00274. [PMID: 32923867 PMCID: PMC7448804 DOI: 10.1200/po.19.00274] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Genomic testing is recognized in national guidelines as essential to guide appropriate therapy selection in metastatic colorectal cancer. Previous studies report adherence to testing guidelines is suboptimal, but current testing rates have not been assessed. This study reports testing rates in metastatic colon cancer (mCC) for guideline-recommended biomarkers in a US-based population. MATERIALS AND METHODS A retrospective review of data extracted from electronic medical records was performed to identify patients with pathologically confirmed mCC and describe patterns of guideline-aligned biomarker testing. Data were extracted from the electronic health records of 1,497 patients treated at 23 practices across the United States. Both community and academic centers were represented. RESULTS A total of 1,497 patients with mCC diagnosed between January 1, 2013 and December 31, 2017 were identified. Guideline-aligned biomarker testing rates for RAS, BRAF, and microsatellite instability/mismatch repair deficiency over this study period were 41%, 43%, and 51%, respectively. Patients were more likely to have guideline-aligned testing for RAS and BRAF if they were treated at an academic center, were diagnosed with de novo metastatic disease, and were female. In addition, patients < 65 years of age were more likely to have guideline-aligned RAS testing. Of the 177 patients (12% of cohort) who received anti-epidermal growth factor receptor therapy, only 50 (28%) had complete guideline-aligned biomarker testing. CONCLUSION Despite guideline recommendations and significant therapeutic implications, overall biomarker testing rates in mCC remain suboptimal. Adherence to guideline-recommended biomarker testing would potentially reduce exposure to expensive and ineffective therapies, resulting in improved patient outcomes.
Collapse
Affiliation(s)
- Martin E. Gutierrez
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | | | | | | | | | | | | | | | - Stuart L. Goldberg
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
- Cota Healthcare, New York, NY
| |
Collapse
|
29
|
Integrated genomic profiling expands clinical options for patients with cancer. Nat Biotechnol 2019; 37:1351-1360. [PMID: 31570899 DOI: 10.1038/s41587-019-0259-z] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/05/2019] [Indexed: 01/14/2023]
Abstract
Genomic analysis of paired tumor-normal samples and clinical data can be used to match patients to cancer therapies or clinical trials. We analyzed 500 patient samples across diverse tumor types using the Tempus xT platform by DNA-seq, RNA-seq and immunological biomarkers. The use of a tumor and germline dataset led to substantial improvements in mutation identification and a reduction in false-positive rates. RNA-seq enhanced gene fusion detection and cancer type classifications. With DNA-seq alone, 29.6% of patients matched to precision therapies supported by high levels of evidence or by well-powered studies. This proportion increased to 43.4% with the addition of RNA-seq and immunotherapy biomarker results. Combining these data with clinical criteria, 76.8% of patients were matched to at least one relevant clinical trial on the basis of biomarkers measured by the xT assay. These results indicate that extensive molecular profiling combined with clinical data identifies personalized therapies and clinical trials for a large proportion of patients with cancer and that paired tumor-normal plus transcriptome sequencing outperforms tumor-only DNA panel testing.
Collapse
|
30
|
Emerging epigenomic landscapes of pancreatic cancer in the era of precision medicine. Nat Commun 2019; 10:3875. [PMID: 31462645 PMCID: PMC6713756 DOI: 10.1038/s41467-019-11812-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
Genetic studies have advanced our understanding of pancreatic cancer at a mechanistic and translational level. Genetic concepts and tools are increasingly starting to be applied to clinical practice, in particular for precision medicine efforts. However, epigenomics is rapidly emerging as a promising conceptual and methodological paradigm for advancing the knowledge of this disease. More importantly, recent studies have uncovered potentially actionable pathways, which support the prediction that future trials for pancreatic cancer will involve the vigorous testing of epigenomic therapeutics. Thus, epigenomics promises to generate a significant amount of new knowledge of both biological and medical importance. In pancreatic cancer, the epigenomic landscape can strongly impact the disease phenotype. Here, the authors discuss recent advances in our understanding of pancreatic cancer epigenomics, and how this knowledge can integrate with precision medicine approaches in this lethal disease.
Collapse
|
31
|
Stenzinger A, Allen JD, Maas J, Stewart MD, Merino DM, Wempe MM, Dietel M. Tumor mutational burden standardization initiatives: Recommendations for consistent tumor mutational burden assessment in clinical samples to guide immunotherapy treatment decisions. Genes Chromosomes Cancer 2019; 58:578-588. [PMID: 30664300 PMCID: PMC6618007 DOI: 10.1002/gcc.22733] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 01/10/2023] Open
Abstract
Characterization of tumors utilizing next‐generation sequencing methods, including assessment of the number of somatic mutations (tumor mutational burden [TMB]), is currently at the forefront of the field of personalized medicine. Recent clinical studies have associated high TMB with improved patient response rates and survival benefit from immune checkpoint inhibitors; hence, TMB is emerging as a biomarker of response for these immunotherapy agents. However, variability in current methods for TMB estimation and reporting is evident, demonstrating a need for standardization and harmonization of TMB assessment methodology across assays and centers. Two uniquely placed organizations, Friends of Cancer Research (Friends) and the Quality Assurance Initiative Pathology (QuIP), have collaborated to coordinate efforts for international multistakeholder initiatives to address this need. Friends and QuIP, who have partnered with several academic centers, pharmaceutical organizations, and diagnostic companies, have adopted complementary, multidisciplinary approaches toward the goal of proposing evidence‐based recommendations for achieving consistent TMB estimation and reporting in clinical samples across assays and centers. Many factors influence TMB assessment, including preanalytical factors, choice of assay, and methods of reporting. Preliminary analyses highlight the importance of targeted gene panel size and composition, and bioinformatic parameters for reliable TMB estimation. Herein, Friends and QuIP propose recommendations toward consistent TMB estimation and reporting methods in clinical samples across assays and centers. These recommendations should be followed to minimize variability in TMB estimation and reporting, which will ensure reliable and reproducible identification of patients who are likely to benefit from immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg (on behalf of QuIP® GmbH, Berlin, Germany), Heidelberg, Germany
| | - Jeffrey D Allen
- Science Policy, Friends of Cancer Research, Washington, District of Columbia
| | - Jörg Maas
- Management Department, QuIP® GmbH, Berlin, Germany
| | - Mark D Stewart
- Science Policy, Friends of Cancer Research, Washington, District of Columbia
| | - Diana M Merino
- Science Policy, Friends of Cancer Research, Washington, District of Columbia
| | - Madison M Wempe
- Science Policy, Friends of Cancer Research, Washington, District of Columbia
| | - Manfred Dietel
- Institute of Pathology, Charité Berlin (on behalf of QuIP® GmbH, Berlin, Germany), Berlin, Germany
| |
Collapse
|
32
|
Abstract
The treatment landscape for non-small-cell lung cancer (NSCLC) has dramatically shifted over the past two decades. Targeted or precision medicine has primarily been responsible for this shift. Older paradigms of treating metastatic NSCLC with cytotoxic chemotherapy, while still important, have given way to evaluating tumor tissues for specific driver mutations that can be treated with targeted agents. Patients treated with targeted agents frequently have improved progression-free survival and overall survival compared to patients without a targetable driver mutation, highlighting the clinical benefit of precision medicine. In this chapter, we explore the historic landmark trials, the current state of the field, and potential future targets under investigation, in this exciting, rapidly evolving discipline of precision medicine in lung cancer.
Collapse
|
33
|
Roberts JS, Gornick MC, Le LQ, Bartnik NJ, Zikmund-Fisher BJ, Chinnaiyan AM. Next-generation sequencing in precision oncology: Patient understanding and expectations. Cancer Med 2019; 8:227-237. [PMID: 30600607 PMCID: PMC6346219 DOI: 10.1002/cam4.1947] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Implementation of precision oncology interventions poses several challenges to informed consent and patient education. This study assessed cancer patients' understanding, expectations, and outcomes regarding participation in research examining the impact of matched tumor and germline sequencing on their clinical care. METHODS A total of 297 patients (mean age: 59 years; 50% female; 96% white) with refractory, metastatic cancer were surveyed, including 217 who completed surveys both before and after undergoing integrated whole exome and transcriptome sequencing as part of a larger clinical research study. RESULTS At baseline, the vast majority of patients expected to receive several potential direct benefits from study participation, including written reports of sequencing findings (88%), greater understanding of the causes of their cancer (74%), and participation in clinical trials for which sequencing results would make them eligible (84%). In most cases, these benefits were not realized by study completion. Despite explanations from study personnel to the contrary, most participants (67%-76%) presumed that incidental germline sequencing findings relevant to noncancerous health conditions (eg, diabetes) would automatically be disclosed to them. Patients reported low levels of concern about study risks at baseline and low levels of regret about study participation at follow-up. CONCLUSIONS Findings suggest that cancer patients participating in precision oncology intervention research have largely unfulfilled expectations of direct benefits related to their study participation. Increased focus on patient education to supplement the informed consent process may help manage patients' expectations regarding the extent and likelihood of benefits received as a result of undergoing genomic sequencing.
Collapse
Affiliation(s)
- J. Scott Roberts
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
- Center for Bioethics & Social Sciences in MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Michele C. Gornick
- Center for Bioethics & Social Sciences in MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Lan Q. Le
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
| | - Natalie J. Bartnik
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
| | - Brian J. Zikmund-Fisher
- Department of Health Behavior and Health EducationUniversity of Michigan School of Public HealthAnn ArborMichigan
- Center for Bioethics & Social Sciences in MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Arul M. Chinnaiyan
- Rogel Cancer CenterUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of PathologyUniversity of Michigan Medical SchoolAnn ArborMichigan
- Michigan Center for Translational PathologyUniversity of Michigan Medical SchoolAnn ArborMichigan
- Department of UrologyUniversity of Michigan Medical SchoolAnn ArborMichigan
- Howard Hughes Medical InstituteUniversity of Michigan Medical SchoolAnn ArborMichigan
| |
Collapse
|
34
|
Abstract
Genomic information is increasingly being incorporated into clinical cancer care. Large-scale sequencing efforts have deepened our understanding of the genomic landscape of cancer and contributed to the expanding catalog of alterations being leveraged to aid in cancer diagnosis, prognosis, and treatment. Genomic profiling can provide clinically relevant information regarding somatic point mutations, copy number alterations, translocations, and gene fusions. Genomic features, such as mutational burden, can also be measured by more comprehensive sequencing strategies and have shown value in informing potential treatment options. Ongoing clinical trials are evaluating the use of molecularly targeted agents in genomically defined subsets of cancers within and across tumor histologies. Continued advancements in clinical genomics promise to further expand the application of genomics-enabled medicine to a broader spectrum of oncology patients.
Collapse
Affiliation(s)
- Alison Roos
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Sara A Byron
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
35
|
Meléndez B, Van Campenhout C, Rorive S, Remmelink M, Salmon I, D'Haene N. Methods of measurement for tumor mutational burden in tumor tissue. Transl Lung Cancer Res 2018; 7:661-667. [PMID: 30505710 DOI: 10.21037/tlcr.2018.08.02] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Immunotherapies based on immune checkpoint inhibitors are emerging as an innovative treatment for different types of advanced cancers. While the utility of immune checkpoint inhibitors has been clearly demonstrated, the response rate is highly variable across individuals. Due to the cost and toxicity of these immunotherapies, a critical challenge in this field is the identification of predictive biomarkers to discriminate which patients may respond to immunotherapy. Recently, a high tumor mutational burden (TMB) has been identified as a genetic signature that is associated with a favorable outcome for immune checkpoint inhibitor therapy. The TMB is defined as the total number of nonsynonymous mutations per coding area of a tumor genome. Initially, it was determined using whole exome sequencing, but due to the high costs and long turnaround time of this method, targeted panel sequencing is currently being explored to measure TMB. In the near future, TMB evaluation may play an important role in immuno-oncology, but its implementation in a routine setting involves robust analytical and clinical validation. Standardization is also needed in order to make informed decisions about patients. This review presents the methodologies employed for determining TMB and discusses the factors that may have an impact on its measurement.
Collapse
Affiliation(s)
- Bárbara Meléndez
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Claude Van Campenhout
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Myriam Remmelink
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicky D'Haene
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
36
|
Lopez JV, Kamel B, Medina M, Collins T, Baums IB. Multiple Facets of Marine Invertebrate Conservation Genomics. Annu Rev Anim Biosci 2018; 7:473-497. [PMID: 30485758 DOI: 10.1146/annurev-animal-020518-115034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Conservation genomics aims to preserve the viability of populations and the biodiversity of living organisms. Invertebrate organisms represent 95% of animal biodiversity; however, few genomic resources currently exist for the group. The subset of marine invertebrates includes the most ancient metazoan lineages and possesses codes for unique gene products and possible keys to adaptation. The benefits of supporting invertebrate conservation genomics research (e.g., likely discovery of novel genes, protein regulatory mechanisms, genomic innovations, and transposable elements) outweigh the various hurdles (rare, small, or polymorphic starting materials). Here we review best conservation genomics practices in the laboratory and in silico when applied to marine invertebrates and also showcase unique features in several case studies of acroporid corals, crown-of-thorns starfish, apple snails, and abalone. Marine conservation genomics should also address how diversity can lead to unique marine innovations, the impact of deleterious variation, and how genomic monitoring and profiling could positively affect broader conservation goals (e.g., value of baseline data for in situ/ex situ genomic stocks).
Collapse
Affiliation(s)
- Jose V Lopez
- Department of Biological Sciences, Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Dania Beach, Florida 33004, USA;
| | - Bishoy Kamel
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, New Mexico 87131, USA;
| | - Mónica Medina
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA; ,
| | - Timothy Collins
- Department of Biological Sciences, Florida International University, Miami, Florida 33199, USA;
| | - Iliana B Baums
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA; ,
| |
Collapse
|
37
|
Mambetsariev I, Pharaon R, Nam A, Knopf K, Djulbegovic B, Villaflor VM, Vokes EE, Salgia R. Heuristic value-based framework for lung cancer decision-making. Oncotarget 2018; 9:29877-29891. [PMID: 30042820 PMCID: PMC6057456 DOI: 10.18632/oncotarget.25643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/04/2018] [Indexed: 11/25/2022] Open
Abstract
Heuristics and the application of fast-and-frugal trees may play a role in establishing a clinical decision-making framework for value-based oncology. We determined whether clinical decision-making in oncology can be structured heuristically based on the timeline of the patient's treatment, clinical intuition, and evidence-based medicine. A group of 20 patients with advanced non-small cell lung cancer (NSCLC) were enrolled into the study for extensive treatment analysis and sequential decision-making. The extensive clinical and genomic data allowed us to evaluate the methodology and efficacy of fast-and-frugal trees as a way to quantify clinical decision-making. The results of the small cohort will be used to further advance the heuristic framework as a way of evaluating a large number of patients within registries. Among the cohort whose data was analyzed, substitution and amplification mutations occurred most frequently. The top five most prevalent genomic alterations were TP53 (45%), ALK (40%), LRP1B (30%), CDKN2A (25%), and MYC (25%). These 20 cases were analyzed by this clinical decision-making process and separated into two distinctions: 10 straightforward cases that represented a clearer decision-making path and 10 complex cases that represented a more intricate treatment pathway. The myriad of information from each case and their distinct pathways was applied to create the foundation of a framework for lung cancer decision-making as an aid for oncologists. In late-stage lung cancer patients, the fast-and-frugal heuristics can be utilized as a strategy of quantifying proper decision-making with limited information.
Collapse
Affiliation(s)
- Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, USA
| | - Rebecca Pharaon
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, USA
| | - Arin Nam
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, USA
| | - Kevin Knopf
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | - Victoria M. Villaflor
- Department of Medicine (Hematology and Oncology), Northwestern University, Chicago, IL, USA
| | | | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA, USA
| |
Collapse
|