1
|
van Schaik LF, Engelhardt EG, Wilthagen EA, Steeghs N, Fernández Coves A, Joore MA, van Harten WH, Retèl VP. Factors for a broad technology assessment of comprehensive genomic profiling in advanced cancer, a systematic review. Crit Rev Oncol Hematol 2024; 202:104441. [PMID: 39002790 DOI: 10.1016/j.critrevonc.2024.104441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/12/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024] Open
Abstract
Comprehensive Genomic Profiling (CGP) allows for the identification of many targets. Reimbursement decision-making is, however, challenging because besides the health benefits of on-label treatments and costs, other factors related to diagnostic and treatment pathways may also play a role. The aim of this study was to identify which other factors are relevant for the technology assessment of CGP and to summarize the available evidence for these factors. After a scoping search and two expert sessions, five factors were identified: feasibility, test journey, wider implications of diagnostic results, organisation of laboratories, and "scientific spillover". Subsequently, a systematic search identified 83 studies collecting mainly evidence for the factors "test journey" and "wider implications of diagnostic results". Its nature was, however, of limited value for decision-making. We recommend the use of comparative strategies, uniformity in outcome definitions, and the inclusion of a comprehensive set of factors in future evidence generation.
Collapse
Affiliation(s)
- L F van Schaik
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands.
| | - E G Engelhardt
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands.
| | - E A Wilthagen
- Scientific Information Service, Netherlands Cancer Institute, Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam CX 1066, the Netherlands.
| | - N Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam CX 1066, the Netherlands.
| | - A Fernández Coves
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), P. Debyelaan 25, Oxford Building, P.O. Box 5800a, Maastricht, Limburg, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands.
| | - M A Joore
- Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), P. Debyelaan 25, Oxford Building, P.O. Box 5800a, Maastricht, Limburg, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands.
| | - W H van Harten
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands.
| | - V P Retèl
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, P.O. Box 90103, Amsterdam 1006 BE, the Netherlands; Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
2
|
Stackland S, Schnabel D, Dinan M, Presley CJ, Gross CP. Strength of Evidence Underlying the CMS-FDA Parallel Review of Comprehensive Genomic Profiling Tests in the Cancer Setting. J Natl Cancer Inst 2024:djae196. [PMID: 39288939 DOI: 10.1093/jnci/djae196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/17/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Although use of comprehensive genomic profiling (CGP) was approved by a novel CMS/FDA parallel review process, the quality of the supporting evidence is unclear. We evaluated the rigor of the peer-reviewed literature cited in the National Coverage Determination Memorandum for the FoundationOne CDx (F1CDx). METHODS We identified studies cited in the memorandum. Two independent researchers evaluated each study and applied a modified version of the Fryback and Thornbury hierarchy[1], an established framework for evaluating the efficacy of diagnostic tests. Studies focused on clinical outcomes were then categorized by study design, guided by recommendations from the Center for Medical Technology Policy. RESULTS The sample included 113 scientific studies. The majority (n = 60, 53.1%) used CGP outside the course of clinical care, and there was significant heterogeneity in the cancer types assessed and sequencing depth. We found 8 (7.1%) studies that assessed whether clinical care had changed due to CGP testing, and 38 (33.6%) assessed clinical outcomes. After excluding studies that tested for five or fewer genomic alterations, 25 remained in the clinical outcomes sample: Of these, only one included a comparator group that did not receive CGP testing. Only four studies used F1CDx as the primary genomic test, none of which compared the outcomes of patients who did vs did not receive the F1CDx test. CONCLUSIONS The findings indicate gaps in the supporting evidence for broad CGP use in patients with solid tumors. More rigorous studies that assess clinical utility would better inform the approval process for novel diagnostic tests.
Collapse
Affiliation(s)
| | | | - Michaela Dinan
- Yale School of Public Health, New Haven, CT, USA
- Cancer Outcomes Public Policy and Effectiveness Research Center, Yale School of Medicine, New Haven, CT, USA
| | - Carolyn J Presley
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Cary P Gross
- Cancer Outcomes Public Policy and Effectiveness Research Center, Yale School of Medicine, New Haven, CT, USA
- Yale School of Medicine, Section of General Internal Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Kraus FBT, Sultova E, Heinrich K, Jung A, Westphalen CB, Tauber CV, Kumbrink J, Rudelius M, Klauschen F, Greif PA, König A, Chelariu-Raicu A, Czogalla B, Burges A, Mahner S, Wuerstlein R, Trillsch F. Genetics and beyond: Precision Medicine Real-World Data for Patients with Cervical, Vaginal or Vulvar Cancer in a Tertiary Cancer Center. Int J Mol Sci 2024; 25:2345. [PMID: 38397025 PMCID: PMC10888648 DOI: 10.3390/ijms25042345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Advances in molecular tumor diagnostics have transformed cancer care. However, it remains unclear whether precision oncology has the same impact and transformative nature across all malignancies. We conducted a retrospective analysis of patients with human papillomavirus (HPV)-related gynecologic malignancies who underwent comprehensive molecular profiling and subsequent discussion at the interdisciplinary Molecular Tumor Board (MTB) of the University Hospital, LMU Munich, between 11/2017 and 06/2022. We identified a total cohort of 31 patients diagnosed with cervical (CC), vaginal or vulvar cancer. Twenty-two patients (fraction: 0.71) harbored at least one mutation. Fifteen patients (0.48) had an actionable mutation and fourteen (0.45) received a recommendation for a targeted treatment within the MTB. One CC patient received a biomarker-guided treatment recommended by the MTB and achieved stable disease on the mTOR inhibitor temsirolimus for eight months. Factors leading to non-adherence to MTB recommendations in other patient cases included informed patient refusal, rapid deterioration, stable disease, or use of alternative targeted but biomarker-agnostic treatments such as antibody-drug conjugates or checkpoint inhibitors. Despite a remarkable rate of actionable mutations in HPV-related gynecologic malignancies at our institution, immediate implementation of biomarker-guided targeted treatment recommendations remained low, and access to targeted treatment options after MTB discussion remained a major challenge.
Collapse
Affiliation(s)
- Fabian B. T. Kraus
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Elena Sultova
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Kathrin Heinrich
- Department of Medicine III, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Andreas Jung
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - C. Benedikt Westphalen
- Department of Medicine III, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Christina V. Tauber
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Frederick Klauschen
- Institute of Pathology, Comprehensive Cancer Center Munich, LMU University Hospital, Ludwig Maximilians University (LMU), 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Philipp A. Greif
- Department of Medicine III, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
- German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Alexander König
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Alexander Burges
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Rachel Wuerstlein
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| |
Collapse
|
4
|
Ballatore Z, Bozzi F, Cardea S, Savino FD, Migliore A, Tarantino V, Chiodi N, Ambrosini E, Bianchi F, Goteri G, Filosa A, Barbisan F, Bartoli E, Papa R, Berardi R. Molecular Tumour Board (MTB): From Standard Therapy to Precision Medicine. J Clin Med 2023; 12:6666. [PMID: 37892804 PMCID: PMC10607087 DOI: 10.3390/jcm12206666] [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: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Background: In the metastatic setting, cancer patients may not benefit from standard care regimes and their diseases undergo drug resistance due to tumour cell heterogeneity and genomic landscape complexity. In recent years, there have been several attempts to personalise the diagnostic-therapeutic path and to propose novel strategies based on not only histological test results but also on each patient's clinical history and molecular biology. Profiling molecular tests allows physicians to investigate the single tumour genomic landscape and to promote targeted approaches. The Molecular Tumour Board (MTB) is a multidisciplinary committee dedicated to selecting individualised and targeted therapeutic strategies appropriate for patients suffering from diseases that present resistance to standard care. Materials and Methods: Our MTB settled in "Azienda Ospedaliero Universitaria delle Marche", Ancona (AN), Italy, and includes oncologists, molecular biologists, geneticists, and other specialists. Clinical cases are referred by physicians to the MTB, through the Cancer and Research Centre of the Marche Region (CORM), through a telemedicine platform. Four possible molecular profiles are available: FoundationOne® CDx e FoundationOne®Liquid CDx and two local Next Generation Sequencing (NGS) panels, with 16 DNA genes and 10 RNA genes respectively. The resulting genetic mutations and their analyses are evaluated by all the members of the Board and a report for each patient is provided with medical recommendations. Results: from June 2021 to May 2023, we collected data from 97 referral patients (M: 49, F: 48). The mean age was 60.6 years (range 22-83 years). 90 cases were approved for testing. Only seven patients were not eligible for genomic profiling. In two patients who were eligible, molecular profiling was not performed because a tissue sample was not available. Off-label therapy was recommended for three patients. 5% of cases (5/88) showed addressable driver mutations associated with an existing targeted therapy and were immediately enrolled. Conclusions: MTB presents a powerful tool for offering precise medical goals. Our Department of Clinical Oncology also takes advantage of the important role of multidisciplinary teams, through the establishment of CORM and MTB meetings, within which there is the chance to perform NGS-based analyses. It will be important in the future to implement the use of genomic profiling to improve personalised care and to guide the choice of suitable therapies and more appropriate management of patients.
Collapse
Affiliation(s)
- Zelmira Ballatore
- Department of Medical Oncology, AOU delle Marche, 60126 Ancona, Italy; (Z.B.); (V.T.)
| | - Francesco Bozzi
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Sara Cardea
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Francesco Domenico Savino
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Antonella Migliore
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Valentina Tarantino
- Department of Medical Oncology, AOU delle Marche, 60126 Ancona, Italy; (Z.B.); (V.T.)
| | - Natalia Chiodi
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Elisa Ambrosini
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Francesca Bianchi
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| | - Gaia Goteri
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Alessandra Filosa
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Francesca Barbisan
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Elisa Bartoli
- Anatomia Patologica, AOU delle Marche, Università Politecnica delle Marche, 60126 Ancona, Italy; (G.G.); (A.F.); (F.B.); (E.B.)
| | - Roberto Papa
- Quality, Risk Management and Health Technology Innovation Unit, Department of Staff, AOU delle Marche, 60126 Ancona, Italy;
| | - Rossana Berardi
- Department of Medical Oncology, AOU delle Marche, 60126 Ancona, Italy; (Z.B.); (V.T.)
- Medical Oncology, Università Politecnica delle Marche, 60126 Ancona, Italy; (F.B.); (S.C.); (F.D.S.); (A.M.); (N.C.); (E.A.); (F.B.)
| |
Collapse
|
5
|
Gruber JJ, Afghahi A, Timms K, DeWees A, Gross W, Aushev VN, Wu HT, Balcioglu M, Sethi H, Scott D, Foran J, McMillan A, Ford JM, Telli ML. A phase II study of talazoparib monotherapy in patients with wild-type BRCA1 and BRCA2 with a mutation in other homologous recombination genes. NATURE CANCER 2022; 3:1181-1191. [PMID: 36253484 PMCID: PMC9586861 DOI: 10.1038/s43018-022-00439-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 08/29/2022] [Indexed: 11/09/2022]
Abstract
Talazoparib, a PARP inhibitor, is active in germline BRCA1 and BRCA2 (gBRCA1/2)-mutant advanced breast cancer, but its activity beyond gBRCA1/2 is poorly understood. We conducted Talazoparib Beyond BRCA ( NCT02401347 ), an open-label phase II trial, to evaluate talazoparib in patients with pretreated advanced HER2-negative breast cancer (n = 13) or other solid tumors (n = 7) with mutations in homologous recombination (HR) pathway genes other than BRCA1 and BRCA2. In patients with breast cancer, four patients had a Response Evaluation Criteria in Solid Tumors (RECIST) partial response (overall response rate, 31%), and three additional patients had stable disease of ≥6 months (clinical benefit rate, 54%). All patients with germline mutations in PALB2 (gPALB2; encoding partner and localizer of BRCA2) had treatment-associated tumor regression. Tumor or plasma circulating tumor DNA (ctDNA) HR deficiency (HRD) scores were correlated with treatment outcomes and were increased in all gPALB2 tumors. In addition, a gPALB2-associated mutational signature was associated with tumor response. Thus, talazoparib has been demonstrated to have efficacy in patients with advanced breast cancer who have gPALB2 mutations, showing activity in the context of HR pathway gene mutations beyond gBRCA1/2.
Collapse
Affiliation(s)
- Joshua J Gruber
- Department of Internal Medicine and Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Anosheh Afghahi
- Department of Medicine, University of Colorado, Aurora, CO, USA
| | | | - Alyssa DeWees
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wyatt Gross
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | | | - Danika Scott
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jessica Foran
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Alex McMillan
- Department of Statistics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - James M Ford
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
- Department of Genetics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Melinda L Telli
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
6
|
Milbury CA, Creeden J, Yip WK, Smith DL, Pattani V, Maxwell K, Sawchyn B, Gjoerup O, Meng W, Skoletsky J, Concepcion AD, Tang Y, Bai X, Dewal N, Ma P, Bailey ST, Thornton J, Pavlick DC, Frampton GM, Lieber D, White J, Burns C, Vietz C. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors. PLoS One 2022; 17:e0264138. [PMID: 35294956 PMCID: PMC8926248 DOI: 10.1371/journal.pone.0264138] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
FoundationOne®CDx (F1CDx) is a United States (US) Food and Drug Administration (FDA)-approved companion diagnostic test to identify patients who may benefit from treatment in accordance with the approved therapeutic product labeling for 28 drug therapies. F1CDx utilizes next-generation sequencing (NGS)-based comprehensive genomic profiling (CGP) technology to examine 324 cancer genes in solid tumors. F1CDx reports known and likely pathogenic short variants (SVs), copy number alterations (CNAs), and select rearrangements, as well as complex biomarkers including tumor mutational burden (TMB) and microsatellite instability (MSI), in addition to genomic loss of heterozygosity (gLOH) in ovarian cancer. CGP services can reduce the complexity of biomarker testing, enabling precision medicine to improve treatment decision-making and outcomes for cancer patients, but only if test results are reliable, accurate, and validated clinically and analytically to the highest standard available. The analyses presented herein demonstrate the extensive analytical and clinical validation supporting the F1CDx initial and subsequent FDA approvals to ensure high sensitivity, specificity, and reliability of the data reported. The analytical validation included several in-depth evaluations of F1CDx assay performance including limit of detection (LoD), limit of blank (LoB), precision, and orthogonal concordance for SVs (including base substitutions [SUBs] and insertions/deletions [INDELs]), CNAs (including amplifications and homozygous deletions), genomic rearrangements, and select complex biomarkers. The assay validation of >30,000 test results comprises a considerable and increasing body of evidence that supports the clinical utility of F1CDx to match patients with solid tumors to targeted therapies or immunotherapies based on their tumor's genomic alterations and biomarkers. F1CDx meets the clinical needs of providers and patients to receive guideline-based biomarker testing, helping them keep pace with a rapidly evolving field of medicine.
Collapse
Affiliation(s)
- Coren A. Milbury
- Department Product Development, Cambridge, MA, United States of America
| | - James Creeden
- Global Medical Affairs, Basel, MA, United States of America
| | - Wai-Ki Yip
- Department Product Development, Cambridge, MA, United States of America
| | - David L. Smith
- Department of Franchise Development, Cambridge, MA, United States of America
| | - Varun Pattani
- Department Product Development, Cambridge, MA, United States of America
| | - Kristi Maxwell
- Department of Health Economic and Outcomes Research & Payer Policy, Reimbursement, Cambridge, MA, United States of America
| | - Bethany Sawchyn
- Department of Scientific and Medical Publications, Clinical Operations, Cambridge, MA, United States of America
| | - Ole Gjoerup
- Department of Scientific and Medical Publications, Clinical Operations, Cambridge, MA, United States of America
| | - Wei Meng
- Department Product Development, Cambridge, MA, United States of America
| | - Joel Skoletsky
- Department Product Development, Cambridge, MA, United States of America
| | | | - Yanhua Tang
- Department Product Development, Cambridge, MA, United States of America
| | - Xiaobo Bai
- Department Product Development, Cambridge, MA, United States of America
| | - Ninad Dewal
- Department Product Development, Cambridge, MA, United States of America
| | - Pei Ma
- Department Product Development, Cambridge, MA, United States of America
| | - Shannon T. Bailey
- Department Product Development, Cambridge, MA, United States of America
| | - James Thornton
- Department Product Development, Cambridge, MA, United States of America
| | - Dean C. Pavlick
- Department of Cancer Genomics, Cambridge, MA, United States of America
| | | | - Daniel Lieber
- Department of Computational Biology, Cambridge, MA, United States of America
| | - Jared White
- Department of Computational Biology, Cambridge, MA, United States of America
| | - Christine Burns
- Department Product Development, Cambridge, MA, United States of America
| | - Christine Vietz
- Department Product Development, Cambridge, MA, United States of America
| |
Collapse
|
7
|
Charo LM, Eskander RN, Sicklick J, Kim KH, Lim HJ, Okamura R, Lee S, Subramanian R, Schwab R, Shatsky R, Plaxe S, Kato S, Kurzrock R. Real-World Data From a Molecular Tumor Board: Improved Outcomes in Breast and Gynecologic Cancers Patients With Precision Medicine. JCO Precis Oncol 2022; 6:e2000508. [PMID: 35005995 PMCID: PMC8769125 DOI: 10.1200/po.20.00508] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/20/2021] [Accepted: 11/17/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Next-generation sequencing is increasingly used in gynecologic and breast cancers. Multidisciplinary Molecular Tumor Board (MTB) may guide matched therapy; however, outcome data are limited. We evaluate the effect of the degree of matching of tumors to treatment as well as compliance to MTB recommendations on outcomes. METHODS Overall, 164 patients with consecutive gynecologic and breast cancers presented at MTB were assessed for clinicopathologic data, next-generation sequencing results, MTB recommendations, therapy received, and outcomes. Matching score (MS), defined as percentage of alterations targeted by treatment over total pathogenic alterations, and compliance to MTB recommendations were analyzed in context of oncologic outcomes. RESULTS Altogether, 113 women were evaluable for treatment after MTB; 54% received matched therapy. Patients with MS ≥ 40% had higher overall response rate (30.8% v 7.1%; P = .001), progression-free survival (PFS; hazard ratio [HR] 0.51; 95% CI, 0.31 to 0.85; P = .002), and a trend toward improved overall survival (HR 0.64; 95% CI, 0.34 to 1.25; P = .082) in univariate analysis. The PFS advantage remained significant in multivariate analysis (HR 0.5; 95% CI, 0.3 to 0.8; P = .006). Higher MTB recommendation compliance was significantly associated with improved median PFS (9.0 months for complete; 6.0 months for partial; 4.0 months for no compliance; P = .004) and overall survival (17.1 months complete; 17.8 months partial; 10.8 months none; P = .046). Completely MTB-compliant patients had higher MS (P < .001). In multivariate analysis comparing all versus none MTB compliance, overall response (HR 9.5; 95% CI, 2.6 to 35.0; P = .001) and clinical benefit (HR 8.8; 95% CI, 2.4 to 33.2; P = .001) rates were significantly improved with higher compliance. CONCLUSION Compliance to MTB recommendations resulted in higher degrees of matched therapy and correlates with improved outcomes in patients with gynecologic and breast cancers.
Collapse
Affiliation(s)
- Lindsey M. Charo
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Center, La Jolla, CA
| | - Ramez N. Eskander
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Center, La Jolla, CA
| | - Jason Sicklick
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
- Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, San Diego, CA
| | - Ki Hwan Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Hyo Jeong Lim
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, South Korea
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| | - Rupa Subramanian
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| | - Richard Schwab
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| | - Rebecca Shatsky
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| | - Steven Plaxe
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Center, La Jolla, CA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA
| |
Collapse
|
8
|
Čerina D, Matković V, Katić K, Lovasić IB, Šeparović R, Canjko I, Jakšić B, Fröbe A, Pleština S, Bajić Ž, Vrdoljak E. Precision Oncology in Metastatic Uterine Cancer; Croatian First-Year Experience of the Comprehensive Genomic Profiling in Everyday Clinical Practice. Pathol Oncol Res 2021; 27:1609963. [PMID: 34646088 PMCID: PMC8504363 DOI: 10.3389/pore.2021.1609963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/07/2021] [Indexed: 01/10/2023]
Abstract
Comprehensive genomic profiling (CGP) is gradually becoming an inevitable part of the everyday oncology clinical practice. The interpretation and optimal implementation of the results is one of the hot topics of modern-day oncology. According to the recent findings, uterine cancer harbors a high level of gene alterations but is still insufficiently explored. The primary goal of this project was to assess the proportion of patients with targetable mutations. Also, the aim was to define and emphasize potential opportunities as well as the problems we have faced in the first year of testing on the national level. We performed a multicentric, retrospective, nested cross-sectional analysis on the total population of Croatian patients with advanced/metastatic uterine cancer where the tumor CGP was performed during 2020. CGP of the tumor tissue of 32 patients revealed clinically relevant genomic alterations (CRGA) in 27 patients (84%) with a median of 3 (IQR 1-4) CRGA per patient. The most common CRGAs were those of phosphatide-inositol-3 kinases (PIK3) in 22 patients (69%), with 13/22 (59%) of those patients harboring PIK3CA mutation. The next most common CGRAs were ARID1A and PTEN mutations in 13 (41%) and 11 (34%) patients, respectively. Microsatellite status was determined as stable in 21 patients (66%) and highly unstable in 10 patients (31%). A high tumor mutational burden (≥10Muts/Mb) was reported in 12 patients (38%). CGP analysis reported some kind of targeted therapy for 28 patients (88%). CGP determined clinically relevant genomic alterations in the significant majority of patients with metastatic uterine cancer, defining it as a rich ground for further positioning and development of precision oncology.
Collapse
Affiliation(s)
- Dora Čerina
- Department of Oncology, School of Medicine, University Hospital Center Split, University of Split, Split, Croatia
| | - Višnja Matković
- Department of Gynecologic Oncology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Kristina Katić
- Department of Gynecologic Oncology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Ingrid Belac Lovasić
- Department of Radiotherapy and Oncology, University Hospital Center Rijeka, Rijeka, Croatia
| | - Robert Šeparović
- Department of Medical Oncology, Division of Medical Oncology, University Hospital for Tumors, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Ivana Canjko
- Department of Radiotherapy Oncology, University Hospital Center Osijek, Osijek, Croatia
| | - Blanka Jakšić
- Department of Oncology and Nuclear Medicine, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Ana Fröbe
- Department of Oncology and Nuclear Medicine, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Stjepko Pleština
- Department of Oncology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Žarko Bajić
- Research Unit "Dr. Mirko Grmek", University Psychiatric Hospital "Sveti Ivan", Zagreb, Croatia
| | - Eduard Vrdoljak
- Department of Oncology, School of Medicine, University Hospital Center Split, University of Split, Split, Croatia
| |
Collapse
|
9
|
Somasegar S, Hoppenot C, Kuchta K, Sereika A, Khandekar J, Rodriguez G, Moore E, Hurteau J, Vogel TJ. Outcomes after targeted treatment based on somatic tumor genetic testing for women with gynecologic cancers. Gynecol Oncol 2021; 163:220-228. [PMID: 34511240 DOI: 10.1016/j.ygyno.2021.08.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Molecular tumor profiling and next-generation sequencing are being increasingly utilized, but there are limited data on the therapeutic implications and potential benefits of targeted treatments. We aim to characterize gynecologic oncology patients referred for somatic tumor genetic mutation testing and assess survival outcomes, efficacy, and toxicities of those receiving targeted therapy. METHODS We conducted a retrospective chart review of gynecologic oncology patients referred for somatic tumor testing by next generation sequencing between 1/1/2012-8/23/2019. The primary objective was to compare overall and progression free survival between those treated with targeted therapy (group 1) versus traditional treatment (group 2). RESULTS Most patients (70%) had additional treatment options available based on actionable mutations. The median number of somatic mutations identified was 5 (range 0-53). Patients in group 1 had more actionable somatic mutations (median 2 versus 0, p < 0.001). There was no difference in OS (median 64 versus 76 months, p = 0.97) or PFS (median 2 versus 8 months, p = 0.05) between the groups. While fewer patients in group 1 experienced neuropathy (0 versus 5, p = 0.02), grade I/II thrombocytopenia (7 versus 13, p = 0.03), grade III/IV thrombocytopenia (0 versus 4, p = 0.02), and grade III/IV neutropenia (1 versus 9, p = 0.002), all other non-hematologic toxicities were similar in the two groups. CONCLUSIONS Most gynecologic cancer patients have actionable mutations and may benefit from a personalized targeted therapy treatment plan. Next generation sequencing can be used to identify clinically actionable mutations in gynecologic cancers and guide the selection of treatments, thereby expanding treatment options without worsening survival or toxicity.
Collapse
Affiliation(s)
- Sahana Somasegar
- Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL, United States of America.
| | - Claire Hoppenot
- Department of Obstetrics and Gynecology, University of Chicago Medicine, Chicago, IL, United States of America
| | - Kristine Kuchta
- Biostatistical Core, NorthShore University HealthSystem Research Institute, Evanston, IL, USA
| | - Annette Sereika
- Division of Oncology, Program in Personalized Medicine, Kellogg Cancer Center, Northshore University HealthSystem, Evanston, IL, United States of America
| | - Janardan Khandekar
- Division of Oncology, Program in Personalized Medicine, Kellogg Cancer Center, Northshore University HealthSystem, Evanston, IL, United States of America
| | - Gustavo Rodriguez
- Division of Gynecologic Oncology, Kellogg Cancer Center, Northshore University HealthSystem, Evanston, IL, United States of America
| | - Elena Moore
- Division of Gynecologic Oncology, Kellogg Cancer Center, Northshore University HealthSystem, Evanston, IL, United States of America
| | - Jean Hurteau
- Division of Gynecologic Oncology, Kellogg Cancer Center, Northshore University HealthSystem, Evanston, IL, United States of America; GlaxoSmithKline, US Medical Affairs, Women's Oncology Program, Waltham, MA, United States of America
| | - Tilley Jenkins Vogel
- Division of Gynecologic Oncology, Kellogg Cancer Center, Northshore University HealthSystem, Evanston, IL, United States of America
| |
Collapse
|
10
|
Arend RC, Goel N, Roane BM, Foxall ME, Dholakia J, Londoño AI, Wall JA, Leath CA, Huh WK. Systematic Next Generation Sequencing is feasible in clinical practice and identifies opportunities for targeted therapy in women with uterine cancer: Results from a prospective cohort study. Gynecol Oncol 2021; 163:85-92. [PMID: 34372972 DOI: 10.1016/j.ygyno.2021.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Both incidence and mortality of uterine cancer are on the rise and mortality is higher for African American women. The aim of our study was to evaluate how Next Generation Sequencing (NGS) may facilitate identification of and intervention for treatment disparities when integrated into clinical workflows. RESULTS Our cohort included 159 uterine cancer patients with recurrent/progressive and newly diagnosed advanced stage and/or high-risk histology. The most common tumor histological subtypes included EEC (n = 67), SEC (n = 34), UCS (n = 20), and mixed (n = 14). Black patients were most likely to present with aggressive histology: (SEC, 34.0%) and carcinosarcoma (UCS, 14.0%). The four most common mutations across all subtypes were TP53, PIK3CA, PTEN, and ARID1A. There was racial disparity between Black versus non-Black patients who were initiated on targeted therapy (28.2% vs. 38.2%, respectively) and clinical trial (15% vs. 22.6%, respectively). Compared to non-Black patients, Black patients had a significantly higher percentage TP53 mutations (p < 0.05) and a significantly lower percentage ARID1A mutations (p < 0.05). CONCLUSIONS NGS for uterine malignancies provides actionable information for targetable mutations and/or clinical trial enrollment in most patients; further investigation is necessary to identify potentially modifiable factors contributing to current disparities that may improve targeted therapy uptake and clinical trial participation.
Collapse
Affiliation(s)
- Rebecca C Arend
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America.
| | - Nidhi Goel
- University of Alabama School of Medicine, Birmingham, AL, United States of America
| | - Brandon M Roane
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - McKenzie E Foxall
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Jhalak Dholakia
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Angelina I Londoño
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Jaclyn A Wall
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Charles A Leath
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| | - Warner K Huh
- University of Alabama in Birmingham, Division of Gynecologic Oncology, Birmingham, AL, United States of America
| |
Collapse
|
11
|
Larson KL, Huang B, Weiss HL, Hull P, Westgate PM, Miller RW, Arnold SM, Kolesar JM. Clinical Outcomes of Molecular Tumor Boards: A Systematic Review. JCO Precis Oncol 2021; 5:PO.20.00495. [PMID: 34632252 PMCID: PMC8277300 DOI: 10.1200/po.20.00495] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/18/2021] [Accepted: 06/09/2021] [Indexed: 01/12/2023] Open
Abstract
We conducted this systematic review to evaluate the clinical outcomes associated with molecular tumor board (MTB) review in patients with cancer. METHODS A systematic search of PubMed was performed to identify studies reporting clinical outcomes in patients with cancer who were reviewed by an MTB. To be included, studies had to report clinical outcomes, including clinical benefit, response, progression-free survival, or overall survival. Two reviewers independently selected studies and assessed quality with the Quality Assessment Tool for Before-After (Pre-Post) Studies with No Control Group or the Quality Assessment Tool for Observational Cohort and Cross-Sectional Studies depending on the type of study being reviewed. RESULTS Fourteen studies were included with a total of 3,328 patients with cancer. All studies included patients without standard-of-care treatment options and usually with multiple prior lines of therapy. In studies reporting response rates, patients receiving MTB-recommended therapy had overall response rates ranging from 0% to 67%. In the only trial powered on clinical outcome and including a control group, the group receiving MTB-recommended therapy had significantly improved rate of progression-free survival compared with those receiving conventional therapy. CONCLUSION Although data quality is limited by a lack of prospective randomized controlled trials, MTBs appear to improve clinical outcomes for patients with cancer. Future research should concentrate on prospective trials and standardization of approach and outcomes.
Collapse
Affiliation(s)
- Kara L. Larson
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
| | - Bin Huang
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
- Kentucky Cancer Registry, University of
Kentucky, Lexington, Kentucky
| | - Heidi L. Weiss
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
| | - Pam Hull
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
| | - Philip M. Westgate
- Department of Biostatistics, University of
Kentucky, Lexington, Kentucky
| | - Rachel W. Miller
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
- Department of Obstetrics and Gynecology,
University of Kentucky, Lexington, Kentucky
| | - Susanne M. Arnold
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
- Department of Internal Medicine,
University of Kentucky, Lexington, Kentucky
| | - Jill M. Kolesar
- Markey Cancer Center, University of
Kentucky, Lexington, Kentucky
- Department of Pharmacy Practice and
Science, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
12
|
Survival outcomes are associated with genomic instability in luminal breast cancers. PLoS One 2021; 16:e0245042. [PMID: 33534788 PMCID: PMC7857737 DOI: 10.1371/journal.pone.0245042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the leading cause of cancer related death among women. Breast cancers are generally diagnosed and treated based on clinical and histopathological features, along with subtype classification determined by the Prosigna Breast Cancer Prognostic Gene Signature Assay (also known as PAM50). Currently the copy number alteration (CNA) landscape of the tumour is not considered. We set out to examine the role of genomic instability (GI) in breast cancer survival since CNAs reflect GI and correlate with survival in other cancers. We focused on the 70% of breast cancers classified as luminal and carried out a comprehensive survival and association analysis using Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) data to determine whether CNA Score Quartiles derived from absolute CNA counts are associated with survival. Analysis revealed that patients diagnosed with luminal A breast cancer have a CNA landscape associated with disease specific survival, suggesting that CNA Score can provide a statistically robust prognostic factor. Furthermore, stratification of patients into subtypes based on gene expression has shown that luminal A and B cases overlap, and it is in this region we largely observe luminal A cases with reduced survival outlook. Therefore, luminal A breast cancer patients with quantitatively elevated CNA counts may benefit from more aggressive therapy. This demonstrates how individual genomic landscapes can facilitate personalisation of therapeutic interventions to optimise survival outcomes.
Collapse
|
13
|
Hay MA, Severson EA, Miller VA, Liebner DA, Vergilio JA, Millis SZ, Chen JL. Identifying Opportunities and Challenges for Patients With Sarcoma as a Result of Comprehensive Genomic Profiling of Sarcoma Specimens. JCO Precis Oncol 2020; 4:1900227. [PMID: 32923870 PMCID: PMC7446311 DOI: 10.1200/po.19.00227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Comprehensive genomic profiling (CGP) of sarcomas is rapidly being integrated into routine clinical care to help refine diagnosis and prognosis and determine treatment. However, little is known about barriers to successful CGP or its clinical utility in sarcoma. We set out to determine whether CGP alters physician treatment decision-making, and whether sarcoma subtypes influence the frequency of successful technical performance of CGP. METHODS A single-institution study evaluated profiling outcomes of 392 samples from patients with sarcoma, using a commercially available CGP panel. Of this group, 34 patients were evaluated prospectively (Decision Impact Trial) to evaluate the utility of CGP in physician decision-making. All cases were retrospectively analyzed to identify causes of CGP failure. RESULTS CGP successfully interrogated 75.3% (n = 295 of 392) of patients with sarcoma. Bone sarcomas had lower passing rates at 65.3% (n = 32 of 49) compared with soft tissue sarcomas at 76.7% (n = 263 of 343; P = .0008). Biopsy location also correlated with profiling efficiency. Bone biopsy specimens had a 52.8% (n = 19 of 36) passing rate versus lung (61.1%; n = 33 of 54) and abdomen (80.1%; n = 109 of 136) specimens. CGP altered physician treatment selection in 25% of evaluable patients (n = 7 of 28) and was associated with improved progression-free survival. CONCLUSION To our knowledge, this is the largest technical evaluation of the performance of CGP in sarcoma. CGP was effectively performed in the vast majority of sarcoma samples and altered physician treatment selection. Tumor location and tissue subtype were key determinants of profiling success and associated with preanalytic variables that affect DNA and RNA quality. These results support standardized biopsy collection protocols to improve profiling outcomes.
Collapse
|
14
|
Przybylski DJ, Dow-Hillgartner EN, Reed MP, Fallon MJ. Current state assessment survey of challenges of pharmacogenomics within oncology pharmacy practice. J Oncol Pharm Pract 2020; 26:1374-1381. [DOI: 10.1177/1078155219896395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Purpose The goal of this survey was to identify opportunities for health systems to increase implementation and adoption of oncology-focused pharmacogenomics services. Methods An online survey assessing respondent demographics, baseline knowledge and training in pharmacogenomics, comfort level with pharmacogenomic data, and challenges of implementing clinical pharmacogenomic platforms was distributed to professional colleagues and over national oncology pharmacy listservs. Pharmacists were grouped based on their comfort level with pharmacogenomic data. Results were analyzed utilizing Pearson chi-square test. A p value of <0.05 was considered significant. Results A total of 84 participants from 58 cancer centers participated in the survey. Most participants were post-graduate year 2 trained and a majority reported being comfortable assessing oncology pharmacogenomic data. Respondents indicated that pharmacogenomics reported within the electronic medical record was the most common institutional process to support pharmacogenomics for oncology patients. Despite this, poor visibility of pharmacogenomics within the electronic medical record was the most challenging aspect of implementing a pharmacogenomic program. Additional challenges included lack of resources for pharmacogenomic programs, insurance denials for pharmacogenomic-driven testing and medication, and prolonged turnaround time of pharmacogenetic results. Length of practice, post-graduate year 2 residency training, institutions with pharmacist involvement on hematology/oncology molecular tumor board, and institutions where a pharmacist helped create local pharmacogenomic policies were significantly associated with respondents’ comfortability in assessing pharmacogenomics. Conclusion Oncology pharmacists reported substantial challenges in implementing a pharmacogenomic program. Future efforts to assist in developing pharmacogenomic efforts should focus on increasing pharmacist involvement, expanding education and training, and improving clinical decision support tools.
Collapse
|
15
|
Taghizadeh H, Mader RM, Müllauer L, Aust S, Polterauer S, Kölbl H, Seebacher V, Grimm C, Reinthaller A, Prager GW. Molecular Guided Treatments in Gynecologic Oncology: Analysis of a Real-World Precision Cancer Medicine Platform. Oncologist 2020; 25:e1060-e1069. [PMID: 32369643 PMCID: PMC7356753 DOI: 10.1634/theoncologist.2019-0904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Advanced gynecologic cancers have a poor prognosis and constitute a major challenge for adequate treatment strategies. By analyzing and targeting molecular alterations, molecular guided treatments may be a viable option for the treatment of advanced gynecologic cancers. PATIENTS AND METHODS In this single-center, real-world retrospective analysis of our platform for precision cancer medicine (PCM), we describe the molecular profiling of 72 patients diagnosed with different types of advanced gynecologic malignancies. Tumor samples of the patients were examined by next-generation sequencing panel and immunohistochemistry (IHC). RESULTS In total, we identified 209 genetic aberrations in 72 patients. The ten most frequent alterations were TP53 (n = 42, 20%), KRAS (n = 14, 6.6%), PIK3CA (n = 11, 5.2%), PIK3R1 (n = 9, 4.3%), ATR (n = 8, 3.8%), PTEN (n = 8, 3.8%), BRCA1 (n = 6, 2.8%), NF1 (n = 4, 1.9%), NOTCH1 (n = 4, 1.9%), and POLE (n = 4, 1.9%), which account for more than half of all molecular alterations (52.6%). In 21 (29.1%) patients only one mutation could be detected, and 44 (61.1%) patients had more than one mutation. No molecular alterations were detected in seven (9.7%) patients. IHC detected expression of phosphorylated mammalian target of rapamycin and epidermal growth factor receptor in 58 (80.6%) and 53 (73.6%) patients, respectively. In over two thirds (n = 49, 68.1%), a targeted therapy was suggested, based on the identified genetic aberrations. The most frequently recommended specific treatment was the combination of everolimus with exemestane (n = 18, 25 %). CONCLUSION Based on our observations, it seems that PCM might be a feasible approach for advanced gynecologic cancers with limited treatment options. IMPLICATIONS FOR PRACTICE Nowadays molecular profiling of advanced gynecologic malignancies is feasible in the clinical routine. A molecular portrait should be done for every patient with an advanced therapy-refractory gynecologic malignancy to offer molecular-based treatment concepts.
Collapse
Affiliation(s)
- Hossein Taghizadeh
- Clinical Division of Oncology, Department of Medicine I, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Robert M. Mader
- Clinical Division of Oncology, Department of Medicine I, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Leonhard Müllauer
- Clinical Institute of Pathology, Medical University of ViennaViennaAustria
| | - Stefanie Aust
- Department of Obstetrics and Gynecology, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Stephan Polterauer
- Department of Obstetrics and Gynecology, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Heinz Kölbl
- Department of Obstetrics and Gynecology, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Veronika Seebacher
- Department of Obstetrics and Gynecology, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Christoph Grimm
- Department of Obstetrics and Gynecology, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Alexander Reinthaller
- Department of Obstetrics and Gynecology, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| | - Gerald W. Prager
- Clinical Division of Oncology, Department of Medicine I, Medical University of ViennaViennaAustria
- Comprehensive Cancer Center ViennaViennaAustria
| |
Collapse
|
16
|
|
17
|
Actionable molecular alterations in advanced gynaecologic malignancies: updated results from the ProfiLER programme. Eur J Cancer 2019; 118:156-165. [PMID: 31351267 DOI: 10.1016/j.ejca.2019.06.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/05/2019] [Accepted: 06/15/2019] [Indexed: 11/20/2022]
Abstract
OBJECTIVES The objectives of this study were to identify actionable genomic alterations in the gynaecological subpopulation of the ProfiLER programme and to report clinical efficacy of recommended targeted treatment (RTT). METHODS The ProfiLER programme (NCT01774409) is a multicentric prospective trial aiming to implement molecular profiling in patients with advanced refractory cancers. In this programme, tumour DNA is analysed by targeted next-generation sequencing (69 genes) and by whole genome array comparative genomic hybridisation. Clinical cases and genomic profiles are presented in a dedicated molecular tumour board to guide treatment strategies. We report here an analysis of patients with gynaecological cancers included in this trial. RESULTS From February 2013 to February 2017, 309 patients with gynaecologic cancer were included; 279 (90%) had sufficient quality, and 131 patients (42.4%) had at least one actionable genomic alteration in cancer cells. Four alterations were shared by at least 3% of the patients: 27 (9.7%) PIK3CA mutations, 15 (5.4%) KRAS mutations, 11 (3.9%) ERBB2 amplifications and 9 (3.2%) CDKN2A deletions. Forty-one treatments were initiated among 39 patients (12.6% of the screened population): 8 (20%) had a partial response, and other 10 (24%) had a stable disease. The median progression-free survival was 2.7 months. The median overall survival was 15.6 months for patients who received a RTT. CONCLUSION Molecular profiling identified actionable alterations in 42.4% of patients with advanced refractory gynaecologic cancer, but only 12.6% were treated with a RTT. Among them, 46% derived clinical benefit (5.8% of the screened population).
Collapse
|
18
|
Rodriguez-Freixinos V, Lheureux S, Mandilaras V, Clarke B, Dhani NC, Mackay H, Butler MO, Wang L, Siu LL, Kamel-Reid S, Stockley T, Bedard PL, Oza AM. Impact of somatic molecular profiling on clinical trial outcomes in rare epithelial gynecologic cancer patients. Gynecol Oncol 2019; 153:304-311. [PMID: 30792002 DOI: 10.1016/j.ygyno.2019.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Conducting clinical trials in rare malignancies is challenging due to the limited number of patients and differences in biologic behavior. We investigated the feasibility and clinical utility of using genomic profiling for rare gynecologic malignancies. METHODS Rare epithelial gynecologic cancer patients were analyzed for somatic variants through an institutional molecular profiling program using the Sequenom MassArray platform or the TruSeq Amplicon Cancer Panel on the MiSeq platform. Clinical trial outcomes by RECIST 1.1, and time on treatment were evaluated. RESULTS From March 2012 to November 2015, 767 gynecologic patients were enrolled and 194 (27%) were classified as rare epithelial malignancies. At least one somatic mutation was identified in 72% of patients, most commonly in TP53 (39%), KRAS (28%) and PIK3CA (27%). A total of 14% of patients were treated on genotype-matched trials. There were no significant differences in overall response rate between genotype-matched versus unmatched trials, nor in median time on treatment between genotype trials and the immediate prior systemic standard treatment. Among 13 evaluable Low Grade Serous ovarian cancer patients treated on genotype-matched trials with MEK inhibitor-based targeted combinations, there were four partial responses. CONCLUSIONS Somatic molecular profiling is feasible and enables the identification of patients with rare gynecologic cancers who are candidates for genotype-matched clinical trials. Genotype-matched trials, predominantly MEK-based combinations in KRAS and/or NRAS mutant Low Grade Serous ovarian cancer patients, and genotype-unmatched trials, have shown potential clinical activity. Prospective trials with integrated genotyping are warranted to assess the clinical utility of next generation sequencing tests as a standard clinical application in rare malignancies.
Collapse
Affiliation(s)
- V Rodriguez-Freixinos
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - S Lheureux
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - V Mandilaras
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - B Clarke
- Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - N C Dhani
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - H Mackay
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - M O Butler
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - L Wang
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - L L Siu
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Cancer Genomics Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - S Kamel-Reid
- Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - T Stockley
- Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - P L Bedard
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Cancer Genomics Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - A M Oza
- Department of Medicine, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Colombo I, Kurnit KC, Westin SN, Oza AM. Moving From Mutation to Actionability. Am Soc Clin Oncol Educ Book 2018; 38:495-503. [PMID: 30231353 DOI: 10.1200/edbk_199665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The diffusion of high-throughput next-generation sequencing technologies has sustained massive parallel sequencing of tumor tissue providing a deep insight into tumor biology and advancement of personalized medicine. A substantial number of targeted agents have been investigated in gynecologic cancer and some have received U.S. Food and Drug Administration approval, like PARP inhibitors in ovarian cancer, bevacizumab in ovarian and cervical cancers, and pembrolizumab in microsatellite-unstable or mismatch repair-deficient endometrial cancer. To improve effectiveness of targeted therapy, identification of predictive biomarkers able to guide the selection of the correct drug for the correct patient is crucial. Different limitations must be addressed to favor a more rapid implementation of a genotyping approach in treatment selection, such as the possibility to easily assess tumor heterogeneity and clonal evolution along the disease trajectory and the need for innovative trial designs like adaptive or basket trials incorporating molecular features as selection criteria. A deep dive into the genomic features of exceptional responders may also favor better understanding of tumor biology, mechanism of action of a specific target agent, and identification or predictive biomarkers for subsequent tailored studies.
Collapse
Affiliation(s)
- Ilaria Colombo
- From the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; University of Toronto, Department of Medicine, Toronto, ON, Canada
| | - Katherine C Kurnit
- From the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; University of Toronto, Department of Medicine, Toronto, ON, Canada
| | - Shannon N Westin
- From the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; University of Toronto, Department of Medicine, Toronto, ON, Canada
| | - Amit M Oza
- From the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; University of Toronto, Department of Medicine, Toronto, ON, Canada
| |
Collapse
|
20
|
Bhangoo MS, Boasberg P, Mehta P, Elvin JA, Ali SM, Wu W, Klempner SJ. Tumor Mutational Burden Guides Therapy in a Treatment Refractory POLE-Mutant Uterine Carcinosarcoma. Oncologist 2018; 23:518-523. [PMID: 29386312 DOI: 10.1634/theoncologist.2017-0342] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/30/2017] [Indexed: 01/31/2023] Open
Abstract
Gynecologic carcinosarcomas, previously known as malignant mixed Müllerian tumors, are uncommon malignancies that demonstrate an aggressive biology and lack a standard therapeutic approach. Molecular analyses have revealed recurrent alterations in chromatin remodeling genes, but clinical support for therapeutic significance is lacking. We prospectively identified a patient with refractory uterine carcinosarcoma whose tumor was subject to molecular profiling at diagnosis and again at radiographic progression. Initial molecular testing did not assess tumor mutational burden, DNA polymerase ɛ (POLE), or microsatellite status. After the failure of several lines of chemotherapy, comprehensive genomic profiling of a repeat biopsy identified two missense mutations of the exonuclease domain of POLE (P286R and T323A). Tumor mutational burden was elevated (169 mutations per DNA megabase), consistent with an ultramutator phenotype. As seen in previously reported POLE-endometrioid cases, our patient harbored alterations in PIK3CA, ARID1A, and PTEN and was microsatellite stable, with appreciable tumor-infiltrating lymphocytes. She achieved an ongoing durable response with pembrolizumab. This is the first report of programmed cell death protein 1 response in uterine carcinosarcoma. KEY POINTS Uterine carcinosarcoma is an uncommon and aggressive histologic variant of endometrial carcinoma with a poor prognosis.Inactivating DNA polymerase ɛ (POLE) mutations have been associated with high tumor mutational burden (TMB) and response to immune checkpoint inhibition.To the authors' knowledge, this is the first report of response to immune checkpoint inhibitor therapy in a patient with uterine carcinosarcoma.This case further supports expanding genomic profiling to include assessment of tumor mutational burden across tumor types, given the potential for immune checkpoint inhibitor therapy in TMB-high tumors.
Collapse
Affiliation(s)
- Munveer S Bhangoo
- Division of Hematology Oncology, Scripps Clinic, La Jolla, California, USA
| | - Peter Boasberg
- The Angeles Clinic and Research Institute, Los Angeles, California, USA
| | - Pareen Mehta
- Department of Radiology, The Angeles Clinic and Research Institute, Los Angeles, California, USA
| | - Julia A Elvin
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
| | - Winnie Wu
- Department of Pathology, John Wayne Cancer Institute, Providence Saint John's Medical Center, Los Angeles, California, USA
| | - Samuel J Klempner
- The Angeles Clinic and Research Institute, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
21
|
Gaponova AV, Deneka AY, Beck TN, Liu H, Andrianov G, Nikonova AS, Nicolas E, Einarson MB, Golemis EA, Serebriiskii IG. Identification of evolutionarily conserved DNA damage response genes that alter sensitivity to cisplatin. Oncotarget 2017; 8:19156-19171. [PMID: 27863405 PMCID: PMC5386675 DOI: 10.18632/oncotarget.13353] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/27/2016] [Indexed: 01/08/2023] Open
Abstract
Ovarian, head and neck, and other cancers are commonly treated with cisplatin and other DNA damaging cytotoxic agents. Altered DNA damage response (DDR) contributes to resistance of these tumors to chemotherapies, some targeted therapies, and radiation. DDR involves multiple protein complexes and signaling pathways, some of which are evolutionarily ancient and involve protein orthologs conserved from yeast to humans. To identify new regulators of cisplatin-resistance in human tumors, we integrated high throughput and curated datasets describing yeast genes that regulate sensitivity to cisplatin and/or ionizing radiation. Next, we clustered highly validated genes based on chemogenomic profiling, and then mapped orthologs of these genes in expanded genomic networks for multiple metazoans, including humans. This approach identified an enriched candidate set of genes involved in the regulation of resistance to radiation and/or cisplatin in humans. Direct functional assessment of selected candidate genes using RNA interference confirmed their activity in influencing cisplatin resistance, degree of γH2AX focus formation and ATR phosphorylation, in ovarian and head and neck cancer cell lines, suggesting impaired DDR signaling as the driving mechanism. This work enlarges the set of genes that may contribute to chemotherapy resistance and provides a new contextual resource for interpreting next generation sequencing (NGS) genomic profiling of tumors.
Collapse
Affiliation(s)
- Anna V Gaponova
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan 420008, Russian Federation
| | - Alexander Y Deneka
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan 420008, Russian Federation
| | - Tim N Beck
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Biochemistry & Molecular Biology, Program in Molecular and Cell Biology and Genetics, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Hanqing Liu
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Pharmaceutics, Jiangsu University, School of Pharmacy, Jingkou District Zhenjiang, Jiangsu 212013, China
| | - Gregory Andrianov
- Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan 420008, Russian Federation
| | - Anna S Nikonova
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Emmanuelle Nicolas
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Margret B Einarson
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Erica A Golemis
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Ilya G Serebriiskii
- Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.,Department of Biochemistry and Biotechnology, Kazan Federal University, Kazan 420008, Russian Federation
| |
Collapse
|
22
|
Bezak B, Lehrke H, Elvin J, Gay L, Schembri-Wismayer D, Viozzi C. Comprehensive Genomic Profiling of Central Giant Cell Lesions Identifies Clinically Relevant Genomic Alterations. J Oral Maxillofac Surg 2017; 75:955-961. [DOI: 10.1016/j.joms.2016.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/13/2016] [Accepted: 10/21/2016] [Indexed: 12/27/2022]
|
23
|
Statz CM, Patterson SE, Mockus SM. Barriers preventing the adoption of comprehensive cancer genomic profiling in the clinic. Expert Rev Mol Diagn 2017; 17:549-555. [DOI: 10.1080/14737159.2017.1319280] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Foran DJ, Chen W, Chu H, Sadimin E, Loh D, Riedlinger G, Goodell LA, Ganesan S, Hirshfield K, Rodriguez L, DiPaola RS. Roadmap to a Comprehensive Clinical Data Warehouse for Precision Medicine Applications in Oncology. Cancer Inform 2017; 16:1176935117694349. [PMID: 28469389 PMCID: PMC5392017 DOI: 10.1177/1176935117694349] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/26/2017] [Indexed: 11/16/2022] Open
Abstract
Leading institutions throughout the country have established Precision Medicine programs to support personalized treatment of patients. A cornerstone for these programs is the establishment of enterprise-wide Clinical Data Warehouses. Working shoulder-to-shoulder, a team of physicians, systems biologists, engineers, and scientists at Rutgers Cancer Institute of New Jersey have designed, developed, and implemented the Warehouse with information originating from data sources, including Electronic Medical Records, Clinical Trial Management Systems, Tumor Registries, Biospecimen Repositories, Radiology and Pathology archives, and Next Generation Sequencing services. Innovative solutions were implemented to detect and extract unstructured clinical information that was embedded in paper/text documents, including synoptic pathology reports. Supporting important precision medicine use cases, the growing Warehouse enables physicians to systematically mine and review the molecular, genomic, image-based, and correlated clinical information of patient tumors individually or as part of large cohorts to identify changes and patterns that may influence treatment decisions and potential outcomes.
Collapse
Affiliation(s)
- David J Foran
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Wenjin Chen
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Huiqi Chu
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Evita Sadimin
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Doreen Loh
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Gregory Riedlinger
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Lauri A Goodell
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Kim Hirshfield
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Lorna Rodriguez
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | | |
Collapse
|
25
|
Genomic profiling of gynecologic cancers and implications for clinical practice. Curr Opin Obstet Gynecol 2016; 29:18-25. [PMID: 27984344 DOI: 10.1097/gco.0000000000000335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW This article summarizes advances in the application of next-generation sequencing (NGS) to the personalized treatment of gynecologic malignancies. RECENT FINDINGS Many recurrent genomic alterations (GA) in gynecologic malignancies have been identified by studies applying NGS to tumor tissue, which can provide insights into tumor biology, diagnostic or prognostic information, and potential targeted therapy options. NGS can be used to assay single genes, portions of multiple genes ("hot-spot" panels), or the complete coding sequence of a broad range of cancer-associated genes [i.e. comprehensive genomic profiling (CGP)]. CGP of a patient's tumor reveals to practitioners clinically relevant GA (CRGA) and associated biomarker-matched treatments, with a goal of improving therapeutic response while limiting cumulative chemotherapeutic toxicities. Although the use of precision medicine for gynecologic cancers holds much promise, the data detailing impact on survival and quality of life is still accumulating, lagging behind other areas of oncology. Enrolling gynecologic oncology patients in genotype-matched trials remains challenging and highlights the need for more molecular-based basket trials for reproductive tract malignancies. SUMMARY Identification of molecular subsets with distinct clinical attributes, prognostic significance, and targeted therapy directed options is now feasible in clinical gynecologic oncology practice.
Collapse
|
26
|
Conley BA, Sorg BS, Tricoli JV. Implications and opportunities of precision medicine in rare malignancies. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1214071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Barbara A. Conley
- Cancer Diagnosis Program, Division of Cancer Diagnosis and Treatment, National Cancer Institute, Bethesda, MD, USA
| | - Brian S. Sorg
- Cancer Diagnosis Program, Division of Cancer Diagnosis and Treatment, National Cancer Institute, Bethesda, MD, USA
| | - James V. Tricoli
- Cancer Diagnosis Program, Division of Cancer Diagnosis and Treatment, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
27
|
Ganesan S, Rodriguez-Rodriguez L, DiPaola RS. Precision Medicine: Implications for Science and Practice. J Am Coll Surg 2016; 223:433-439.e1. [PMID: 27321387 DOI: 10.1016/j.jamcollsurg.2016.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Shridar Ganesan
- Precision Medicine Initiative, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | | | - Robert S DiPaola
- Office of the Dean, University of Kentucky, College of Medicine, Lexington, KY
| |
Collapse
|