1
|
Wang F, Chen G, Ruan W, Wang B, Zhu Z, Hu W, Chen S, Zang L. Application of tissue pneumoperitoneum technique around lymph nodes in thoracoscopic lung cancer resection. Front Oncol 2024; 14:1443088. [PMID: 39252943 PMCID: PMC11381222 DOI: 10.3389/fonc.2024.1443088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Background Thoracoscopic surgery is a primary treatment for lung cancer, with lobectomy and mediastinal lymph node dissection being the predominant surgical approaches for invasive lung cancer. While many thoracic surgeons can proficiently perform lobectomy, thorough and standardized lymph node dissection remains challenging. This study aimed to explore a safer and more efficient surgical method for mediastinal lymph node dissection in lung cancer. Methods A prospective randomized controlled study was conducted, involving 100 patients with right lung cancer who were admitted to our hospital from January 2021 to April 2024 and met the inclusion criteria. These patients were randomly divided into an observation group (tissue pneumoperitoneum technique around lymph nodes group) and a control group (conventional surgery group). Thoracoscopic lobectomy and mediastinal lymph node dissection were performed. Intraoperative and postoperative related indicators were observed to validate the effectiveness and safety of the tissue pneumoperitoneum technique around lymph nodes. Results The observation group showed a significantly shorter lymph node dissection surgery time compared to the control group, with a statistically significant difference (p < 0.05). The number of lymph nodes dissected in the observation group was significantly higher than that in the control group, with a statistically significant difference (p < 0.05). Although the observation group had slightly more mediastinal lymph node stations dissected than the control group, the difference was not statistically significant (p > 0.05). The total drainage volume within three days postoperatively was comparable between the two groups, with no statistically significant difference (p > 0.05). The observation group had shorter chest tube indwelling time and postoperative hospital stay than the control group, with statistically significant differences (p < 0.05). The incidence of surgical complications was similar between the two groups, and there were no perioperative deaths. Conclusion The tissue pneumoperitoneum technique around lymph nodes is a more efficient method for mediastinal lymph node dissection in lung cancer, demonstrating safety and feasibility, and is worthy of promotion.
Collapse
Affiliation(s)
- Fangqing Wang
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Gang Chen
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Weimin Ruan
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Binkui Wang
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Zhaowang Zhu
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Weijian Hu
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Sheng Chen
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| | - Lin Zang
- Department of Cardiothoracic Surgery, The People's Hospital of Tongling, Tongling, Anhui, China
| |
Collapse
|
2
|
Lefkovits YR, Heriot N, Sporik A, Perera S, Friedlander M, Dixon C, Cohen PA, Lee YC, Hyde S, Richardson G, Webb P, Rome R, King M, Zalcberg J, Schofield P. Incorporating patient-reported outcome measures (PROMs) into a clinical quality registry (CQR) for ovarian cancer: considerations and challenges. BMC Health Serv Res 2024; 24:778. [PMID: 38978033 PMCID: PMC11232149 DOI: 10.1186/s12913-024-11042-8] [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: 10/12/2022] [Accepted: 04/24/2024] [Indexed: 07/10/2024] Open
Abstract
As medical treatment increasingly focuses on improving health-related quality of life, patient-reported outcome measures (PROMs) are an essential component of clinical research. The National Gynae-Oncology Registry (NGOR) is an Australian clinical quality registry. A suitable PROM was required for the NGOR ovarian cancer module to complement clinical outcomes and provide insights into outcomes important to patients. Our narrative review aimed to identify existing ovarian cancer-specific PROMs and ascertain which tool would be most appropriate for implementation into the NGOR ovarian cancer module.A literature review of Cochrane Library, Embase, MEDLINE and PubMed databases was performed to identify existing ovarian cancer-specific PROM tools. A steering committee was convened to (1) determine the purpose of, and criteria for our required PROM; and (2) to review the available tools against the criteria and recommend the most appropriate one for implementation within the NGOR.The literature review yielded five tools: MOST, EORTC QLQ-OV28, FACIT-O, NFOSI-18 and QOL-OVCA. All were developed and validated for use in clinical trials, but none had been validated for use in clinical quality registry. Our expert steering committee pre-determined purpose of a PROM tool for use within the NGOR was to enable cross-service comparison and benchmarking to drive quality improvements. They identified that while there was no ideal, pre-existing, ovarian cancer-specific PROM tool for implementation into the NGOR, on the basis of its psychometric properties, its available translations, its length and its ability to be adapted, the EORTC tool is most fit-for-purpose for integration into the NGOR.This process enabled identification of the tool most appropriate to provide insights into how ovarian cancer treatments impact patients' quality of life and permit benchmarking across health services.
Collapse
Affiliation(s)
- Yael R Lefkovits
- Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia.
- School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, Melbourne, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, Victoria, Australia.
| | - Natalie Heriot
- School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, Melbourne, Australia
| | - Alice Sporik
- School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, Melbourne, Australia
| | - Sharnel Perera
- School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, Melbourne, Australia
| | - Michael Friedlander
- Department of Medical Oncology, University of New South Wales, Sydney, Australia
| | - Cyril Dixon
- Ovarian Cancer Australia, Melbourne, Victoria, Australia
| | - Paul A Cohen
- Discipline of Obstetrics and Gynaecology, Medical School, University of Western Australia, Crawley, Western Australia, Australia
- Department of Gynaecological Oncology, St John of God Subiaco Hospital, Subiaco, Western Australia, Australia
- Institute of Health Research, University of Notre Dame, Fremantle, Western Australia, Australia
| | - Yeh Chen Lee
- Medical Oncology Department, Prince of Wales Hospital, Randwick, New South Wales, Australia
- Faculty of Medicine and Health, University of New South Wales, Randwick, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Simon Hyde
- Department of Gynaecological Oncology, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Gary Richardson
- Szalmuk Family Department of Medical Oncology, Cabrini Research, Malvern, Victoria, Australia
| | - Penelope Webb
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Robert Rome
- Clinical Institute of Obstetrics and Gynaecology, Epworth HealthCare, East Melbourne, Victoria, Australia
| | - Madeleine King
- School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - John Zalcberg
- School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, Melbourne, Australia
| | - Penelope Schofield
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, Victoria, Australia
- Behavioural Sciences Unit, Health Services Research and Implementation Sciences, Peter MacCallum Centre, Melbourne, Victoria, Australia
- Department of Psychology and Iverson Health Innovation Research Institute, Swinburne University, Melbourne, Victoria, Australia
- Department of Behavioural Sciences in Cancer, Swinburne University, Hawthorne, Victoria, Australia
| |
Collapse
|
3
|
Miller RW, Hutchcraft ML, Weiss HL, Wu J, Wang C, Liu J, Jayswal R, Buchanan M, Anderson A, Allison DB, El Khouli RH, Patel RA, Villano JL, Arnold SM, Kolesar JM. Molecular Tumor Board-Assisted Care in an Advanced Cancer Population: Results of a Phase II Clinical Trial. JCO Precis Oncol 2022; 6:e2100524. [PMID: 36103643 PMCID: PMC9489195 DOI: 10.1200/po.21.00524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/04/2022] [Accepted: 08/10/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Multidisciplinary molecular tumor boards (MTBs) interpret next-generation sequencing reports and help oncologists determine best therapeutic options; however, there is a paucity of data regarding their clinical utility. The purpose of this study was to determine if MTB-directed therapy improves progression-free survival (PFS) over immediately prior therapy in patients with advanced cancer. METHODS This single-arm, prospective phase II clinical trial enrolled patients with advanced cancer with an actionable mutation who received MTB-recommended targeted therapy between January 1, 2017, and October 31, 2020. MTB-recommended both on-label (level 1 evidence) and off-label (evidence levels 2 and 3) therapies. Of the 93 enrolled patients, 43 were treated frontline and 50 received second-line or greater-line therapy. The primary outcome was the probability of patients treated with second-line or greater-line MTB-directed therapy who achieved a PFS ratio ≥ 1.3 (PFS on MTB-directed therapy divided by PFS on the patient's immediately prior therapy). Secondary outcomes included PFS for patients treated frontline and overall survival and adverse effects for the entire study population. RESULTS The most common disease sites were lung (35 of 93, 38%), gynecologic (17 of 93, 18%), GI (16 of 93, 17%), and head and neck (7 of 93, 8%). The Kaplan-Meier estimate of the probability of PFS ratio ≥ 1.3 was 0.59 (95% CI, 0.47 to 0.75) for patients treated with second-line or greater-line MTB-directed therapy. The median PFS was 449 (range 42-1,125) days for patients treated frontline. The median overall survival was 768 (range 22-1,240) days. There were four nontreatment-related deaths. CONCLUSION When treated with MTB-directed therapy, most patients experienced improved PFS compared with immediately prior treatment. MTB-directed targeted therapy may be a strategy to improve outcomes for patients with advanced cancer.
Collapse
Affiliation(s)
- Rachel W. Miller
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Megan L. Hutchcraft
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Heidi L. Weiss
- Shared Resource Facility, University of Kentucky Markey Cancer Center, Lexington, KY
- Division of Cancer Biostatistics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| | - Jianrong Wu
- Division of Cancer Biostatistics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| | - Chi Wang
- Shared Resource Facility, University of Kentucky Markey Cancer Center, Lexington, KY
- Division of Cancer Biostatistics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| | - Jinpeng Liu
- Shared Resource Facility, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Rani Jayswal
- Shared Resource Facility, University of Kentucky Markey Cancer Center, Lexington, KY
- Division of Cancer Biostatistics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| | - Mikayla Buchanan
- Division of Precision Medicine, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Abigail Anderson
- Division of Precision Medicine, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Derek B. Allison
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY
| | | | - Reema A. Patel
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky Markey Cancer Center, Lexington, KY
| | - John L. Villano
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Susanne M. Arnold
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky Markey Cancer Center, Lexington, KY
| | - Jill M. Kolesar
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky Markey Cancer Center, Lexington, KY
- Division of Precision Medicine, University of Kentucky Markey Cancer Center, Lexington, KY
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY
| |
Collapse
|
4
|
Farhangfar CJ, Scarola GT, Morris VA, Farhangfar F, Dumas K, Symanowski J, Hwang JJ, Mileham KF, Carrizosa DR, Naumann RW, Livasy C, Kim ES, Raghavan D. Impact of a Clinical Genomics Program on Trial Accrual for Targeted Treatments: Practical Approach Overcoming Barriers to Accrual for Underserved Patients. JCO Clin Cancer Inform 2022; 6:e2200011. [PMID: 35839431 DOI: 10.1200/cci.22.00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Clinical trials of novel and targeted agents increasingly require biomarkers for eligibility. Precision oncology continues to evolve, but challenges hamper broad use of molecular profiling (MP) that could increase the number of patients benefiting from targeted therapy. We implemented an integrated clinical genomics program (CGP), including a virtual Molecular Tumor Board (MTB), and examined its impact on MP use and impact on clinical trial accrual in a multisite regional-based cancer system with an emphasis on effects for isolated clinicians. METHODS We assessed MP and MTB use from 2010 to 2020 by practice location, physician experience, and patient characteristics. Use of MTB-recommended treatments was assessed. Clinical trial enrollment was evaluated for patients with MP versus MP and MTB review. RESULTS After CGP implementation, the number of physicians using MP and the number of MP tests increased ≥ 10-fold. The proportion of Hispanic patients with MP was the same as that in the system (both 2%) with marginal differences observed in the proportion of African Americans tested compared with the system population (16% v 19%). Physicians followed MTB treatment recommendations in 74% of cases. Rapid clinical decline was the most common reason why physicians did not follow MTB recommendations. Clinical trial accrual was 15% (669 of 4,459) for patients with MP alone and 28% (94 of 334) with both MP and MTB review. Clinical trial availability and patient out-of-pocket costs affected MP use. CONCLUSION Integrating CGP into clinical workflow with decision support tools, trial matching, and management of patient costs led to increased use of MP by physicians with all levels of experience, enhanced clinical trial accrual, and has the potential to reduce disparities in MP.
Collapse
Affiliation(s)
- Carol J Farhangfar
- Department of Translational Research, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Gregory T Scarola
- Department of Translational Research, Levine Cancer Institute, Atrium Health, Charlotte, NC.,Department of Surgery, Atrium Health, Charlotte, NC
| | - Victoria A Morris
- Department of Information and Analytics Services, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Farhang Farhangfar
- Department of Biospecimen Repository, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Kathryn Dumas
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC.,Johns Hopkins Medical Institution, Baltimore, MD
| | - James Symanowski
- Department of Biostatistics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Jimmy J Hwang
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Kathryn F Mileham
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Daniel R Carrizosa
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - R Wendel Naumann
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Chad Livasy
- Department of Pathology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Edward S Kim
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC.,City of Hope, National Medical Center, Los Angeles, CA
| | - Derek Raghavan
- Department of Solid Tumor Oncology, Levine Cancer Institute, Atrium Health, Charlotte, NC
| |
Collapse
|
5
|
Fischer CG, Pallavajjala A, Jiang L, Anagnostou V, Tao J, Adams E, Eshleman JR, Gocke CD, Lin MT, Platz EA, Xian RR. Artificial intelligence-assisted serial analysis of clinical cancer genomics data identifies changing treatment recommendations and therapeutic targets. Clin Cancer Res 2022; 28:2361-2372. [PMID: 35312750 DOI: 10.1158/1078-0432.ccr-21-4061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/15/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Given the pace of predictive biomarker and targeted therapy development, it is unknown if repeat annotation of the same next-generation sequencing data can identify additional clinically actionable targets that could be therapeutically leveraged. In this study, we sought to determine the predictive yield of serial re-analysis of clinical tumor sequencing data. EXPERIMENTAL DESIGN Using artificial intelligence (AI)-assisted variant annotation, we retrospectively re-analyzed sequencing data from 2,219 cancer patients from a single academic medical center at 3-month intervals totaling 9 months in 2020. The yield of serial re-analysis was assessed by the proportion of patients with improved strength of therapeutic recommendations. RESULTS 1,775 patients (80%) had {greater than or equal to}1 potentially clinically actionable mutation at baseline, including 243 (11%) patients who had an alteration targeted by an FDA-approved drug for their cancer type. By month nine, the latter increased to 458 (21%) patients mainly due to a single pan-cancer agent directed against tumors with high tumor mutation burden. Within this timeframe, 67 new therapies became available and 45 were no longer available. Variant pathogenicity classifications also changed leading to changes in treatment recommendations for 124 patients (6%). CONCLUSIONS Serial re-annotation of tumor sequencing data improved the strength of treatment recommendations (based on level of evidence) in a mixed cancer cohort and showed substantial changes in available therapies and variant classifications. These results suggest a role for repeat analysis of tumor sequencing data in clinical practice, which can be streamlined with AI support.
Collapse
Affiliation(s)
| | | | - LiQun Jiang
- Johns Hopkins Medical Institutions, United States
| | - Valsamo Anagnostou
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jessica Tao
- Johns Hopkins Medical Institutions, United States
| | - Emily Adams
- Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | | | | | - Ming-Tseh Lin
- The Johns Hopkins School of Medicine, Baltimore, United States
| | - Elizabeth A Platz
- Johns Hopkins Bloomberg School of Public Health, Baltimore, United States
| | - Rena R Xian
- Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
6
|
Green MF, Bell JL, Hubbard CB, McCall SJ, McKinney MS, Riedel JE, Menendez CS, Abbruzzese JL, Strickler JH, Datto MB. Implementation of a Molecular Tumor Registry to Support the Adoption of Precision Oncology Within an Academic Medical Center: The Duke University Experience. JCO Precis Oncol 2021; 5:PO.21.00030. [PMID: 34568718 PMCID: PMC8457820 DOI: 10.1200/po.21.00030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 07/14/2021] [Accepted: 08/04/2021] [Indexed: 12/27/2022] Open
Abstract
Comprehensive genomic profiling to inform targeted therapy selection is a central part of oncology care. However, the volume and complexity of alterations uncovered through genomic profiling make it difficult for oncologists to choose the most appropriate therapy for their patients. Here, we present a solution to this problem, The Molecular Registry of Tumors (MRT) and our Molecular Tumor Board (MTB). PATIENTS AND METHODS MRT is an internally developed system that aggregates and normalizes genomic profiling results from multiple sources. MRT serves as the foundation for our MTB, a team that reviews genomic results for all Duke University Health System cancer patients, provides notifications for targeted therapies, matches patients to biomarker-driven trials, and monitors the molecular landscape of tumors at our institution. RESULTS Among 215 patients reviewed by our MTB over a 6-month period, we identified 176 alterations associated with therapeutic sensitivity, 15 resistance alterations, and 51 alterations with potential germline implications. Of reviewed patients, 17% were subsequently treated with a targeted therapy. For 12 molecular therapies approved during the course of this work, we identified between two and 71 patients who could qualify for treatment based on retrospective MRT data. An analysis of 14 biomarker-driven clinical trials found that MRT successfully identified 42% of patients who ultimately enrolled. Finally, an analysis of 4,130 comprehensive genomic profiles from 3,771 patients revealed that the frequency of clinically significant therapeutic alterations varied from approximately 20% to 70% depending on the tumor type and sequencing test used. CONCLUSION With robust informatics tools, such as MRT, and the right MTB structure, a precision cancer medicine program can be developed, which provides great benefit to providers and patients with cancer.
Collapse
Affiliation(s)
- Michelle F Green
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Jonathan L Bell
- Department of Pathology, Duke University Medical Center, Durham, NC
| | | | - Shannon J McCall
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Matthew S McKinney
- Division of Hematologic Malignancies, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Jinny E Riedel
- Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | - Carolyn S Menendez
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Department of Surgery, Duke University Medical Center, Durham, NC
| | - James L Abbruzzese
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - John H Strickler
- Duke Cancer Institute, Duke University Medical Center, Durham, NC.,Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Michael B Datto
- Department of Pathology, Duke University Medical Center, Durham, NC
| |
Collapse
|
7
|
Barnell EK, Newcomer KF, Skidmore ZL, Krysiak K, Anderson SR, Wartman LD, Oh ST, Welch JS, Stockerl-Goldstein KE, Vij R, Cashen AF, Pusic I, Westervelt P, Abboud CN, Ghobadi A, Uy GL, Schroeder MA, Dipersio JF, Politi MC, Spencer DH, Duncavage EJ, Ley TJ, Griffith M, Jacoby MA, Griffith OL. Impact of a 40-Gene Targeted Panel Test on Physician Decision Making for Patients With Acute Myeloid Leukemia. JCO Precis Oncol 2021; 5:PO.20.00182. [PMID: 34036230 PMCID: PMC8140802 DOI: 10.1200/po.20.00182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Physicians treating hematologic malignancies increasingly order targeted sequencing panels to interrogate recurrently mutated genes. The precise impact of these panels on clinical decision making is not well understood. METHODS Here, we report our institutional experience with a targeted 40-gene panel (MyeloSeq) that is used to generate a report for both genetic variants and variant allele frequencies for the treating physician (the limit of mutation detection is approximately one AML cell in 50). RESULTS In total, 346 sequencing reports were generated for 325 patients with suspected hematologic malignancies over an 8-month period (August 2018 to April 2019). To determine the influence of genomic data on clinical care for patients with acute myeloid leukemia (AML), we analyzed 122 consecutive reports from 109 patients diagnosed with AML and surveyed the treating physicians with a standardized questionnaire. The panel was ordered most commonly at diagnosis (61.5%), but was also used to assess response to therapy (22.9%) and to detect suspected relapse (15.6%). The panel was ordered at multiple timepoints during the disease course for 11% of patients. Physicians self-reported that 50 of 114 sequencing reports (44%) influenced clinical care decisions in 44 individual patients. Influences were often nuanced and extended beyond identifying actionable genetic variants with US Food and Drug Administration-approved drugs. CONCLUSION This study provides insights into how physicians are currently using multigene panels capable of detecting relatively rare AML cells. The most influential way to integrate these tools into clinical practice will be to perform prospective clinical trials that assess patient outcomes in response to genomically driven interventions.
Collapse
Affiliation(s)
- Erica K Barnell
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO
| | - Kenneth F Newcomer
- Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Zachary L Skidmore
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO
| | - Kilannin Krysiak
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO
| | - Sydney R Anderson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO
| | - Lukas D Wartman
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Stephen T Oh
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO.,Department of Medicine, Division of Hematology, Washington University School of Medicine, St Louis, MO
| | - John S Welch
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Keith E Stockerl-Goldstein
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Ravi Vij
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Amanda F Cashen
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Iskra Pusic
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Peter Westervelt
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Camille N Abboud
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Armin Ghobadi
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Geoffrey L Uy
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO
| | - Mark A Schroeder
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - John F Dipersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Mary C Politi
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St Louis, MO
| | - David H Spencer
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO
| | - Timothy J Ley
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Malachi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO.,Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO.,Department of Genetics, Washington University School of Medicine, St Louis, MO
| | - Meagan A Jacoby
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Obi L Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO.,Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO.,Siteman Cancer Center, Washington University School of Medicine, St Louis, MO.,Department of Genetics, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
8
|
Xu L, Su H, She Y, Dai C, Zhao M, Gao J, Xie H, Ren Y, Xie D, Chen C. Which N Descriptor Is More Predictive of Prognosis in Resected Non-small Cell Lung Cancer: The Number of Involved Nodal Stations or the Location-Based Pathological N Stage? Chest 2020; 159:2458-2469. [PMID: 33352193 DOI: 10.1016/j.chest.2020.12.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The eighth edition of nodal classification for non-small cell lung cancer (NSCLC) is defined only by the anatomical location of metastatic lymph nodes. RESEARCH QUESTION We sought to evaluate the prognostic significance and discriminatory capability of the number of involved nodal stations (nS) in a large Chinese cohort. STUDY DESIGN AND METHODS A total of 4,011 patients with NSCLC undergoing surgical resection between 2009 and 2013 were identified. The optimal cutoff values for nS classification were determined with X-tile software. Kaplan-Meier and multivariate Cox analysis were used to examine the prognostic performance of nS classification in comparison with location-based N classification. A decision curve analysis was performed to evaluate the standardized net benefit of nS classification in predicting prognosis. RESULTS All the patients were classified into four prognostically different subgroups according to the number of involved nodal stations: (1) nS0 (none positive), (2) nS1 (one involved station), (3) nS2 (two involved stations), and (4) nS ≥ 3 (three or more involved stations). The prognoses among all the neighboring categories of nS classification were statistically significantly different in terms of disease-free survival and overall survival. The multivariate Cox analysis demonstrated that nS was an independent prognostic factor of disease-free survival and overall survival. Patients with N1 or N2 stage disease could be divided into three prognostically different subgroups according to nS classification. However, the prognosis was similar between the N1 and N2 subgroups when patients were staged in the same nS category. The decision curve analysis showed that nS classification tended to have a higher predictive capability than location-based N classification. INTERPRETATION The nS classification could be used to provide a more accurate prognosis for patients with resected NSCLC. The nS is worth taking into consideration when defining nodal category in the forthcoming ninth edition of the staging system.
Collapse
Affiliation(s)
- Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mengmeng Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Sun W, Su H, Liu J, Zhang L, Li M, Xie H, Xu L, Zhao S, She Y, Tang H, Wu C, Ke H, Chen C. Impact of histological components on selecting limited lymphadenectomy for lung adenocarcinoma ≤ 2 cm. Lung Cancer 2020; 150:36-43. [PMID: 33059150 DOI: 10.1016/j.lungcan.2020.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES There is still some dispute regarding the performance of limited mediastinal lymphadenectomy (LML) even for lung adenocarcinoma ≤ 2 cm. We aimed to recognize the potential candidates who can benefit from LML based on the percentage of histological components (PHC). METHODS We analyzed 1160 surgical patients with invasive lung adenocarcinoma ≤ 2 cm from seven institutions between January 2012 and December 2015. All histological subtypes were listed in 5% increments by pathological slices. To test the accuracy of frozen section in judging PHC, frozen section slides from 140 cases were reviewed by three pathologists. RESULTS There were 882 patients with systematic mediastinal lymphadenectomy (SML) and 278 with LML. Multivariable analysis indicated that, the total percentage of micropapillary and solid components (PHCMIP+S) > 5 % was the independent predictor of N2 metastasis (P < 0.001). Overall, recurrence-free survival (RFS) and overall survival (OS) favored SML compared with LML, but the subgroup analysis revealed LML and SML had similar prognosis in the group of PHCMIP+S ≤ 5 %. Moreover, multivariable Cox analysis showed LML (vs. SML) was independently associated with worse prognosis for patients with PHCMIP+S > 5 % (RFS, HR = 2.143, P < 0.001; OS, HR=1.963, P < 0.001), but not for those with PHCMIP+S ≤ 5 % (RFS, P = 0.398; OS, P = 0.298). The sensitivity and specificity of frozen section to intraoperatively identify PHCMIP+S ≤ 5 % were 97.6 % and 84.2 %, respectively. CONCLUSIONS PHCMIP+S showed the predictive value for N2 metastasis and procedure-specific outcome (LML vs. SML). It may serve as a feasible indicator for identifying proper candidates of LML by using intraoperative frozen section.
Collapse
Affiliation(s)
- Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hang Su
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jinshi Liu
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Lei Zhang
- Department of Thoracic Surgery, The First People's Hospital of Changzhou, Changzhou, People's Republic of China
| | - Ming Li
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Nanjing, People's Republic of China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, TongjiUniversity School of Medicine, Shanghai, People's Republic of China
| | - Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shengnan Zhao
- Department of Pathology, Shanghai Pulmonary Hospital, TongjiUniversity School of Medicine, Shanghai, People's Republic of China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, TongjiUniversity School of Medicine, Shanghai, People's Republic of China
| | - Honggang Ke
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
10
|
Pan L, Mo R, Zhu L, Yu W, Lv W, Hu J. Time trend of mediastinal lymph node dissection in stage IA non-small cell lung cancer patient who undergo lobectomy: a retrospective study of surveillance, epidemiology, and end results (SEER) database. J Cardiothorac Surg 2020; 15:207. [PMID: 32738925 PMCID: PMC7395351 DOI: 10.1186/s13019-020-01215-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/07/2020] [Indexed: 12/25/2022] Open
Abstract
Background Although lobectomy with mediastinal lymph node dissection (MLND) is the first option for early-stage non-small cell lung cancer (NSCLC) patients, the time trends of MLND in stage IA NSCLC patients who undergo a lobectomy are not clear still. Methods We included stage IA NSCLC patients who underwent lobectomy or lobectomy with MLND between 2003 and 2013 in the SEER database. The time trend of MLND was compared among patients who underwent a lobectomy. Results For stage T1a patients, the lobectomy group and lobectomy with MLND group had no differences in postoperative overall survival (OS) (P = 0.34) or lung-cancer specific survival (LCSS) (P = 0.18) between 2003 and 2013. For stage T1b patients, the OS (P = 0.01) and LCSS (P = 0.01) were different between the lobectomy group and the lobectomy with MLND group in the period from 2003 to 2009; however, only OS (P = 0.04), not LCSS (P = 0.14), was different between the lobectomy group and the lobectomy with MLND group between 2009 and 2013. For T1c patients, the OS (P = 0.01) and LCSS (P = 0.02) were different between the two groups between 2003 and 2009 but not between 2009 and 2013 (P = 0.60; P = 0.39). From the Cox regression analysis, we found that the factors affecting OS/LCSS in T1b and T1c patients were age, sex, year of diagnosis, histology, and grade, in which year of diagnosis was the obvious factor (HR = 0.79, CI = 0.71–0.87; HR = 0.73, CI = 0.64–0.84). Conclusions There was a time trend in prognosis differences between the lobectomy group and lobectomy with MLND group for T1b and T1c stage NSCLC patients.
Collapse
Affiliation(s)
- Liang Pan
- Department of thoracic surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Ran Mo
- Department of thoracic surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Linhai Zhu
- Department of thoracic surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Wenfeng Yu
- Department of thoracic surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Wang Lv
- Department of thoracic surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China
| | - Jian Hu
- Department of thoracic surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
11
|
Williams D, Vilar E, Shakrukh Hashmi S, Choates M, Noblin S, Mork M. Somatic mismatch repair testing in evaluation of Lynch syndrome: The gap between preferred and current practices. J Genet Couns 2020; 29:728-736. [PMID: 31896172 DOI: 10.1002/jgc4.1198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 11/05/2022]
Abstract
Lynch syndrome (LS) is a hereditary cancer predisposition syndrome primarily defined by increased risk for colorectal and uterine cancers. Individuals with germline pathogenic variants in the mismatch repair (MMR) genes (MLH1, MSH2/EPCAM, MSH6, and PMS2) are diagnosed with LS and recommended high-risk screening protocols to increase prevention and early detection of LS-related cancers. Tumor testing can help identify those at high risk for LS, but sometimes creates uncertainty with discordant screening and germline results, or unexplained mismatch repair deficiency (UMMRD). Somatic testing for MMR genes may help resolve UMMRD, potentially clarifying LS status and modifying cancer surveillance. However, guidelines for such testing are currently limited. This survey of cancer genetic counselors (GCs) aimed to examine current versus preferred ordering practices and interpretation of somatic MMR testing results in LS evaluation. Two hundred eligible GCs practicing in the United States and Canada were recruited from the National Society of Genetic Counselors. Participants answered questions regarding ordering practices, barriers to somatic MMR testing, theoretical scenarios, and desire for further guidelines. Statistical analysis was performed using chi-square, Fisher's exact, and Wilcoxon rank-sum tests, while themes were identified from free-text responses. Most respondents did not include somatic MMR testing in the LS work-up, despite three-quarters reporting they were 'somewhat comfortable' or 'extremely comfortable' with interpreting these results. Approximately half of participants indicated interest in ordering concurrent somatic MMR and germline testing for each of the four theoretical scenarios. Over three-quarters of individuals reported barriers to ordering somatic MMR testing, with cost and coordinating tissue samples most commonly cited. The frequently reported laboratory- and insurance-related barriers may contribute to the gap between preferred and current ordering practices for somatic MMR testing. Nearly all respondents endorsed additional guidelines for this testing, which could reduce barriers and inform screening recommendations for patients with UMMRD and their family members.
Collapse
Affiliation(s)
- Danielle Williams
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Cancer Genetics, The Center for Cancer Prevention and Treatment, St. Joseph Health, Orange, California
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - S Shakrukh Hashmi
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, Texas
| | - Meagan Choates
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Obstetrics, Gynecology, and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, Texas
| | - Sarah Noblin
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Invitae Genetics, San Francisco, California
| | - Maureen Mork
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.,Department of Clinical Cancer Prevention, Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
12
|
Pishvaian MJ, Blais EM, Bender RJ, Rao S, Boca SM, Chung V, Hendifar AE, Mikhail S, Sohal DPS, Pohlmann PR, Moore KN, He K, Monk BJ, Coleman RL, Herzog TJ, Halverson DD, DeArbeloa P, Petricoin EF, Madhavan S. A virtual molecular tumor board to improve efficiency and scalability of delivering precision oncology to physicians and their patients. JAMIA Open 2019; 2:505-515. [PMID: 32025647 PMCID: PMC6994017 DOI: 10.1093/jamiaopen/ooz045] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/28/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Scalable informatics solutions that provide molecularly tailored treatment recommendations to clinicians are needed to streamline precision oncology in care settings. MATERIALS AND METHODS We developed a cloud-based virtual molecular tumor board (VMTB) platform that included a knowledgebase, scoring model, rules engine, an asynchronous virtual chat room and a reporting tool that generated a treatment plan for each of the 1725 patients based on their molecular profile, previous treatment history, structured trial eligibility criteria, clinically relevant cancer gene-variant assertions, biomarker-treatment associations, and current treatment guidelines. The VMTB systematically allows clinician users to combine expert-curated data and structured data from clinical charts along with molecular testing data to develop consensus on treatments, especially those that require off-label and clinical trial considerations. RESULTS The VMTB was used as part of the cancer care process for a focused subset of 1725 patients referred by advocacy organizations wherein resultant personalized reports were successfully delivered to treating oncologists. Median turnaround time from data receipt to report delivery decreased from 14 days to 4 days over 4 years while the volume of cases increased nearly 2-fold each year. Using a novel scoring model for ranking therapy options, oncologists chose to implement the VMTB-derived therapies over others, except when pursuing immunotherapy options without molecular support. DISCUSSION VMTBs will play an increasingly critical role in precision oncology as the compendium of biomarkers and associated therapy options available to a patient continues to expand. CONCLUSION Further development of such clinical augmentation tools that systematically combine patient-derived molecular data, real-world evidence from electronic health records and expert curated knowledgebases on biomarkers with computational tools for ranking best treatments can support care pathways at point of care.
Collapse
Affiliation(s)
- Michael J Pishvaian
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
- Perthera, Inc, McLean, Virginia, USA
| | | | | | - Shruti Rao
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington DC, USA
| | - Simina M Boca
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington DC, USA
| | | | | | - Sam Mikhail
- Mark H. Zangmeister Cancer Center, Columbus, Ohio, USA
| | - Davendra P S Sohal
- Case Comprehensive Cancer Center, University Hospitals Seidman Cancer Center, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Paula R Pohlmann
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Kathleen N Moore
- Stephenson Oklahoma Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Kai He
- Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Bradley J Monk
- Arizona Oncology, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Robert L Coleman
- University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Thomas J Herzog
- University of Cincinnati Cancer Institute, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | - Emanuel F Petricoin
- Perthera, Inc, McLean, Virginia, USA
- George Mason University, Fairfax, Virginia, USA
| | - Subha Madhavan
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
13
|
Wang W, Chen D, Xi K, Chen Y, Zhang X, Wen Y, Huang Z, Yu X, Wang G, Zhang R, Zhang L. Impact of Different Types of Lymphadenectomy Combined With Different Extents of Tumor Resection on Survival Outcomes of Stage I Non-small-cell Lung Cancer: A Large-Cohort Real-World Study. Front Oncol 2019; 9:642. [PMID: 31396479 PMCID: PMC6668052 DOI: 10.3389/fonc.2019.00642] [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: 04/09/2019] [Accepted: 07/01/2019] [Indexed: 12/25/2022] Open
Abstract
Background: To investigate the prognostic impact of different types of lymphadenectomy with different extents of tumor resection on the outcomes of stage I non-small-cell lung cancer (NSCLC). Methods: Patients were classified into lobectomy and sublobectomy groups, and then each group was subdivided according to the types of lymphadenectomy. The end points of the study were overall survival (OS) and disease-free survival (DFS). Propensity score matched (PSM) comparative analysis and univariate and multivariate Cox regression analyses were performed. Result: A total of 1,336 patients were included in the current study. Lobectomy was associated with better OS and DFS. In the lobectomy group, lobectomy with bilateral mediastinal lymphadenectomy (BML) was associated with better OS than lobectomy with systematic nodal dissection (SND) or lobe-specific systematic node dissection (L-SND). Lobectomy with SND or L-SND was associated with better OS than lobectomy with systematic nodal sampling (SNS) or selected lymph node biopsy (SLNB). Additionally, lobectomy with BML or SND was associated with better DFS than lobectomy with L-SND or SNS or SLNB. After PSM, compared with lobectomy with SNS or SLNB, lobectomy with SND resulted in more favorable OS and DFS. There was no survival difference between different types of lymphadenectomy for patients who underwent sublobectomy. A multivariable analysis revealed independent associations of lobectomy with BML or SND with better OS and DFS compared with those of lobectomy with SNS or SLNB. Conclusion: This study reveals an association of lobectomy with more systematic and complete lymph node dissection, such as BML or SND, with better prognosis in stage I NSCLC patients.
Collapse
Affiliation(s)
- Weidong Wang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Thoracic Surgery, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dongni Chen
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kexing Xi
- Department of Colorectal Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongqiang Chen
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xuewen Zhang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yinsheng Wen
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zirui Huang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiangyang Yu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gongming Wang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rusi Zhang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lanjun Zhang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
14
|
Barroso-Sousa R, Guo H, Srivastava P, James T, Birch W, Siu LL, Tew WP, Tolaney SM. Utilization of tumor genomics in clinical practice: an international survey among ASCO members. Future Oncol 2019; 15:2463-2470. [DOI: 10.2217/fon-2019-0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: To identify patterns of use and barriers to tumor genomic testing among oncologists. Methods: We surveyed American Society of Clinical Oncology physician members about their use of genomic testing. Results: Among 11,900 members surveyed, a total of 1000 responded to the survey (participation rate, 8.4%). A total of 75% of the respondents included in the analysis reported ordering tests for at least 1–10% of their patients. Practice setting (academic vs community) was only a determinant in the ordering frequency in North America. Regardless of location, academic oncologists were more likely to prescribe medicine in the context of a clinical trial. Access to clinical trials and costs associated with testing were the barriers identified worldwide. Conclusion: There is substantial variation in the use of genomic tools according to region and practice setting; yet, the barriers are similar worldwide.
Collapse
Affiliation(s)
- Romualdo Barroso-Sousa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Current affiliation: Oncology Center, Hospital Sírio-Libanês, Brasilia-DF, 70200-730, Brazil
| | - Hao Guo
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Piyush Srivastava
- Division of Hematology/Oncology, Kaiser Permanente, Walnut Creek, CA 94596, USA
| | - Ted James
- Division of Surgical Oncology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Walter Birch
- Division of Practice Management, American Society of Clinical Oncology, Alexandria, VA 22314, USA
| | - Lillian L Siu
- Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - William P Tew
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
15
|
Zhang L. [The Argument and Consensus of Lymphadenectomy on Lung Cancer Surgery]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2018; 21:176-179. [PMID: 29587935 PMCID: PMC5973022 DOI: 10.3779/j.issn.1009-3419.2018.03.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
淋巴结转移是肺癌的重要转移途径,淋巴结清扫术已成为肺癌的标准术式,同时也决定着肺癌的分期、预后及治疗策略。在临床实践中肺癌淋巴结的清扫方式各有不同,从选择性的淋巴结采样到扩大的淋巴结清扫,目前各种清扫方式存在着很大的争议,本文就目前的纵隔淋巴结清扫方式的共识及争议进行综述,为今后开展多中心临床研究提供参考。
Collapse
Affiliation(s)
- Lanjun Zhang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
16
|
Toor OM, Ahmed Z, Bahaj W, Boda U, Cummings LS, McNally ME, Kennedy KF, Pluard TJ, Hussain A, Subramanian J, Masood A. Correlation of Somatic Genomic Alterations Between Tissue Genomics and ctDNA Employing Next-Generation Sequencing: Analysis of Lung and Gastrointestinal Cancers. Mol Cancer Ther 2018; 17:1123-1132. [PMID: 29500272 DOI: 10.1158/1535-7163.mct-17-1015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/19/2017] [Accepted: 02/23/2018] [Indexed: 11/16/2022]
Abstract
Next-generation Sequencing (NGS) of cancer tissues is increasingly being carried out to identify somatic genomic alterations that may guide physicians to make therapeutic decisions. However, a single tissue biopsy may not reflect complete genomic architecture due to the heterogeneous nature of tumors. Circulating tumor DNA (ctDNA) analysis is a robust noninvasive method to detect and monitor genomic alterations in blood in real time. We analyzed 28 matched tissue NGS and ctDNA from gastrointestinal and lung cancers for concordance of somatic genomic alterations, driver, and actionable alterations. Six patients (21%) had at least one concordant mutation between tissue and ctDNA sequencing. At the gene level, among all the mutations (n = 104) detected by tissue and blood sequencing, 7.7% (n = 8) of mutations were concordant. Tissue and ctDNA sequencing identified driver mutations in 60% and 64% of the tested samples, respectively. We found high discordance between tissue and ctDNA testing, especially with respect to the driver and actionable alterations. Both tissue and ctDNA NGS detected actionable alterations in 25% of patients. When somatic alterations identified by each test were combined, the total number of patients with actionable mutations increased to 32%. Our data show significant discordance between tissue NGS and ctDNA analysis. These results suggest tissue NGS and ctDNA NGS are complementary approaches rather than exclusive of each other. When performed in isolation, tissue and ctDNA NGS can each potentially miss driver and targetable alterations, suggesting that both approaches should be incorporated to enhance mutation detection rates. Larger prospective studies are needed to better clarify this emerging precision oncology landscape. Mol Cancer Ther; 17(5); 1123-32. ©2018 AACR.
Collapse
Affiliation(s)
- Omer M Toor
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri
- Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
| | - Zaheer Ahmed
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri
| | - Waled Bahaj
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri
| | - Urooge Boda
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri
| | - Lee S Cummings
- Department of Surgery, University of Missouri Kansas City, Missouri
- Division of Hepatobiliary Surgery, Saint Luke's Hospital, Kansas City, Missouri
| | - Megan E McNally
- Division of Surgical Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
| | - Kevin F Kennedy
- Division of Cardiovascular Research, Saint Luke's Hospital, Kansas City, Missouri
| | - Timothy J Pluard
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri
- Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
- Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
| | - Arif Hussain
- Division of Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- The Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Janakiraman Subramanian
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri
- Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
- Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
| | - Ashiq Masood
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, Missouri.
- Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
- Division of Oncology, Saint Luke's Cancer Institute, Kansas City, Missouri
| |
Collapse
|
17
|
Nagarajan R, Bartley AN, Bridge JA, Jennings LJ, Kamel-Reid S, Kim A, Lazar AJ, Lindeman NI, Moncur J, Rai AJ, Routbort MJ, Vasalos P, Merker JD. A Window Into Clinical Next-Generation Sequencing-Based Oncology Testing Practices. Arch Pathol Lab Med 2017; 141:1679-1685. [PMID: 29028368 DOI: 10.5858/arpa.2016-0542-cp] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - Detection of acquired variants in cancer is a paradigm of precision medicine, yet little has been reported about clinical laboratory practices across a broad range of laboratories. OBJECTIVE - To use College of American Pathologists proficiency testing survey results to report on the results from surveys on next-generation sequencing-based oncology testing practices. DESIGN - College of American Pathologists proficiency testing survey results from more than 250 laboratories currently performing molecular oncology testing were used to determine laboratory trends in next-generation sequencing-based oncology testing. RESULTS - These presented data provide key information about the number of laboratories that currently offer or are planning to offer next-generation sequencing-based oncology testing. Furthermore, we present data from 60 laboratories performing next-generation sequencing-based oncology testing regarding specimen requirements and assay characteristics. The findings indicate that most laboratories are performing tumor-only targeted sequencing to detect single-nucleotide variants and small insertions and deletions, using desktop sequencers and predesigned commercial kits. Despite these trends, a diversity of approaches to testing exists. CONCLUSIONS - This information should be useful to further inform a variety of topics, including national discussions involving clinical laboratory quality systems, regulation and oversight of next-generation sequencing-based oncology testing, and precision oncology efforts in a data-driven manner.
Collapse
|
18
|
Baik CS, Myall NJ, Wakelee HA. Targeting BRAF-Mutant Non-Small Cell Lung Cancer: From Molecular Profiling to Rationally Designed Therapy. Oncologist 2017; 22:786-796. [PMID: 28487464 PMCID: PMC5507646 DOI: 10.1634/theoncologist.2016-0458] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/06/2017] [Indexed: 12/28/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related deaths globally. However, the identification of oncogenic driver alterations involved in the initiation and maintenance of NSCLC, such as epidermal growth factor receptor mutations and anaplastic lymphoma kinase translocation, has led to the development of novel therapies that directly target mutant proteins and associated signaling pathways, resulting in improved clinical outcomes. As sequencing techniques have improved, the molecular heterogeneity of NSCLC has become apparent, leading to the identification of a number of potentially actionable oncogenic driver mutations. Of these, one of the most promising therapeutic targets is B-Raf proto-oncogene, serine/threonine kinase (BRAF). Mutations in BRAF, observed in 2%-4% of NSCLCs, typically lead to constitutive activation of the protein and, as a consequence, lead to activation of the mitogen-activated protein kinase signaling pathway. Direct inhibition of mutant BRAF and/or the downstream mitogen-activated protein kinase kinase (MEK) has led to prolonged survival in patients with BRAF-mutant metastatic melanoma. This comprehensive review will discuss the clinical characteristics and prognostic implications of BRAF-mutant NSCLC, the clinical development of BRAF and MEK inhibitors from melanoma to NSCLC, and practical considerations for clinicians involving BRAF mutation screening and the choice of targeted therapy. IMPLICATIONS FOR PRACTICE Personalized medicine has begun to provide substantial benefit to patients with oncogene-driven non-small cell lung cancer (NSCLC). However, treatment options for patients with oncogenic driver mutations lacking targeted treatment strategies remain limited. Direct inhibition of mutant B-Raf proto-oncogene, serine/threonine kinase (BRAF) and/or downstream mitogen-activated protein kinase kinase (MEK) has the potential to change the course of the disease for patients with BRAF-mutant NSCLC, as it has in BRAF-mutant melanoma. Optimization of screening strategies for rare mutations and the choice of appropriate agents on an individual basis will be key to providing timely and successful intervention.
Collapse
Affiliation(s)
- Christina S Baik
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
19
|
Dalton WB, Forde PM, Kang H, Connolly RM, Stearns V, Gocke CD, Eshleman JR, Axilbund J, Petry D, Geoghegan C, Wolff AC, Loeb DM, Pratilas CA, Meyer CF, Christenson ES, Slater SA, Ensminger J, Parsons HA, Park BH, Lauring J. Personalized Medicine in the Oncology Clinic: Implementation and Outcomes of the Johns Hopkins Molecular Tumor Board. JCO Precis Oncol 2017; 2017. [PMID: 30003184 DOI: 10.1200/po.16.00046] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose Tumor genomic profiling for personalized oncology therapy is being widely applied in clinical practice even as it is being evaluated more formally in clinical trials. Given the complexities of genomic data and its application to clinical use, molecular tumor boards with diverse expertise can provide guidance to oncologists and patients seeking to implement personalized genetically targeted therapy in practice. Methods A multidisciplinary molecular tumor board reviewed tumor molecular profiling reports from consecutive referrals at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins over a 3-year period. The tumor board weighed evidence for actionability of genomic alterations identified by molecular profiling and provided recommendations including US Food and Drug Administration-approved drug therapy, clinical trials of matched targeted therapy, off-label use of such therapy, and additional tumor or germline genetic testing. Results One hundred fifty-five patients were reviewed. Actionable genomic alterations were identified in 132 patients (85%). Off-label therapies were recommended in 37 patients (24%). Eleven patients were treated off-label, and 13 patients were enrolled onto clinical trials of matched targeted therapies. Median progression-free survival of patients treated with matched therapies was 5 months (95% CI, 2.9 months to not reached), and the progression-free survival probability at 6 months was 43%(95% CI, 26% to 71%). Lack of locally available clinical trials was the major limitation on clinical actionability of tumor profiling reports. Conclusion The molecular tumor board recommended off-label targeted therapies for a quarter of all patients reviewed. Outcomes were heterogeneous, although 43% of patients receiving genomically matched therapy derived clinical benefit lasting at least 6 months. Until more data become available from precision oncology trials, molecular tumor boards can help guide appropriate use of tumor molecular testing to direct therapy.
Collapse
Affiliation(s)
- W Brian Dalton
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Patrick M Forde
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Hyunseok Kang
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Roisin M Connolly
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Vered Stearns
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Christopher D Gocke
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - James R Eshleman
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | | | - Dana Petry
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | | | - Antonio C Wolff
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - David M Loeb
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | | | - Christian F Meyer
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Eric S Christenson
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Shannon A Slater
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Jennifer Ensminger
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Heather A Parsons
- Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Ben H Park
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Josh Lauring
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
20
|
Abstract
INTRODUCTION Next-Generation-Sequencing (NGS) has enabled gene mutation profiling - cataloguing sequence variants and modifications in clinical assays encompassing tens to thousands of genes in tumors and in germlines. The clinical benefit of applying multi-gene NGS to diverse applications in various malignancies remains to be demonstrated. AREAS COVERED Applications of gene mutation profiling in oncology include screening cancer-prone families, classification of malignancies, treatment selection, and monitoring the response to treatment of solid tumors (the 'liquid biopsy'). Google Scholar was used to search PubMed for the period 2011-2016 using combinations of the following search terms: 'clinical utility', NGS, 'molecular diagnostics'. Expert commentary: Clinical studies are in progress pairing mutation profiling with streamlined new trial designs to speed identification of promising drug-target combinations and to see if genotype-informed treatment selection will improve outcome across a spectrum of histologies. The analytical advantages and falling cost of NGS make focused gene panels likely to become the dominant modality in molecular diagnostic testing even if trials eventually discourage use of large panels to test all malignancies.
Collapse
Affiliation(s)
- Loren Joseph
- a Department of Pathology, Beth Israel Deaconess Medical Center, Molecular Diagnostics Laboratory , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
21
|
Day D, Bedard PL. Beta-Testing of Next-Generation DNA Sequencing for Patients With Advanced Cancers Treated at Community Hospitals. J Oncol Pract 2016; 12:279-81. [DOI: 10.1200/jop.2015.010165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Daphne Day
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | - Philippe L. Bedard
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|