1
|
Arch JJ, Slivjak ET, Finkelstein LB. A Novel Intervention to Reduce Fear of Progression and Trauma Symptoms in Advanced Cancer Using Written Exposure to Worst-Case Scenarios. J Palliat Med 2024; 27:1009-1017. [PMID: 38579139 DOI: 10.1089/jpm.2023.0658] [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] [Indexed: 04/07/2024] Open
Abstract
Background: Adults with advanced cancer experience profound future uncertainty, reflected in elevated fear of cancer progression (FoP) and cancer-related trauma symptoms. These symptoms are associated with physical symptom burden and poorer quality of life, and few interventions exist to manage them. Objective: To develop and pilot a written exposure-based coping intervention (EASE) focused on worst-case scenarios among adults with advanced cancer reporting elevated cancer-related trauma symptoms or FoP. Design: A single-arm intervention development and pilot trial. Participants: The trial enrolled 29 U.S. adults with stage III or stage IV solid tumor cancer (n = 24) or incurable or higher-risk blood cancer (n = 5) reporting elevated cancer-related trauma symptoms or FoP. Among those screened, 74% were eligible, with an eligible-to-enrolled rate of 85%. Design/Measurements: EASE was delivered over five 1:1 videoconferencing sessions. Feasibility and acceptability were evaluated via attendance, surveys, and exit interviews. Outcomes were assessed at five time points through 3-month (FU1, main assessment of interest) and 4.5-month (FU2) follow-up. Results: Participant and interventionist feedback was used to iteratively refine EASE. Among participants, 86% (25/29) completed all five sessions and FU1; surveys and exit interviews indicated high acceptability. Primary outcomes of cancer-related trauma symptoms and FoP improved significantly from pre to both follow-ups by predominantly large effect sizes. Secondary outcomes of anxiety, depression, hopelessness, fear of death/dying, and fatigue, and most process measures improved significantly by FU1 or FU2. Conclusions: EASE, a novel adaptation of written exposure therapy, is a promising approach to reducing FoP and cancer-related trauma symptoms among adults with advanced cancer that warrants further study.
Collapse
Affiliation(s)
- Joanna J Arch
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Division of Cancer Prevention and Control, University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elizabeth T Slivjak
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lauren B Finkelstein
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
2
|
Wallenta Law J, Bapat B, Sweetnam C, Mohammed H, McBratney A, Izano MA, Scannell Bryan M, Spencer S, Schroeder B, Hostin D, Simon GR, Berry AB. Real-World Impact of Comprehensive Genomic Profiling on Biomarker Detection, Receipt of Therapy, and Clinical Outcomes in Advanced Non-Small Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2400075. [PMID: 38754057 PMCID: PMC11371096 DOI: 10.1200/po.24.00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/29/2024] [Indexed: 05/18/2024] Open
Abstract
PURPOSE Therapeutic decision making for patients with advanced non-small cell lung cancer (aNSCLC) includes a growing number of options for genomic, biomarker-guided, targeted therapies. We compared actionable biomarker detection, targeted therapy receipt, and real-world overall survival (rwOS) in patients with aNSCLC tested with comprehensive genomic profiling (CGP) versus small panel testing (SP) in real-world community health systems. METHODS Patients older than 18 years diagnosed with aNSCLC between January 1, 2015, and December 31, 2020, who received biomarker testing were followed until death or study end (September 30, 2021), and categorized by most comprehensive testing during follow-up: SP (≤52 genes) or CGP (>52 genes). RESULTS Among 3,884 patients (median age, 68 years; 50% female; 73% non-Hispanic White), 20% received CGP and 80% SP. The proportion of patients with ≥one actionable biomarker (actionability) was significantly higher in CGP than in SP (32% v 14%; P < .001). Of patients with actionability, 43% (CGP) and 38% (SP) received matched therapies (P = .20). Among treated patients, CGP before first-line treatment was associated with higher likelihood of matched therapy in any line (odds ratio, 3.2 [95% CI, 1.84 to 5.53]). CGP testing (hazard ratio [HR], 0.80 [95% CI, 0.72 to 0.89]) and actionability (HR, 0.84 [95% CI, 0.77 to 0.91]) were associated with reduced risk of mortality. Among treated patients with actionability, matched therapy receipt showed improved median rwOS in months in CGP (34 [95% CI, 21 to 49] matched v 14 [95% CI, 10 to 18] unmatched) and SP (27 [95% CI, 21 to 43] matched v 10 [95% CI, 8 to 14] unmatched). CONCLUSION Patients who received CGP had improved detection of actionable biomarkers and greater use of matched therapies, both of which were associated with significant increases in survival.
Collapse
|
3
|
Chon E, Hendricks W, White M, Rodrigues L, Haworth D, Post G. Precision Medicine in Veterinary Science. Vet Clin North Am Small Anim Pract 2024; 54:501-521. [PMID: 38212188 DOI: 10.1016/j.cvsm.2023.12.006] [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] [Indexed: 01/13/2024]
Abstract
Precision medicine focuses on the clinical management of the individual patient, not on population-based findings. Successes from human precision medicine inform veterinary oncology. Early evidence of success for canines shows how precision medicine can be integrated into practice. Decreasing genomic profiling costs will allow increased utilization and subsequent improvement of knowledge base from which to make better informed decisions. Utility of precision medicine in canine oncology will only increase for improved cancer characterization, enhanced therapy selection, and overall more successful management of canine cancer. As such, practitioners are called to interpret and leverage precision medicine reports for their patients.
Collapse
Affiliation(s)
- Esther Chon
- Vidium Animal Health, 7201 East Henkel Way, Suite 210, Scottsdale, AZ 85255, USA
| | - William Hendricks
- Vidium Animal Health, 7201 East Henkel Way, Suite 210, Scottsdale, AZ 85255, USA
| | - Michelle White
- OneHealthCompany, Inc, 530 Lytton Avenue, 2nd Floor, Palo Alto, CA 94301, USA
| | - Lucas Rodrigues
- OneHealthCompany, Inc, 530 Lytton Avenue, 2nd Floor, Palo Alto, CA 94301, USA
| | - David Haworth
- Vidium Animal Health, 7201 East Henkel Way, Suite 210, Scottsdale, AZ 85255, USA
| | - Gerald Post
- OneHealthCompany, Inc, 530 Lytton Avenue, 2nd Floor, Palo Alto, CA 94301, USA.
| |
Collapse
|
4
|
Ferreira-Gonzalez A, Hocum B, Ko G, Shuvo S, Appukkuttan S, Babajanyan S. Next-Generation Sequencing Trends among Adult Patients with Select Advanced Tumor Types: A Real-World Evidence Evaluation. J Mol Diagn 2024; 26:292-303. [PMID: 38296192 DOI: 10.1016/j.jmoldx.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/10/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
There are limited data on the prevalence of next-generation sequencing (NGS) in the United States, especially in light of the increasing importance of identifying actionable oncogenic variants due to molecular biomarker-based therapy approvals. This retrospective study of adult patients with select metastatic solid tumors and central nervous system tumors from the Optum Clinformatics Data Mart US health care claims database (January 1, 2014, to June 30, 2021; N = 63,209) examined NGS use trends over time. A modest increase in NGS was observed across tumor types from 2015 (0.0% to 1.5%) to 2021 (2.1% to 17.4%). A similar increase in NGS rates was also observed across key periods; however, rates in the final key period remained <10% for patients with breast, colorectal, head and neck, soft tissue sarcoma, and thyroid cancers, as well as central nervous system tumors. The median time to NGS from diagnosis was shortest among patients with non-small-cell lung cancer and longest for patients with breast cancer. Predictors of NGS varied by tumor type; test rates for minorities in select tumor types appeared comparable to the White population. Despite improving payer policies to expand coverage of NGS and molecular biomarker-based therapy approvals, NGS rates remained low across tumor types. Given the potential for improved patient outcomes with molecular biomarker-based therapy, further efforts to improve NGS rates are warranted.
Collapse
Affiliation(s)
| | - Brian Hocum
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | - Gilbert Ko
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey.
| | - Sohul Shuvo
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | | |
Collapse
|
5
|
Sakthikumar S, Warrier M, Whitley D, Facista S, Adkins J, Aman S, Tsinajinnie D, Duran N, Siravegna G, Ahmed Z, Day K, Jenkins B, Patel N, Ryden K, Nadai J, Banovich K, Powers B, Edwards J, Steinberg J, Fielder S, Wong S, Byron SA, Izatt T, Zismann V, Boateng M, Zhu Z, Chuang HY, Trent JM, Haworth D, Chon E, Hendricks W, Wang G. Genomic analysis across 53 canine cancer types reveals novel mutations and high clinical actionability potential. Vet Comp Oncol 2024; 22:30-41. [PMID: 38053317 DOI: 10.1111/vco.12944] [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: 07/28/2023] [Revised: 09/27/2023] [Accepted: 11/12/2023] [Indexed: 12/07/2023]
Abstract
A genomic understanding of the oncogenic processes and individual variability of human cancer has steadily fueled improvement in patient outcomes over the past 20 years. Mutations within tumour tissues are routinely assessed through clinical genomic diagnostic assays by academic and commercial laboratories to facilitate diagnosis, prognosis and effective treatment stratification. The application of genomics has unveiled a wealth of mutation-based biomarkers in canine cancers, suggesting that the transformative principles that have revolutionized human cancer medicine can be brought to bear in veterinary oncology. To advance clinical genomics and genomics-guided medicine in canine oncology, we have developed and validated a canine cancer next-generation sequencing gene panel for the identification of multiple mutation types in clinical specimens. With this panel, we examined the genomic landscapes of 828 tumours from 813 dogs, spanning 53 cancer types. We identified 7856 alterations, encompassing copy number variants, single nucleotide variants, indels and internal tandem duplications. Additionally, we evaluated the clinical utility of these alterations by incorporating a biomarker framework from comprehensive curation of primary canine literature and inferences from human cancer genomic biomarker literature and clinical diagnostics. Remarkably, nearly 90% of the cases exhibited mutations with diagnostic, prognostic or therapeutic implications. Our work represents a thorough assessment of genomic landscapes in a large cohort of canine cancers, the first of its kind for its comprehensive inclusion of multiple mutation types and structured annotation of biomarkers, demonstrating the clinical potential of leveraging mutation-based biomarkers in veterinary oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Sara Aman
- Vidium Animal Health, Phoenix, Arizona, USA
| | | | | | | | | | | | | | | | - Kirk Ryden
- Vidium Animal Health, Phoenix, Arizona, USA
| | - Joe Nadai
- Vidium Animal Health, Phoenix, Arizona, USA
| | | | | | | | - Jennifer Steinberg
- Center for Cancer Research at the National Cancer Institute, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Shukmei Wong
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Sara A Byron
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Tyler Izatt
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Victoria Zismann
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | | | | | | | - Jeffrey M Trent
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | | | | | | | | |
Collapse
|
6
|
Wilczyński J, Paradowska E, Wilczyński M. Personalization of Therapy in High-Grade Serous Tubo-Ovarian Cancer-The Possibility or the Necessity? J Pers Med 2023; 14:49. [PMID: 38248751 PMCID: PMC10817599 DOI: 10.3390/jpm14010049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/17/2023] [Accepted: 12/25/2023] [Indexed: 01/23/2024] Open
Abstract
High-grade serous tubo-ovarian cancer (HGSTOC) is the most lethal tumor of the female genital tract. The foregoing therapy consists of cytoreduction followed by standard platinum/taxane chemotherapy; alternatively, for primary unresectable tumors, neo-adjuvant platinum/taxane chemotherapy followed by delayed interval cytoreduction. In patients with suboptimal surgery or advanced disease, different forms of targeted therapy have been accepted or tested in clinical trials. Studies on HGSTOC discovered its genetic and proteomic heterogeneity, epigenetic regulation, and the role of the tumor microenvironment. These findings turned attention to the fact that there are several distinct primary tumor subtypes of HGSTOC and the unique biology of primary, metastatic, and recurrent tumors may result in a differential drug response. This results in both chemo-refractoriness of some primary tumors and, what is significantly more frequent and destructive, secondary chemo-resistance of metastatic and recurrent HGSTOC tumors. Treatment possibilities for platinum-resistant disease include several chemotherapeutics with moderate activity and different targeted drugs with difficult tolerable effects. Therefore, the question appears as to why different subtypes of ovarian cancer are predominantly treated based on the same therapeutic schemes and not in an individualized way, adjusted to the biology of a specific tumor subtype and temporal moment of the disease. The paper reviews the genomic, mutational, and epigenetic signatures of HGSTOC subtypes and the tumor microenvironment. The clinical trials on personalized therapy and the overall results of a new, comprehensive approach to personalized therapy for ovarian cancer have been presented and discussed.
Collapse
Affiliation(s)
- Jacek Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Street, 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Street, 93-232 Lodz, Poland;
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Street, 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Street, 90-419 Lodz, Poland
| |
Collapse
|
7
|
Bertoletti L, Madridano O, Jiménez D, Muriel A, Bikdeli B, Ay C, Trujillo-Santos J, Bosevski M, Sigüenza P, Monreal M. Cancer-Associated Thrombosis: Trends in Clinical Features, Treatment, and Outcomes From 2001 to 2020. JACC CardioOncol 2023; 5:758-772. [PMID: 38205016 PMCID: PMC10774792 DOI: 10.1016/j.jaccao.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 01/12/2024] Open
Abstract
Background Despite advances in cancer and venous thromboembolism (VTE) management, the epidemiology of cancer-associated thrombosis management over time remains unclear. Objectives We analyzed data from the RIETE (Registro Informatizado de la Enfermedad Trombo Embólica) registry spanning 2001 to 2020 to investigate temporal trends in clinical characteristics and treatments for cancer-associated thrombosis. Methods Using multivariable survival regression, we examined temporal trends in risk-adjusted rates of symptomatic VTE recurrences, major bleeding, and death within 30 days after incident VTE. Results Among the 17,271 patients with cancer-associated thrombosis, there was a progressive increase in patients presenting with pulmonary embolism (from 44% in 2001-2005 to 55% in 2016-2020; P < 0.001 for trend), lung (from 12.7% to 18.1%; P < 0.001) or pancreatic cancer (from 3.8% to 5.6%; P = 0.003), and utilization of immunotherapy (from 0% to 7.4%; P < 0.001). Conversely, there was a decline in patients with prostate cancer (from 11.7% to 6.6%; P < 0.001) or carcinoma of unknown origin (from 3.5% to 0.7%; P < 0.001). At the 30-day follow-up, a reduction was observed in the proportion of patients experiencing symptomatic VTE recurrences (from 3.1% to 1.1%; P < 0.001), major bleeding (from 3.1% to 2.2%; P = 0.004), and death (from 11.9% to 8.4%; P < 0.001). Multivariable analyses revealed a decreased risk over time for VTE recurrence (adjusted subdistribution HR [asHR]: 0.94 per year; 95% CI: 0.92-0.98), major bleeding (asHR: 0.98; 95% CI: 0.96-0.99), and death (aHR: 0.97; 95% CI: 0.96-0.98). Conclusions In this multicenter study of cancer patients with VTE, there was a decline in thrombotic, hemorrhagic, and fatal events from 2001 to 2020. (Registro Informatizado de la Enfermedad Trombo Embólica [RIETE]; NCT02832245).
Collapse
Affiliation(s)
- Laurent Bertoletti
- Service de Médecine Vasculaire et Thérapeutique, Centre Hospitalier Universitaire de St-Etienne, Saint-Etienne, France
- INSERM, UMR1059, Equipe Dysfonction Vasculaire et Hémostase, Université Jean-Monnet, Saint-Etienne, France
- INSERM, CIC-1408, CHU Saint-Etienne, Saint-Etienne, France
| | - Olga Madridano
- Department of Internal Medicine, Hospital Infanta Sofía, Madrid, Spain
| | - David Jiménez
- Respiratory Department, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS, Madrid, Spain
- Medicine Department, Universidad de Alcalá, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
| | - Alfonso Muriel
- Biostatistics Unit, Ramón y Cajal Hospital IRYCIS, CIBERESP, Nursing and Physiotherapy Department, University of Alcala, Madrid, Spain
| | - Behnood Bikdeli
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Thrombosis Research Group, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Yale/YNHH Center for Outcomes Research & Evaluation, New Haven, Connecticut, USA
- Cardiovascular Research Foundation, New York, New York, USA
| | - Cihan Ay
- Department of Medicine I, Clinical Division of Haematology and Haemostaseology, Medical University of Vienna, Vienna, Austria
| | - Javier Trujillo-Santos
- Internal Medicine Department, Hospital Universitario Santa Lucía, Complejo Hospitalario Universitario de Cartagena, Murcia, Spain
- Universidad Católica San Antonio de Murcia, Murcia, Spain
| | - Marijan Bosevski
- University Cardiology Clinic Faculty of Medicine, Skopje, North Macedonia
| | - Patricia Sigüenza
- Department of Internal Medicine, Hospital Germans Trias I Pujol, Badalona, Barcelona, Spain
| | - Manuel Monreal
- Faculty of Health Sciences, Universidad Católica San Antonio de Murcia, Murcia, Spain
| |
Collapse
|
8
|
Vaidya R, Unger JM, Qian L, Minichiello K, Herbst RS, Gandara DR, Neal JW, Leal TA, Patel JD, Dragnev KH, Waqar SN, Edelman MJ, Sigal EV, Adam SJ, Malik S, Blanke CD, LeBlanc ML, Kelly K, Gray JE, Redman MW. Representativeness of Patients Enrolled in the Lung Cancer Master Protocol (Lung-MAP). JCO Precis Oncol 2023; 7:e2300218. [PMID: 37677122 PMCID: PMC10581630 DOI: 10.1200/po.23.00218] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/18/2023] [Accepted: 07/20/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE Lung Cancer Master Protocol (Lung-MAP), a public-private partnership, established infrastructure for conducting a biomarker-driven master protocol in molecularly targeted therapies. We compared characteristics of patients enrolled in Lung-MAP with those of patients in advanced non-small-cell lung cancer (NSCLC) trials to examine if master protocols improve trial access. METHODS We examined patients enrolled in Lung-MAP (2014-2020) according to sociodemographic characteristics. Proportions for characteristics were compared with those for a set of advanced NSCLC trials (2001-2020) and the US advanced NSCLC population using SEER registry data (2014-2018). Characteristics of patients enrolled in Lung-MAP treatment substudies were examined in subgroup analysis. Two-sided tests of proportions at an alpha of .01 were used for all comparisons. RESULTS A total of 3,556 patients enrolled in Lung-MAP were compared with 2,215 patients enrolled in other NSCLC studies. Patients enrolled in Lung-MAP were more likely to be 65 years and older (57.2% v 46.3%; P < .0001), from rural areas (17.3% v 14.4%; P = .004), and from socioeconomically deprived neighborhoods (42.2% v 36.7%, P < .0001), but less likely to be female (38.6% v 47.2%; P < .0001), Asian (2.8% v 5.1%; P < .0001), or Hispanic (2.4% v 3.8%; P = .003). Among patients younger than 65 years, Lung-MAP enrolled more patients using Medicaid/no insurance (27.6% v 17.8%; P < .0001). Compared with the US advanced NSCLC population, Lung-MAP under represented patients 65 years and older (57.2% v 69.8%; P < .0001), females (38.6% v 46.0%; P < .0001), and racial or ethnic minorities (14.8% v 21.5%; P < .0001). CONCLUSION Master protocols may improve access to trials using novel therapeutics for older patients and socioeconomically vulnerable patients compared with conventional trials, but specific patient exclusion criteria influenced demographic composition. Further research examining participation barriers for under represented racial or ethnic minorities in precision medicine clinical trials is warranted.
Collapse
Affiliation(s)
- Riha Vaidya
- Fred Hutchinson Cancer Center, Seattle, WA
- SWOG Statistics and Data Management Center, Seattle, WA
| | - Joseph M. Unger
- Fred Hutchinson Cancer Center, Seattle, WA
- SWOG Statistics and Data Management Center, Seattle, WA
| | - Lu Qian
- Fred Hutchinson Cancer Center, Seattle, WA
| | - Katherine Minichiello
- Fred Hutchinson Cancer Center, Seattle, WA
- SWOG Statistics and Data Management Center, Seattle, WA
| | | | | | | | | | - Jyoti D. Patel
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | - Martin J. Edelman
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA
| | | | - Stacey J. Adam
- Foundations for the National Institutes of Health, North Bethesda, MD
| | | | - Charles D. Blanke
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR
| | - Michael L. LeBlanc
- Fred Hutchinson Cancer Center, Seattle, WA
- SWOG Statistics and Data Management Center, Seattle, WA
| | - Karen Kelly
- UC Davis Comprehensive Cancer Center, Sacramento, CA
| | | | - Mary W. Redman
- Fred Hutchinson Cancer Center, Seattle, WA
- SWOG Statistics and Data Management Center, Seattle, WA
| |
Collapse
|
9
|
Lee PWP, Strum SW, Tsvetkova E. It Is All in the Genes: A Story of Unexpected Survival in a 67-Year-Old Male with Metastatic Pancreatic Cancer. Case Rep Oncol Med 2023; 2023:8751205. [PMID: 37547629 PMCID: PMC10400294 DOI: 10.1155/2023/8751205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Background We describe a case report of a 67-year-old male with PDAC who experienced an exceptional survival outcome during systemic therapy and its implications in precision medicine. We hypothesize that his outcomes are attributable, in part, to a germline BRCA2 deletion and somatic GNAS substitution. Methods Retrospective single-patient chart review was performed at the London Regional Cancer Program, as well as a structured literature search spanning all years in PubMed of BRCA and GNAS mutations in pancreatic cancer. Results The case described herein represents a 67-year-old male who survived over 27 months after third-line treatment with gemcitabine, docetaxel, and capecitabine (GTX) chemotherapy for metastatic PDAC after progression on gemcitabine and Abraxane and then on FOLFIRINOX. His survival far exceeded the median overall survival metrics. Genetic testing revealed a pathogenic heterozygous germline BRCA2 6643delT p.(Tyr2215Thrfs∗14) frameshift mutation and somatic GNAS 2531G > A p.(Arg844His) mutation. Conclusions This case highlights the urgent need to expand our knowledge of cancer biology to advance personalized cancer treatment and therapy development.
Collapse
Affiliation(s)
- Patsy W. P. Lee
- Department of Internal Medicine, Schulich School of Medicine and Dentistry, Western University, Canada
| | - Scott W. Strum
- Department of Medical Oncology, London Regional Cancer Program, Western University, Canada
| | - Elena Tsvetkova
- Department of Medical Oncology, London Regional Cancer Program, Western University, Canada
| |
Collapse
|
10
|
Fortune EE, Zaleta AK, Saxton MC. Biomarker testing communication, familiarity, and informational needs among people living with breast, colorectal, and lung cancer. PATIENT EDUCATION AND COUNSELING 2023; 112:107720. [PMID: 37062167 DOI: 10.1016/j.pec.2023.107720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/06/2023] [Accepted: 03/29/2023] [Indexed: 05/09/2023]
Abstract
OBJECTIVES This study aims to characterize patient experiences with biomarker testing, including history of biomarker testing, related communication and education, self-perceived familiarity and informational needs. METHODS 436 U.S. adults diagnosed with lung (38%), colorectal (35%) or breast cancer (27%) from 2018 to 2022 completed a survey. Two logistic regressions were conducted to predict patients' familiarity with biomarker testing and informational needs. RESULTS Despite high biomarker testing rates (85%), half of respondents reported low familiarity with biomarker testing and three-quarters reported outstanding informational needs. Regression models indicate those patients who have greater health literacy and report having conversations with their oncologists about biomarker testing have more familiarity with biomarker testing and less informational needs, even after controlling for important sociodemographic factors. CONCLUSIONS There is an opportunity to improve patients' familiarity with biomarker testing and decrease outstanding informational needs by focusing on factors such as health literacy and patient-provider communication, which could further cultivate patients' understanding of the importance of biomarker testing in cancer care. PRACTICE IMPLICATIONS These findings underscore the importance of patient-provider relationships and the need for additional tools that assist providers in assessing patients' health literacy and facilitating conversations with patients, especially those focused on complex topics such as biomarker testing.
Collapse
Affiliation(s)
- Erica E Fortune
- Cancer Support Community, Research and Training Institute, Philadelphia, PA, USA.
| | - Alexandra K Zaleta
- Cancer Support Community, Research and Training Institute, Philadelphia, PA, USA
| | | |
Collapse
|
11
|
Martens UM, Schröder J, Bengsch F, Sellmann L, Busies S, Frank-Gleich S, Zaiss M, Decker T, Schneeweiss A, Schuler M, Grebhardt S, Zacharias S, Marschner N, Kasenda B, Potthoff K, Vannier C. The INFINITY study protocol: a retrospective cohort study on decision making and clinical impact of biomarker-driven precision oncology in routine clinical practice. BMC Cancer 2023; 23:543. [PMID: 37312086 DOI: 10.1186/s12885-023-11046-3] [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: 05/05/2022] [Accepted: 06/04/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Precision oncology, defined as treatment of patients with targeted therapies matched to specific molecular alterations, has entered routine clinical practice. Particularly in patients with advanced cancer or hematologic malignancies, for whom no further standard therapies are available, this approach is increasingly applied as last resort option outside of the approved indication. However, data on patient outcomes are not systematically collected, analyzed, reported, and shared. We have initiated the INFINITY registry to provide evidence from routine clinical practice to fill this knowledge gap. METHODS INFINITY is a retrospective, non-interventional cohort study conducted at approximately 100 sites in Germany (office-based oncologists/hematologists and hospitals). We aim to include 500 patients with advanced solid tumors or hematologic malignancies who received a non-standard targeted therapy based on potentially actionable molecular alterations or biomarkers. INFINITY aims to provide insights into the use of precision oncology in routine clinical practice within Germany. We systematically collect details on patient and disease characteristics, molecular testing, clinical decision-making, treatment, and outcome. DISCUSSION INFINITY will provide evidence on the current biomarker landscape driving treatment decisions in routine clinical care. It will also provide insights on effectiveness of precision oncology approaches in general, and of specific drug class/alteration matches used outside their approved indications. TRIAL REGISTRATION The study is registered at ClinicalTrials.gov, NCT04389541.
Collapse
Affiliation(s)
- Uwe M Martens
- Clinic for Hematology, Oncology and Palliative Care, SLK Kliniken Heilbronn, Heilbronn, Germany
- MOLIT Institute for Personalized Medicine, Heilbronn, Germany
| | - Jan Schröder
- Practice for Hematology and Internal Oncology, Mülheim a.d.R, Germany
| | - Fee Bengsch
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany
| | | | - Sabine Busies
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany
| | - Stefanie Frank-Gleich
- Joint Practice for Internal Medicine, Hematology, Oncology, Gastroenterology, Halle (Saale), Germany
| | - Matthias Zaiss
- Center for Interdisciplinary Oncology & Hematology, Freiburg, Germany
| | - Thomas Decker
- Study Center for Oncology Ravensburg, Ravensburg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center, Heidelberg, Germany
| | - Martin Schuler
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sina Grebhardt
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany
| | - Stefan Zacharias
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany
| | - Norbert Marschner
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany
| | - Benjamin Kasenda
- Medical Oncology, University Hospital and University of Basel, Basel, Switzerland
| | - Karin Potthoff
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany.
| | - Corinne Vannier
- iOMEDICO, Ellen-Gottlieb-Straße 19, 79106, Freiburg im Breisgau, Germany
| |
Collapse
|
12
|
Gibbs SN, Peneva D, Cuyun Carter G, Palomares MR, Thakkar S, Hall DW, Dalglish H, Campos C, Yermilov I. Comprehensive Review on the Clinical Impact of Next-Generation Sequencing Tests for the Management of Advanced Cancer. JCO Precis Oncol 2023; 7:e2200715. [PMID: 37285561 DOI: 10.1200/po.22.00715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/14/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023] Open
Abstract
PURPOSE This review summarizes the published evidence on the clinical impact of using next-generation sequencing (NGS) tests to guide management of patients with cancer in the United States. METHODS We performed a comprehensive literature review to identify recent English language publications that presented progression-free survival (PFS) and overall survival (OS) of patients with advanced cancer receiving NGS testing. RESULTS Among 6,475 publications identified, 31 evaluated PFS and OS among subgroups of patients who received NGS-informed cancer management. PFS and OS were significantly longer among patients who were matched to targeted treatment in 11 and 16 publications across tumor types, respectively. CONCLUSION Our review indicates that NGS-informed treatment can have an impact on survival across tumor types.
Collapse
Affiliation(s)
- Sarah N Gibbs
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | - Desi Peneva
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | | | | | | | | | - Hannah Dalglish
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | - Cynthia Campos
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | - Irina Yermilov
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| |
Collapse
|
13
|
Stallings SC, Richmond J, Canedo JR, Beard K, Bonnet K, Schlundt DG, Wilkins CH, Aldrich MC. Assessing patient-level knowledge of precision medicine in a community health center setting. J Community Genet 2023; 14:197-210. [PMID: 36609637 PMCID: PMC10104983 DOI: 10.1007/s12687-023-00632-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/02/2023] [Indexed: 01/09/2023] Open
Abstract
As precision medicine approaches are implemented, cancer treatment decisions have come to require comprehension of genetic tests and their role in risk stratification and treatment options. Acceptance and implementation of precision medicine requires patient understanding of numeracy, genetic literacy, health literacy, and medical trust. Implementing precision medicine in a US federally qualified community health center (FQCHC) setting has received little attention. Using a mixed-methods approach, we sought to identify patient-level factors influencing the understanding of cancer risk and precision medicine among FQCHC patients. We enrolled 26 English-speaking adults aged 40-79 years. Participants enrolled in focus groups and completed surveys to assess patient-level understanding of precision medicine, numeracy, and health literacy. The majority of participants were female (77%) and self-identified as African American (89%). Approximately one-third reported having a high school degree or less. While health literacy was generally high, 42% felt that genes or genetics had little impact on health and most (69%) reported little familiarity with precision medicine. Many participants reported that trust in their providers was extremely or very important when receiving genetic tests. Numeracy levels were moderate, with nearly half reporting some discomfort working with fractions and 38% finding numerical information only occasionally useful. Findings suggest that patients may lack familiarity with precision medicine concepts relevant for understanding cancer treatment decisions. Future educational efforts may help bridge the gap in patient understanding and facilitate equitable opportunities for precision medicine for all patients, including those seeking care from community health centers.
Collapse
Affiliation(s)
- Sarah C. Stallings
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jennifer Richmond
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Juan R. Canedo
- Department of Internal Medicine, University of Kentucky, Lexington, KY USA
| | - Katina Beard
- Matthew Walker Comprehensive Health Center, Nashville, TN USA
| | - Kemberlee Bonnet
- Department of Psychology, Vanderbilt University, Nashville, TN USA
- Qualitative Research Core, Vanderbilt University Medical Center, Nashville, TN USA
| | - David G. Schlundt
- Department of Psychology, Vanderbilt University, Nashville, TN USA
- Qualitative Research Core, Vanderbilt University Medical Center, Nashville, TN USA
| | - Consuelo H. Wilkins
- Department of Medicine, Division of Geriatrics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Melinda C. Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
14
|
Lemmon CA, Zhou J, Hobbs B, Pennell NA. Modeling Costs and Life-Years Gained by Population-Wide Next-Generation Sequencing or Single-Gene Testing in Nonsquamous Non-Small-Cell Lung Cancer in the United States. JCO Precis Oncol 2023; 7:e2200294. [PMID: 36634300 PMCID: PMC9928881 DOI: 10.1200/po.22.00294] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
PURPOSE Many patients with actionable driver oncogenes (ADOs) are never identified and thus never receive targeted treatment. This study evaluated the economic impact and the potential life-years gained (LYG) that can be attributed to the extent of next-generation sequencing (NGS) testing in the United States compared with single-gene testing (SGT) in patients with metastatic nonsquamous non-small-cell lung cancer in the United States. METHODS A model was developed to evaluate incremental rates of SGT or NSG testing on the basis of LYG and cost per LYG. ADOs included for NGS included EGFR, ALK, ROS1, BRAF, RET, MET, and NTRK. SGT included EGFR and ALK. Assumptions were made for expected incidence of ADOs. Survival distributions were fit to published trial averages of median and 5-year overall survival. Treatment costs were estimated from drug cost averages. Reimbursement costs were based on data from the Center for Medicare and Medicaid Services. RESULTS Each incremental 10% increase in NGS testing produces an average of 2,627.4 additional LYG, with an average cost savings per LYG of $75 US dollars (USD). Replacing SGT at the current rate of 80% with NGS testing would result in an average additional 21,09.6 LYG and reduce cost per LYG by an average of $599 USD. If 100% of eligible patients were tested with NGS and each identified patient had matched treatment, the total average cost per LYG would be $16,641.57 USD. CONCLUSION On the basis of current evidence, population-level simulations demonstrate that clinically relevant gains in survival with non-negligible reduction in costs are obtainable from widespread adoption of NGS testing and appropriate treatment matching for patients with advanced nonsquamous non-small-cell lung cancer.
Collapse
Affiliation(s)
- Christopher A. Lemmon
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH,Present address: University of Cincinnati College of Medicine, Department of Internal Medicine, Division of Hematology and Oncology, Cincinnati, OH,Christopher A. Lemmon, MD, University of Cincinnati College of Medicine, Department of Internal Medicine, Division of Hematology and Oncology, Vontz Center, 3109 Eden Ave, Room 1330, Cincinnati, OH 45219; e-mail:
| | - Jie Zhou
- Cleveland Clinic Lerner Research Institute, Department of Quantitative Health Sciences, Cleveland, OH,Present address: Department of Neuroscience, Novartis, Cambridge, MA
| | - Brian Hobbs
- Cleveland Clinic Lerner Research Institute, Department of Quantitative Health Sciences, Cleveland, OH,Present address: Department of Population Health, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Nathan A. Pennell
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
15
|
Barbar J, Armach M, Hodroj MH, Assi S, El Nakib C, Chamseddine N, Assi HI. Emerging genetic biomarkers in lung adenocarcinoma. SAGE Open Med 2022; 10:20503121221132352. [PMID: 36277445 PMCID: PMC9583216 DOI: 10.1177/20503121221132352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 09/26/2022] [Indexed: 11/07/2022] Open
Abstract
Comprehensive genomic profiling is a next-generation sequencing approach used to
detect several known and emerging genomic alterations. Many genomic variants
detected by comprehensive genomic profiling have become recognized as
significant cancer biomarkers, leading to the development of major clinical
trials. Lung adenocarcinoma has become one of the most targeted cancers for
genomic profiling with a series of actionable mutations such as EGFR, KRAS,
HER2, BRAF, FGFR, MET, ALK, and many others. The importance of these mutations
lies in establishing targeted therapies that significantly change the outcome in
lung adenocarcinoma besides the prognostic value of some mutations. This review
sheds light on the development of the comprehensive genomic profiling field,
mainly lung adenocarcinoma, and discusses the role of a group of mutations in
this disease.
Collapse
Affiliation(s)
- Jawad Barbar
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Maria Armach
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Mohammad Hassan Hodroj
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Sahar Assi
- Department of Internal Medicine,
American University of Beirut Medical Center, Beirut, Lebanon
| | - Clara El Nakib
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Nathalie Chamseddine
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon
| | - Hazem I Assi
- Department of Internal Medicine,
Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American
University of Beirut Medical Center, Beirut, Lebanon,Hazem I Assi, Department of Internal
Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute,
American University of Beirut Medical Center, P.O. Box: 11-0236, Riad El Solh,
Beirut 1107 2020, Lebanon.
| |
Collapse
|
16
|
Briggs A, Wehler B, Gaultney JG, Upton A, Italiano A, Bokemeyer C, Paracha N, Sullivan SD. Comparison of Alternative Methods to Assess the Cost-Effectiveness of Tumor-Agnostic Therapies: A Triangulation Approach Using Larotrectinib as a Case Study. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2022; 25:1002-1009. [PMID: 35667773 DOI: 10.1016/j.jval.2021.11.1354] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 05/15/2023]
Abstract
OBJECTIVES The study objective was to investigate the economic value of tumor-agnostic therapies when only single-arm effectiveness data are available at launch by applying multiple methodologies to establish comparative effectiveness. METHODS In the absence of direct comparative data, 3 methods were used to estimate the counterfactual: (1) a historical control based on a systematic literature review for each tumor site from the larotrectinib trials, (2) an intracohort comparison using the previous line of therapy time to progression from larotrectinib trials, and (3) a nonresponder control that applied outcomes for larotrectinib nonresponders. Cost-effectiveness was modeled using the partitioned survival approach. Stochastic parameter uncertainty was assessed in a probabilistic sensitivity analysis (PSA). A triangulated estimate of the mean cost-effectiveness result was generated combining all 3 counterfactual estimates. RESULTS Incremental cost-effectiveness ratios were similar across the 3 methodologies in the deterministic analysis ranging from £83 868 (95% uncertainty interval [UI] £65 698-£107 668) to £104 922 per quality-adjusted life-year (95% UI £80 132-£139 658). PSA results for each method substantially overlapped when plotted on the cost-effectiveness plane. Weighting PSA results for each method equally in the triangulation method produced an incremental cost-effectiveness ratios of £95 587 per quality-adjusted life-year gained (95% UI £70 449-£137 431). CONCLUSIONS In the absence of direct comparative data, different methods of estimating a counterfactual are possible, each with strengths and limitations. Triangulating results across the methods provides a composite view of the total uncertainty and a more consistent estimation of the cost-effectiveness of the tumor-agnostic intervention compared with choosing a single method.
Collapse
Affiliation(s)
- Andrew Briggs
- London School of Hygiene and Tropical Medicine, London, England, UK
| | | | | | - Alex Upton
- Bayer Pharmaceuticals, Whippany, NJ, USA.
| | | | | | | | - Sean D Sullivan
- The CHOICE Institute, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
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
|
18
|
Identification of Copy Number Alterations from Next-Generation Sequencing Data. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:55-74. [DOI: 10.1007/978-3-030-91836-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Conroy JM, Pabla S, Glenn ST, Seager RJ, Van Roey E, Gao S, Burgher B, Andreas J, Giamo V, Mallon M, Lee YH, DePietro P, Nesline M, Wang Y, Lenzo FL, Klein R, Zhang S. A scalable high-throughput targeted next-generation sequencing assay for comprehensive genomic profiling of solid tumors. PLoS One 2021; 16:e0260089. [PMID: 34855780 PMCID: PMC8639101 DOI: 10.1371/journal.pone.0260089] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/03/2021] [Indexed: 11/18/2022] Open
Abstract
Timely and accurate identification of molecular alterations in solid tumors is essential for proper management of patients with advanced cancers. This has created a need for rapid, scalable comprehensive genomic profiling (CGP) systems that detect an increasing number of therapeutically-relevant variant types and molecular signatures. In this study, we assessed the analytical performance of the TruSight Oncology 500 High-Throughput assay for detection of somatic alterations from formalin-fixed paraffin-embedded tissue specimens. In parallel, we developed supporting software and automated sample preparation systems designed to process up to 70 clinical samples in a single NovaSeq 6000TM sequencing run with a turnaround time of <7 days from specimen receipt to report. The results demonstrate that the scalable assay accurately and reproducibly detects small variants, copy number alterations, microsatellite instability (MSI) and tumor mutational burden (TMB) from 40ng DNA, and multiple gene fusions, including known and unknown partners and splice variants from 20ng RNA. 717 tumor samples and reference materials with previously known alterations in 96 cancer-related genes were sequenced to evaluate assay performance. All variant classes were reliably detected at consistent and reportable variant allele percentages with >99% overall accuracy and precision. Our results demonstrate that the high-throughput CGP assay is a reliable method for accurate detection of molecular alterations in support of precision therapeutics in oncology. The supporting systems and scalable workflow allow for efficient interpretation and prompt reporting of hundreds of patient cancer genomes per week with excellent analytical performance.
Collapse
Affiliation(s)
- Jeffrey M. Conroy
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
- Research Support Services, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - Sarabjot Pabla
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Sean T. Glenn
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
- Laboratory Operations, OmniSeq Inc., Buffalo, New York, United States of America
- HemePath Molecular, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States of America
| | - R. J. Seager
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Erik Van Roey
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Shuang Gao
- Bioinformatics, OmniSeq Inc., Buffalo, New York, United States of America
| | - Blake Burgher
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Jonathan Andreas
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Vincent Giamo
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Melissa Mallon
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Yong Hee Lee
- Clinical Evidence Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Paul DePietro
- Clinical Evidence Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Mary Nesline
- Clinical Evidence Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Yirong Wang
- Information Technology, OmniSeq Inc., Buffalo, New York, United States of America
| | - Felicia L. Lenzo
- Research and Development, OmniSeq Inc., Buffalo, New York, United States of America
| | - Roger Klein
- Medical Affairs, OmniSeq Inc., Buffalo, New York, United States of America
| | - Shengle Zhang
- Laboratory Operations, OmniSeq Inc., Buffalo, New York, United States of America
| |
Collapse
|
20
|
Universal Germline Testing of Unselected Cancer Patients Detects Pathogenic Variants Missed by Standard Guidelines without Increasing Healthcare Costs. Cancers (Basel) 2021; 13:cancers13225612. [PMID: 34830767 PMCID: PMC8616044 DOI: 10.3390/cancers13225612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/14/2021] [Accepted: 10/31/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Clinical genetic testing likely underestimates the frequency of pathogenic germline variants (PGVs) in a cancer patient population due to strict qualifications designated by practice guidelines. Accurate ascertainment of PGVs in cancer patients can be paramount to the treatment of hereditary cancer syndromes. Our prospective study aimed to (1) elucidate PGV frequency in an unselected cohort of cancer patients by offering universal germline testing regardless of eligibility by clinical guidelines and to (2) evaluate the impact of receiving such testing on cost of care. Results confirmed that over 50% of patients who harbored a PGV would not have qualified for testing under current guidelines and there was no increase in healthcare costs for patients who tested positive for a PGV. It is therefore feasible to offer universal germline testing for cancer patients in the clinic to detect PGVs without increasing healthcare costs. Abstract Purpose: To accurately ascertain the frequency of pathogenic germline variants (PGVs) in a pan-cancer patient population with universal genetic testing and to assess the economic impact of receiving genetic testing on healthcare costs. Methods: In this prospective study, germline genetic testing using a 105-gene panel was administered to an unselected pan-cancer patient population irrespective of eligibility by current guidelines. Financial records of subjects were analyzed to assess the effect of PGV detection on cost of care one year from the date of testing. Results: A total of 284 patients participated in this study, of which 44 patients (15%) tested positive for a PGV in 14 different cancer types. Of the patients with PGVs, 23 patients (52%) were ineligible for testing by current guidelines. Identification of a PGV did not increase cost of care. Conclusion: Implementation of universal genetic testing for cancer patients in the clinic, beyond that specified by current guidelines, is necessary to accurately assess and treat hereditary cancer syndromes and does not increase healthcare costs.
Collapse
|
21
|
Li DY, Yang F, Liao WQ, Zhou XF, Li WB, Cai JR, Liu BL, Luo Y, Zhan HL. Deep Genomic Sequencing of Bladder Urothelial Carcinoma in Southern Chinese Patients: A Single-Center Study. Front Oncol 2021; 11:538927. [PMID: 34055593 PMCID: PMC8160294 DOI: 10.3389/fonc.2021.538927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/21/2021] [Indexed: 12/31/2022] Open
Abstract
Objective Bladder urothelial carcinoma (BUC) is a common urological malignancy with molecular heterogeneity. However, the genetic feature of Chinese BUC patients is still not well-identified. Methods We performed deep sequencing by a large panel (450 genes) on 22 BUC samples and using matched normal bladder tissue as control. Genomic alterations (GAs), pathways and Tumor Mutation Burden (TMB) were investigated. Results The frequencies of GAs (TERT, 54.5%; CREBBP, 27.3%; GATA3, 22.7%; BRAF, 18.2%; TEK, 18.2% and GLI1, 18.2%) were significantly higher in Chinese than Western BUC patients. Other GAs' frequencies were in accordance with previous study (TP53, 50.0%; KDM6A, 31.8%; KMT2D, 22.7%; etc.). Besides, we detected gene amplification in ERBB2, FRS2, FAS, etc. The gene fusion/rearrangement took place in the chromosome 11, 12, 14, 17, 19, 22, and Y. Other than cell cycle and PI3K-AKT-mTOR, mutated genes were more associated with the transcription factor, chromatin modification signaling pathways. Interestingly, the TMB value was significantly higher in the BUC patients at stages T1-T2 than T3-T4 (P = 0.025). Conclusion Deep genomic sequencing of BUC can provide new clues on the unique GAs of Chinese patients and assist in therapeutic decision.
Collapse
Affiliation(s)
- Dong-Yang Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fei Yang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wei-Qiang Liao
- Department of Urology, Luoding People's Hospital, Luoding, China
| | - Xiang-Fu Zhou
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wen-Biao Li
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jia-Rong Cai
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bo-Long Liu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hai-Lun Zhan
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
22
|
Freidlin B, Allegra CJ, Korn EL. Moving Molecular Profiling to Routine Clinical Practice: A Way Forward? J Natl Cancer Inst 2021; 112:773-778. [PMID: 31868907 DOI: 10.1093/jnci/djz240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 01/09/2023] Open
Abstract
Molecular profiling of a patient's tumor to guide targeted treatment selection offers the potential to advance patient care by improving outcomes and minimizing toxicity (by avoiding ineffective treatments). However, current development of molecular profile (MP) panels is often based on applying institution-specific or subjective algorithms to nonrandomized patient cohorts. Consequently, obtaining reliable evidence that molecular profiling is offering clinical benefit and is ready for routine clinical practice is challenging. In particular, we discuss here the problems with interpreting for clinical utility nonrandomized studies that compare outcomes in patients treated based on their MP vs those treated with standard of care, studies that compare the progression-free survival (PFS) seen on a MP-directed treatment to the PFS seen for the same patient on a previous standard treatment (PFS ratio), and multibasket trials that evaluate the response rates of targeted therapies in specific molecularly defined subpopulations (regardless of histology). We also consider some limitations of randomized trial designs. A two-step strategy is proposed in which multiple mutation-agent pairs are tested for activity in one or more multibasket trials in the first step. The results of the first step are then used to identify promising mutation-agent pairs that are combined in a molecular panel that is then tested in the step-two strategy-design randomized clinical trial (the molecular panel-guided treatment for the selected mutations vs standard of care). This two-step strategy should allow rigorous evidence-driven identification of mutation-agent pairs that can be moved into routine clinical practice.
Collapse
Affiliation(s)
- Boris Freidlin
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr, Bethesda, MD 20892, USA
| | - Carmen J Allegra
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr, Bethesda, MD 20892, USA.,Division of Hematology and Oncology, Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608, USA
| | - Edward L Korn
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
John A, Yang B, Shah R. Clinical Impact of Adherence to NCCN Guidelines for Biomarker Testing and First-Line Treatment in Advanced Non-Small Cell Lung Cancer (aNSCLC) Using Real-World Electronic Health Record Data. Adv Ther 2021; 38:1552-1566. [PMID: 33537872 PMCID: PMC7932942 DOI: 10.1007/s12325-020-01617-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/23/2020] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Although clinical guidelines are broadly available, the relationship between adherence and outcomes is not well studied. This study aimed to assess the association between adherence to National Comprehensive Cancer Network (NCCN) guidelines and clinical outcomes for adult patients with advanced non-small-cell lung cancer (aNSCLC). METHODS This was a retrospective cohort study of adult patients with aNSCLC (stages IIIB, IIIC, and IV) from a de-identified real-world database. The objective was accomplished in a two-step analysis process. We first assessed adherence to NCCN recommendations for biomarker testing and overall survival (OS). Next, we assessed adherence to NCCN-recommended first-line therapy and time to treatment discontinuation (TTD). Multivariable Cox regression analyses were conducted to evaluate the association between guideline adherence and patient outcomes. Kaplan-Meier analyses were used to assess median OS and TTD. RESULTS A total of 28,784 patients with a diagnosis for aNSCLC between January 1, 2011 and July 31, 2019 met the inclusion criteria for the analysis of NCCN-recommended biomarker testing adherence. Two-thirds of these patients (n = 19,787) had evidence of biomarker testing (adherent). Multivariable Cox models found that testing-adherent patients had a significantly lower risk of mortality [hazard ratio (HR) = 0.89, 95% confidence interval (CI) 0.86, 0.92; p < 0.01]. Median OS was modestly longer in the testing-adherent group compared to the testing-non-adherent group (15.4 vs. 14.2 months; p < 0.01). For the first-line therapy analysis, 15,898 patients met the inclusion criteria, of which 69.9% had evidence of appropriate first-line therapy (first-line-adherent). The multivariable Cox model found that adherent patients had significantly lower risk of treatment discontinuation versus non-adherent patients (HR = 0.60, 95% CI 0.57, 0.62; p < 0.01). First-line-adherent patients had a modest, yet significantly longer median TTD compared to first-line-non-adherent patients (3.45 vs. 2.40 months; p < 0.01). CONCLUSIONS Improved clinical outcomes were observed in patients who were adherent to NCCN-recommended biomarker testing and first-line therapy. This study demonstrated the value of following NCCN guideline recommendations and the need to prioritize timely access to biomarker testing and individualized treatment.
Collapse
Affiliation(s)
- Ani John
- Roche Diagnostics, Santa Clara, CA, USA.
| | | | - Roma Shah
- Roche Diagnostics, Santa Clara, CA, USA
| |
Collapse
|
24
|
Chen AP, Kummar S, Moore N, Rubinstein LV, Zhao Y, Williams PM, Palmisano A, Sims D, O'Sullivan Coyne G, Rosenberger CL, Simpson M, Raghav KPS, Meric-Bernstam F, Leong S, Waqar S, Foster JC, Konaté MM, Das B, Karlovich C, Lih CJ, Polley E, Simon R, Li MC, Piekarz R, Doroshow JH. Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT): A Randomized Multicenter Phase II Trial. JCO Precis Oncol 2021; 5:PO.20.00372. [PMID: 33928209 PMCID: PMC8078898 DOI: 10.1200/po.20.00372] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
This trial assessed the utility of applying tumor DNA sequencing to treatment selection for patients with advanced, refractory cancer and somatic mutations in one of four signaling pathways by comparing the efficacy of four study regimens that were either matched to the patient's aberrant pathway (experimental arm) or not matched to that pathway (control arm). MATERIALS AND METHODS Adult patients with an actionable mutation of interest were randomly assigned 2:1 to receive either (1) a study regimen identified to target the aberrant pathway found in their tumor (veliparib with temozolomide or adavosertib with carboplatin [DNA repair pathway], everolimus [PI3K pathway], or trametinib [RAS/RAF/MEK pathway]), or (2) one of the same four regimens, but chosen from among those not targeting that pathway. RESULTS Among 49 patients treated in the experimental arm, the objective response rate was 2% (95% CI, 0% to 10.9%). One of 20 patients (5%) in the experimental trametinib cohort had a partial response. There were no responses in the other cohorts. Although patients and physicians were blinded to the sequencing and random assignment results, a higher pretreatment dropout rate was observed in the control arm (22%) compared with the experimental arm (6%; P = .038), suggesting that some patients may have had prior tumor mutation profiling performed that led to a lack of participation in the control arm. CONCLUSION Further investigation, better annotation of predictive biomarkers, and the development of more effective agents are necessary to inform treatment decisions in an era of precision cancer medicine. Increasing prevalence of tumor mutation profiling and preference for targeted therapy make it difficult to use a randomized phase II design to evaluate targeted therapy efficacy in an advanced disease setting.
Collapse
Affiliation(s)
- Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Nancy Moore
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - P. Mickey Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Alida Palmisano
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- General Dynamics Information Technology (GDIT), Falls Church, VA
| | - David Sims
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Mel Simpson
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kanwal P. S. Raghav
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Funda Meric-Bernstam
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Saiama Waqar
- Department of Medical Oncology, Washington University School of Medicine, St Louis, MO
| | - Jared C. Foster
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Mariam M. Konaté
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chris Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chih-Jian Lih
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Eric Polley
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Richard Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Ming-Chung Li
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Richard Piekarz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
25
|
Bonanno L, Pavan A, Ferro A, Calvetti L, Frega S, Pasello G, Aprile G, Guarneri V, Conte P. Clinical Impact of Plasma and Tissue Next-Generation Sequencing in Advanced Non-Small Cell Lung Cancer: A Real-World Experience. Oncologist 2020; 25:e1996-e2005. [PMID: 32557976 PMCID: PMC8108051 DOI: 10.1634/theoncologist.2020-0148] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Targeted agents have improved the outcome of a subset of non-small cell lung cancer (NSCLC). Molecular profiling by next-generation sequencing (NGS) allows screening for multiple genetic alterations both in tissue and in plasma, but limited data are available concerning its feasibility and impact in real-world clinical practice. METHODS Patients with advanced NSCLC consecutively referring to our Institution for potential eligibility to VISION trial (NCT02864992) were prospectively enrolled. They were already screened with standard method, and EGFR/ALK/ROS-1 positive cases were excluded. NGS was performed in plasma and tissue using the Guardant360 test covering 73 genes and the Oncomine Focus Assay covering 59 genes, respectively. RESULTS The study included 235 patients. NGS was performed in plasma in 209 (88.9%) cases; 78 of these (37.3%) were evaluated also in tissue; tissue only was analyzed in 26 cases (11.1%). Half of the tissue samples were deemed not evaluable. Druggable alterations were detected in 13 (25%) out of 52 evaluable samples and 31 of 209 (14.8%) of plasma samples. Improved outcome was observed for patients with druggable alterations if treated with matched targeted agents: they had a longer median overall survival (not reached) compared with the ones who did not start any targeted therapy (9.1 months; 95% confidence interval, 4.6-13.6; p = .046). The results of NGS testing potentially also affected the outcome of patients treated with immunotherapy. CONCLUSION Systematic real-life NGS testing showed the limit of tissue analysis in NSCLC and highlighted the potentiality of genetic characterization in plasma in increasing the number of patients who may benefit from NGS screening, both influencing the clinical decision-making process and affecting treatment outcome. IMPLICATIONS FOR PRACTICE Genetic characterization of cancer has become more important with time, having had positive implications for treatment specificity and efficacy. Such analyses changed the natural history of advanced non-small cell lung cancer (aNSCLC) with the introduction of drugs targeted to specific gene alterations (e.g., EGFR mutations, ALK and ROS-1 rearrangements). In the field of cancer molecular characterization, the applicability of the analysis of a wide panel of genes using a high-throughput sequencing approach, such as next-generation sequencing (NGS), is still a matter of research. This study used NGS in a real-world setting to systematically and prospectively profile patients with aNSCLC. The aim was to evaluate its feasibility and reliability, as well as consequent access to targeted agents and impact on clinical outcome whenever a druggable alteration was detected either in tumor tissue samples or through liquid biopsy.
Collapse
Affiliation(s)
- Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
| | - Alberto Pavan
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
| | - Alessandra Ferro
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
- Department of Surgery, Oncology, and Gastroenterology, University of PadovaPadovaItaly
| | - Lorenzo Calvetti
- Department of Oncology, San Bortolo General HospitalUnità Locale Socio Sanitaria (ULSS) 8 Berica—East DistrictVicenzaItaly
| | - Stefano Frega
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
| | - Giulia Pasello
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General HospitalUnità Locale Socio Sanitaria (ULSS) 8 Berica—East DistrictVicenzaItaly
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
- Department of Surgery, Oncology, and Gastroenterology, University of PadovaPadovaItaly
| | - PierFranco Conte
- Medical Oncology 2, Istituto Oncologico Veneto (IOV)–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)PadovaItaly
- Department of Surgery, Oncology, and Gastroenterology, University of PadovaPadovaItaly
| | | |
Collapse
|
26
|
Bollati V, Ferrari L, Leso V, Iavicoli I. Personalised Medicine: implication and perspectives in the field of occupational health. LA MEDICINA DEL LAVORO 2020; 111:425-444. [PMID: 33311418 PMCID: PMC7809984 DOI: 10.23749/mdl.v111i6.10947] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
"Personalised medicine" relies on identifying and integrating individual variability in genomic, biological, and physiological parameters, as well as in environmental and lifestyle factors, to define "individually" targeted disease prevention and treatment. Although innovative "omic" technologies supported the application of personalised medicine in clinical, oncological, and pharmacological settings, its role in occupational health practice and research is still in a developing phase. Occupational personalised approaches have been currently applied in experimental settings and in conditions of unpredictable risks, e.g.. war missions and space flights, where it is essential to avoid disease manifestations and therapy failure. However, a debate is necessary as to whether personalized medicine may be even more important to support a redefinition of the risk assessment processes taking into consideration the complex interaction between occupational and individual factors. Indeed, "omic" techniques can be helpful to understand the hazardous properties of the xenobiotics, dose-response relationships through a deeper elucidation of the exposure-disease pathways and internal doses of exposure. Overall, this may guide the adoption/implementation of primary preventive measures protective for the vast majority of the population, including most susceptible subgroups. However, the application of personalised medicine into occupational health requires overcoming some practical, ethical, legal, economical, and socio-political issues, particularly concerning the protection of privacy, and the risk of discrimination that the workers may experience. In this scenario, the concerted action of academic, industry, governmental, and stakeholder representatives should be encouraged to improve research aimed to guide effective and sustainable implementation of personalised medicine in occupational health fields.
Collapse
Affiliation(s)
- Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Italy.
| | - Luca Ferrari
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Italy.
| | - Veruscka Leso
- Section of Occupational Medicine, Department of Public Health, Università degli Studi di Napoli Federico II, Napoli, Italy.
| | - Ivo Iavicoli
- Section of Occupational Medicine, Department of Public Health, Università degli Studi di Napoli Federico II, Napoli, Italy.
| |
Collapse
|
27
|
Hsiao SJ, Sireci AN, Pendrick D, Freeman C, Fernandes H, Schwartz GK, Henick BS, Mansukhani MM, Roth KA, Carvajal RD, Oberg JA. Clinical Utilization, Utility, and Reimbursement for Expanded Genomic Panel Testing in Adult Oncology. JCO Precis Oncol 2020; 4:1038-1048. [DOI: 10.1200/po.20.00048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The routine use of large next-generation sequencing (NGS) pan-cancer panels is required to identify the increasing number of, but often uncommon, actionable alterations to guide therapy. Inconsistent coverage and variable payment is hindering NGS adoption into clinical practice. A review of test utilization, clinical utility, coverage, and reimbursement was conducted in a cohort of patients diagnosed with high-risk cancer who received pan-cancer panel testing as part of their clinical care. MATERIALS AND METHODS The Columbia Combined Cancer Panel (CCCP), a 467-gene panel designed to detect DNA variations in solid and liquid tumors, was performed in the Laboratory of Personalized Genomic Medicine at Columbia University Irving Medical Center. Utilization was characterized at test order. Results were reviewed by a molecular pathologist, followed by a multidisciplinary molecular tumor board where clinical utility was classified by consensus. Reimbursement was reviewed after payers provided final coverage decisions. RESULTS NGS was performed on 359 high-risk tumors from 349 patients. Reimbursement data were available for 246 cases. The most common reason providers ordered CCCP testing was for patients diagnosed with a treatment-resistant or recurrent tumor (n = 214; 61%). Findings were clinically impactful for 229 cases (64%). Molecular alterations that may inform future therapy in the event of progression or relapse were found in 42% of cases, and a targeted therapy was initiated in 23 cases (6.6%). The majority of tests were denied coverage by payers (n = 190; 77%). On average, insurers reimbursed 10.75% of the total NGS service charge. CONCLUSION CCCP testing identified clinically impactful alterations in 64% of cases. Limited coverage and low reimbursement remain barriers, and broader reimbursement policies are needed to adopt pan-cancer NGS testing that benefits patients into clinical practice.
Collapse
Affiliation(s)
- Susan J. Hsiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Anthony N. Sireci
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Danielle Pendrick
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Christopher Freeman
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Helen Fernandes
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Gary K. Schwartz
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Brian S. Henick
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Mahesh M. Mansukhani
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Kevin A. Roth
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Richard D. Carvajal
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Jennifer A. Oberg
- Division of Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
28
|
John A, Shah RA, Wong WB, Schneider CE, Alexander M. Value of Precision Medicine in Advanced Non-Small Cell Lung Cancer: Real-World Outcomes Associated with the Use of Companion Diagnostics. Oncologist 2020; 25:e1743-e1752. [PMID: 32627882 PMCID: PMC7648341 DOI: 10.1634/theoncologist.2019-0864] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 06/18/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Companion diagnostic (CDx) testing for patients with advanced non-small cell lung cancer (aNSCLC) identifies patients more likely to benefit from biomarker-driven treatments. METHODS Patients with nonsquamous cell (non-Sq) aNSCLC from the Flatiron Health database (diagnosed January 1, 2011-May 31, 2018) who had CDx testing were compared with those who had no reported evidence of testing. The association between CDx testing and overall survival was evaluated by unadjusted and adjusted Cox proportional hazards regression models. Logistic regression analysis identified characteristics associated with CDx testing. The revised modified Lung Cancer Prognostic Index and other factors identified a priori were included in the adjusted models. RESULTS A total of 17,555 patients with non-Sq aNSCLC (CDx, n = 14,732; no CDx, n = 2,823) with mean ± SD age of 67.2 ± 10.0 years were included. Most were insured (91.7%) and white (67.1%). Asian patients and those who were never-smokers were more likely to undergo CDx testing. Those with CDx testing lived longer than those without (median [95% confidence interval (CI)] survival, 13.04 [12.62-13.40] vs. 6.01 [5.72-6.24] months) and had a decreased mortality risk (adjusted hazard ratio [95% CI], 0.72 [0.69-0.76]). A survival advantage was also seen for patients with CDx testing who received biomarker-driven first-line therapy. CONCLUSION Patients with non-Sq aNSCLC who had CDx testing had a greater survival benefit than those without, supporting broader use of CDx testing in routine clinical practice to identify patients more likely to benefit from precision medicine. IMPLICATIONS FOR PRACTICE Companion diagnostic (CDx) testing coupled with biomarker-driven treatment offers a greater survival benefit for patients with advanced non-small cell lung cancer (aNSCLC). In this study, patients with nonsquamous aNSCLC from Flatiron Health, a large, real-world oncology database, with CDx testing had a reduced mortality risk and lived longer than patients without reported evidence of CDx testing; those who received biomarker-driven therapy as their first line of treatment were likely to survive three times longer than those who did not. These results demonstrate the clinical utility of CDx testing as the first step in treating nonsquamous aNSCLC in real-world clinical practice.
Collapse
Affiliation(s)
- Ani John
- Roche Diagnostics Information SolutionsPleasanton, CaliforniaUSA
| | - Roma A. Shah
- Roche Diagnostics Information SolutionsPleasanton, CaliforniaUSA
| | | | | | - Marliese Alexander
- Pharmacy Department, Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of Oncology, University of MelbourneVictoriaAustralia
| |
Collapse
|
29
|
Zhang Q, Fu Q, Bai X, Liang T. Molecular Profiling-Based Precision Medicine in Cancer: A Review of Current Evidence and Challenges. Front Oncol 2020; 10:532403. [PMID: 33194591 PMCID: PMC7652987 DOI: 10.3389/fonc.2020.532403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Matched therapy based on next-generation sequencing is now a part of routine care to guide the treatment of patients with advanced solid tumors. However, whether and to what extent patients can benefit from this strategy on a large scale remains uncertain. In the past decade, several clinical studies were performed in this field, among which only one was a randomized trial. We reviewed the literature on this topic and summarize the existing data about the efficacy of this treatment strategy. Currently, the evidence is promising but not solid. Multiple ongoing trials are also summarized. We also discuss the limitations of this treatment strategy and certain unsolved important problems, including how to select the sample and target level, how to interpret the results, and the problem of drug accessibility. All these issues should receive more attention in future clinical trial design and the application of target therapy in cancer treatment.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Qihan Fu
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| |
Collapse
|
30
|
Ferreira-Gonzalez A. Plasma PIK3CA Mutation Testing in Advanced Breast Cancer Patients for Personalized Medicine: A Value Proposition. J Appl Lab Med 2020; 5:1076-1089. [PMID: 32901282 DOI: 10.1093/jalm/jfaa117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Even though endocrine therapy is often initially successful in treating advanced breast cancer, most patients inevitably face disease progression. In advanced hormone receptor-positive (HR+) breast cancer, activation of the PI3K downstream pathway is a critical feature of the mechanism of endocrine resistance. A significant recent advance in treating HR+ advanced breast cancer has been the recent introduction of PI3K inhibitor (PI3Ki) for the treatment of patients with HR+, HER2-negative (HER2-) advanced or metastatic breast cancer that harbors PIK3CA mutations. A value proposition concept was applied to assess the potential benefits of cell-free tumor DNA (ctDNA) testing to identify patients who might respond to PI3Ki treatment. CONTENT By applying the framework of the value proposition to >35 publications, in addition to recommendations from professional organizations, it was evident that robust clinical evidence exists to support the role of ctDNA PIK3CA mutation evaluation in identifying patients with advanced breast cancer who could benefit from PI3Ki treatment. SUMMARY Detection of PIK3CA gene mutations in HR+HER2- advanced breast cancer patients allows for the identification of patients who might benefit from more effective personalized treatment with molecularly targeted drugs.
Collapse
|
31
|
Tsimberidou AM, Elkin S, Dumanois R, Pritchard D. Clinical and Economic Value of Genetic Sequencing for Personalized Therapy in Non-small-cell Lung Cancer. Clin Lung Cancer 2020; 21:477-481. [PMID: 32718774 DOI: 10.1016/j.cllc.2020.05.029] [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: 01/21/2020] [Revised: 05/12/2020] [Accepted: 05/26/2020] [Indexed: 11/17/2022]
Abstract
Two recent studies examining the clinical and economic value of next-generation sequencing (NGS)-based diagnostic testing (multi-gene panel examining ≥ 30 genes) for non-small-cell lung cancer therapy compared with single gene ALK, EGFR testing to select therapy demonstrated statistically insignificant improvement in population-level overall survival and only a moderate incremental cost-effectiveness ratio associated with the NGS testing approach. The data, however, revealed a key practice gap: many patients with actionable mutations did not receive targeted therapies. This gap is attributed, in part, to limitations in the availability and interpretation of NGS results, sample processing constraints, limited access to targeted therapies, and lagging awareness of the rapidly evolving field of personalized medicine, all of which result in "clinical inertia," (ie, suboptimal use of targeted therapy against an actionable driver alteration identified by NGS testing). Additional analysis estimated that cost-effectiveness would improve significantly if a higher percentage of patients received testing and if all patients who were eligible for targeted therapies received them. Strategies to address implementation barriers will help to realize the full value of NGS testing in cancer care.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | | | | | | |
Collapse
|
32
|
Melas M, Subbiah S, Saadat S, Rajurkar S, McDonnell KJ. The Community Oncology and Academic Medical Center Alliance in the Age of Precision Medicine: Cancer Genetics and Genomics Considerations. J Clin Med 2020; 9:E2125. [PMID: 32640668 PMCID: PMC7408957 DOI: 10.3390/jcm9072125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Recent public policy, governmental regulatory and economic trends have motivated the establishment and deepening of community health and academic medical center alliances. Accordingly, community oncology practices now deliver a significant portion of their oncology care in association with academic cancer centers. In the age of precision medicine, this alliance has acquired critical importance; novel advances in nucleic acid sequencing, the generation and analysis of immense data sets, the changing clinical landscape of hereditary cancer predisposition and ongoing discovery of novel, targeted therapies challenge community-based oncologists to deliver molecularly-informed health care. The active engagement of community oncology practices with academic partners helps with meeting these challenges; community/academic alliances result in improved cancer patient care and provider efficacy. Here, we review the community oncology and academic medical center alliance. We examine how practitioners may leverage academic center precision medicine-based cancer genetics and genomics programs to advance their patients' needs. We highlight a number of project initiatives at the City of Hope Comprehensive Cancer Center that seek to optimize community oncology and academic cancer center precision medicine interactions.
Collapse
Affiliation(s)
- Marilena Melas
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA;
| | - Shanmuga Subbiah
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Glendora, CA 91741, USA;
| | - Siamak Saadat
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Colton, CA 92324, USA;
| | - Swapnil Rajurkar
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Upland, CA 91786, USA;
| | - Kevin J. McDonnell
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA 91010, USA
- Center for Precision Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
33
|
Burnett-Hartman AN, Udaltsova N, Kushi LH, Neslund-Dudas C, Rahm AK, Pawloski PA, Corley DA, Knerr S, Feigelson HS, Hunter JE, Tabano DC, Epstein MM, Honda SA, Ter-Minassian M, Lynch JA, Lu CY. Clinical Molecular Marker Testing Data Capture to Promote Precision Medicine Research Within the Cancer Research Network. JCO Clin Cancer Inform 2020; 3:1-10. [PMID: 31487201 DOI: 10.1200/cci.19.00026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE To evaluate health care systems for the availability of population-level data on the frequency of use and results of clinical molecular marker tests to inform precision cancer care. METHODS We assessed cancer-related molecular marker test data availability across 12 US health care systems in the Cancer Research Network. Overall, these systems provide care to a diverse population of more than 12 million people in the United States. We performed qualitative analyses of test data availability for five blood-based protein, nine germline, and 14 tissue-based tumor marker tests in each health care system's electronic health record and tumor registry using key informants, test code lists, and manual review of data types and output. We then performed quantitative analyses to estimate the proportion of patients with cancer with test utilization data and results for specific molecular marker tests. RESULTS Health systems were able to systematically capture population-level data on all five blood protein markers, six of 14 tissue-based tumor markers, and none of the nine germline markers. Successful, systematic data capture was achievable for tests with electronic data feeds for test results (blood protein markers) or through prior manual abstraction by tumor registrars (select tumor-based markers). For test results stored in scanned image files (particularly germline and tumor marker tests), information on which test was performed and test results was not readily accessible in an electronic format. CONCLUSION Even in health care systems with sophisticated electronic health records, there were few codified data elements available for evaluating precision cancer medicine test use and results at the population level. Health care organizations should establish standards for electronic reporting of precision medicine tests to expedite cancer research and facilitate the implementation of precision medicine approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sarah Knerr
- University of Washington and Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | | | | | - David C Tabano
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO
| | - Mara M Epstein
- University of Massachusetts Medical School, Worcester, MA
| | | | | | - Julie A Lynch
- Department of Veterans Affairs Salt Lake City Health System, Salt Lake City, UT
| | - Christine Y Lu
- Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| |
Collapse
|
34
|
Jones SF, McKenzie AJ. Molecular Profiling in Drug Development: Paving a Way Forward. Am Soc Clin Oncol Educ Book 2020; 40:309-318. [PMID: 32463698 DOI: 10.1200/edbk_100024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
As researchers learn more about tumor biology and the molecular mechanisms involved in tumorigenesis, metastasis, and tumor evolution, clinical trials are growing more complex and patient selection for clinical trials is becoming more specific. Rather than exploit certain phenotypic characteristics of tumor cells (e.g., rapid cell division and uncontrolled cell growth), pharmaceuticals targeting the genotypic causes of tumorigenesis are emerging. The sequencing of the human genome, advances in chemical techniques, and increased efficiency in drug target identification have changed the way drugs are developed. Now, more precise drugs targeting specific mutations within individual genes are being used to treat narrow patient populations harboring these specific driver mutations, often with greater efficacy and lower toxicity than traditional chemotherapeutic agents. This precision in drug development relies not only on the ability to design exquisitely specific pharmaceuticals but also to identify (with the same level of precision) the patients who are most likely to respond to those therapies. Robust screening techniques and adequate molecular oncology education are required to match the appropriate patient to precision therapies, and these same screening techniques provide the data necessary to advance to the next generation of drug development.
Collapse
|
35
|
Karami E, Ruschin M, Soliman H, Sahgal A, Stanisz GJ, Sadeghi-Naini A. An MR Radiomics Framework for Predicting the Outcome of Stereotactic Radiation Therapy in Brain Metastasis. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:1022-1025. [PMID: 31946067 DOI: 10.1109/embc.2019.8856558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Despite recent advances in cancer treatment, patients with brain metastasis still suffer from poor overall survival (OS) after standard treatment. Predicting the treatment outcome before or early after the treatment can potentially assist the physicians in improving the therapy outcome by adjusting a standard treatment on an individual patient basis. In this study, a data-driven computational framework was proposed and investigated to predict the local control/failure (LC/LF) outcome in patients with brain metastasis treated with hypo-fractionated stereotactic radiation therapy (SRT). The framework extracted several geometrical and textural features from the magnetic resonance (MR) images of the tumour and edema regions acquired for 38 patients. Subsequent to a multi-step feature reduction/selection, a quantitative MR biomarker consisting of two features was constructed. A support vector machine classifier was used for outcome prediction using the constructed MR biomarker. The bootstrap .632+ and leave-one-patient-out cross-validation methods were used to assess the model's performance. The results indicated that the outcome of LF after SRT could be predicted with an area under the curve of 0.80 and a cross-validated accuracy of 82%. The results obtained implied a good potential of the proposed framework for local outcome prediction in patients with brain metastasis treated with SRT and encourage further investigations on a larger cohort of patients.
Collapse
|
36
|
Li X, Warner JL. A Review of Precision Oncology Knowledgebases for Determining the Clinical Actionability of Genetic Variants. Front Cell Dev Biol 2020; 8:48. [PMID: 32117976 PMCID: PMC7026022 DOI: 10.3389/fcell.2020.00048] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/20/2020] [Indexed: 01/25/2023] Open
Abstract
The increased availability of tumor genetic testing and targeted cancer therapies contributes to the advancement of precision medicine in the field of oncology. Precision oncology knowledgebases provide a way of organizing clinically relevant genetic information in a way that is easily accessible for both oncologists and patients, facilitating the genetic-based clinical decision making. Many organizations and companies have built precision oncology knowledgebases, intended for multiple users. In general, these knowledgebases offer information on cancer-related genetic variants as well as their associated diagnostic, prognostic, and therapeutic implications, but they often differ in their information curations, designs, and user experiences. It is advisable that oncologists use multiple knowledgebases during their practice to have them complement each other. In the future, convergence toward common standards and formats is needed to ensure that the comprehensive knowledge across all sources can be unified to bring the oncology community closer to the achievement of the goal of precision oncology.
Collapse
Affiliation(s)
- Xuanyi Li
- Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Jeremy L. Warner
- Department of Medicine, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
37
|
Mambetsariev I, Wang Y, Chen C, Nadaf S, Pharaon R, Fricke J, Amanam I, Amini A, Bild A, Chu P, Erhunmwunsee L, Kim J, Munu J, Pillai R, Raz D, Sampath S, Vora L, Qiu F, Smith L, Batra SK, Massarelli E, Koczywas M, Reckamp K, Salgia R. Precision medicine and actionable alterations in lung cancer: A single institution experience. PLoS One 2020; 15:e0228188. [PMID: 32045431 PMCID: PMC7012442 DOI: 10.1371/journal.pone.0228188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Oncology has become more reliant on new testing methods and a greater use of electronic medical records, which provide a plethora of information available to physicians and researchers. However, to take advantage of vital clinical and research data for precision medicine, we must initially make an effort to create an infrastructure for the collection, storage, and utilization of this information with uniquely designed disease-specific registries that could support the collection of a large number of patients. MATERIALS AND METHODS In this study, we perform an in-depth analysis of a series of lung adenocarcinoma patients (n = 415) with genomic and clinical data in a recently created thoracic patient registry. RESULTS Of the 415 patients with lung adenocarcinoma, 59% (n = 245) were female; the median age was 64 (range, 22-92) years with a median OS of 33.29 months (95% CI, 29.77-39.48). The most common actionable alterations were identified in EGFR (n = 177/415 [42.7%]), ALK (n = 28/377 [7.4%]), and BRAF V600E (n = 7/288 [2.4%]). There was also a discernible difference in survival for 222 patients, who had an actionable alteration, with a median OS of 39.8 months as compared to 193 wild-type patients with a median OS of 26.0 months (P<0.001). We identified an unprecedented number of actionable alterations [53.5% (222/415)], including distinct individual alteration rates, as compared with 15.0% and 22.3% in TCGA and GENIE respectively. CONCLUSION The use of patient registries, focused genomic panels and the appropriate use of clinical guidelines in community and academic settings may influence cohort selection for clinical trials and improve survival outcomes.
Collapse
Affiliation(s)
- Isa Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Yingyu Wang
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Chen Chen
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Sorena Nadaf
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Rebecca Pharaon
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Idoroenyi Amanam
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Arya Amini
- Department of Radiation Oncology, City of Hope, Duarte, California, United States of America
| | - Andrea Bild
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Peiguo Chu
- Department of Pathology, City of Hope, Duarte, California, United States of America
| | - Loretta Erhunmwunsee
- Department of Thoracic Surgery, City of Hope, Duarte, California, United States of America
| | - Jae Kim
- Department of Thoracic Surgery, City of Hope, Duarte, California, United States of America
| | - Janet Munu
- Center for Informatics, City of Hope, Duarte, California, United States of America
| | - Raju Pillai
- Department of Pathology, City of Hope, Duarte, California, United States of America
| | - Dan Raz
- Department of Thoracic Surgery, City of Hope, Duarte, California, United States of America
| | - Sagus Sampath
- Department of Radiation Oncology, City of Hope, Duarte, California, United States of America
| | - Lalit Vora
- Department of Diagnostic Radiology, City of Hope, Duarte, California, United States of America
| | - Fang Qiu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Lynette Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Erminia Massarelli
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Marianna Koczywas
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Karen Reckamp
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California, United States of America
| |
Collapse
|
38
|
Karami E, Soliman H, Ruschin M, Sahgal A, Myrehaug S, Tseng CL, Czarnota GJ, Jabehdar-Maralani P, Chugh B, Lau A, Stanisz GJ, Sadeghi-Naini A. Quantitative MRI Biomarkers of Stereotactic Radiotherapy Outcome in Brain Metastasis. Sci Rep 2019; 9:19830. [PMID: 31882597 PMCID: PMC6934477 DOI: 10.1038/s41598-019-56185-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/08/2019] [Indexed: 02/08/2023] Open
Abstract
About 20-40% of cancer patients develop brain metastases, causing significant morbidity and mortality. Stereotactic radiation treatment is an established option that delivers high dose radiation to the target while sparing the surrounding normal tissue. However, up to 20% of metastatic brain tumours progress despite stereotactic treatment, and it can take months before it is evident on follow-up imaging. An early predictor of radiation therapy outcome in terms of tumour local failure (LF) is crucial, and can facilitate treatment adjustments or allow for early salvage treatment. In this study, an MR-based radiomics framework was proposed to derive and investigate quantitative MRI (qMRI) biomarkers for the outcome of LF in brain metastasis patients treated with hypo-fractionated stereotactic radiation therapy (SRT). The qMRI biomarkers were constructed through a multi-step feature extraction/reduction/selection framework using the conventional MR imaging data acquired from 100 patients (133 lesions), and were applied in conjunction with machine learning techniques for outcome prediction and risk assessment. The results indicated that the majority of the features in the optimal qMRI biomarkers characterize the heterogeneity in the surrounding regions of tumour including edema and tumour/lesion margins. The optimal qMRI biomarker consisted of five features that predict the outcome of LF with an area under the curve (AUC) of 0.79, and a cross-validated sensitivity and specificity of 81% and 79%, respectively. The Kaplan-Meier analyses showed a statistically significant difference in local control (p-value < 0.0001) and overall survival (p = 0.01). Findings from this study are a step towards using qMRI for early prediction of local failure in brain metastasis patients treated with SRT. This may facilitate early adjustments in treatment, such as surgical resection or salvage radiation, that can potentially improve treatment outcomes. Investigations on larger cohorts of patients are, however, required for further validation of the technique.
Collapse
Affiliation(s)
- Elham Karami
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hany Soliman
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Mark Ruschin
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Chia-Lin Tseng
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | | | - Brige Chugh
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Angus Lau
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Greg J Stanisz
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - Ali Sadeghi-Naini
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| |
Collapse
|
39
|
Schroader B, Kong S, Anderson S, Williamson T, Sireci A, Shields K. Current status of biomarker testing in historically rare, high-unmet-need tumors: soft tissue sarcomas and thyroid cancers. Expert Rev Anticancer Ther 2019; 19:929-938. [DOI: 10.1080/14737140.2019.1682554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | - Sheldon Kong
- US Medical Affairs, Bayer HealthCare, Whippany, NJ, USA
| | | | | | | | - Kasia Shields
- Oncology Medical Communications, Xcenda, LLC, Palm Harbor, FL, USA
| |
Collapse
|
40
|
Spizzo G, Siebert U, Gastl G, Voss A, Schuster K, Leonard R, Seeber A. Cost-comparison analysis of a multiplatform tumour profiling service to guide advanced cancer treatment. COST EFFECTIVENESS AND RESOURCE ALLOCATION 2019; 17:23. [PMID: 31641338 PMCID: PMC6802110 DOI: 10.1186/s12962-019-0191-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Background Tumor profiling is increasingly used in advanced cancer patients to define treatment options, especially in refractory cases where no standard treatment is available. Caris Molecular Intelligence (CMI) is a multiplatform tumor profiling service that is comprehensive of next-generation sequencing (NGS) of DNA and RNA, immunohistochemistry (IHC) and in situ hybridisation (FISH). The aim of this study is to compare costs of CMI-guided treatment with prior or planned treatment options in correlation with outcome results. Methods Retrospective data from five clinical trials were collected to define the treatment decision prior to the receipt of the CMI report (n = 137 patients). A systematic review of treatment data from 11 clinical studies of CMI (n = 385 patients) allowed a comparison of planned vs actual (n = 137) and prior vs actual (n = 229) treatment costs. Results Treatment plan was changed in 88% of CMI-profiled cases. The actual CMI guided treatment cost per cycle was £995 in 385 treated patients. Planned treatment costs were comparable to actual treatment costs (£979 vs £945; p = 0.7123) and prior treatment costs were not significantly different to profiling-guided treatments (£892 vs £850; p = 0.631). Conclusions Caris Molecular Intelligence guided treatment cost per cycle was in the range of prior or planned treatment cost/cycle. Due to beneficial overall survival the additional cost of performing CMI's multiplatform testing to the treatment costs seems to be cost-effective.
Collapse
Affiliation(s)
- Gilbert Spizzo
- Department of Internal Medicine, Oncologic Day Hospital, Bressanone Hospital (SABES-ASDAA), Bressanone-Brixen, Italy.,2Department of Haematology and Oncology, Innsbruck Medical University, Innrain 66, 6020 Innsbruck, Austria
| | - Uwe Siebert
- Institute of Public Health, Medical Decision Making and HTA, Hall in Tirol, Austria
| | - Guenther Gastl
- 2Department of Haematology and Oncology, Innsbruck Medical University, Innrain 66, 6020 Innsbruck, Austria
| | | | | | | | - Andreas Seeber
- 2Department of Haematology and Oncology, Innsbruck Medical University, Innrain 66, 6020 Innsbruck, Austria.,6Laboratory for Oncogenomics, Tyrolean Cancer Research Institute, Innsbruck, Austria
| |
Collapse
|
41
|
Gregg JP, Li T, Yoneda KY. Molecular testing strategies in non-small cell lung cancer: optimizing the diagnostic journey. Transl Lung Cancer Res 2019; 8:286-301. [PMID: 31367542 PMCID: PMC6626860 DOI: 10.21037/tlcr.2019.04.14] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/14/2019] [Indexed: 01/21/2023]
Abstract
Molecular testing identifies patients with advanced non-small cell lung cancer (NSCLC) who may benefit from targeted therapy or immunotherapy (i.e., immune checkpoint inhibitor treatment for patients with high tumor mutational burden (TMB), microsatellite instability-high or mismatch repair-deficient tumors). Current guidelines state that molecular testing should be conducted at the time of initial diagnosis and tumor progression on targeted therapy. In real-world clinical practice in the United States (US), molecular testing is often not conducted or happens late in the diagnostic journey, resulting in delayed or inappropriate treatment. Herein, we review the rationale for molecular testing in advanced NSCLC, along with best-practice guidelines based on published recommendations and our own clinical experience, including a case study. We propose three strategies to optimize molecular testing in newly diagnosed patients with advanced NSCLC: (I) pulmonologists, interventional radiologists, or thoracic surgeons order molecular tests as soon as advanced NSCLC with an adenocarcinoma component is suspected; (II) liquid biopsies conducted early in the diagnostic pathway; and (III) pathologist-directed reflex testing, as conducted in other areas of oncology. To help facilitate these strategies, we outline our recommendations for optimal sample collection techniques and stewardship. In summary, we believe that implementation of these individual strategies will allow clinicians to effectively leverage available treatment options for advanced NSCLC, reducing the time to optimal treatment and improving patient outcomes.
Collapse
Affiliation(s)
- Jeffrey P. Gregg
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, CA, USA
| | - Tianhong Li
- Division of Hematology & Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis Comprehensive Cancer Center, CA, USA
| | - Ken Y. Yoneda
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis Medical Center, CA, USA
| |
Collapse
|
42
|
Reitsma M, Fox J, Borre PV, Cavanaugh M, Chudnovsky Y, Erlich RL, Gribbin TE, Anhorn R. Effect of a Collaboration Between a Health Plan, Oncology Practice, and Comprehensive Genomic Profiling Company from the Payer Perspective. J Manag Care Spec Pharm 2019; 25:601-611. [PMID: 30632889 PMCID: PMC10398083 DOI: 10.18553/jmcp.2019.18309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Comprehensive genomic profiling (CGP) is a next-generation sequencing-based methodology that detects 4 classes of genomic alterations, as well as gene signature biomarkers such as microsatellite instability and tumor mutational burden. In the context of precision oncology, CGP can help to direct treatment to genomically matched therapies. OBJECTIVE To describe the results of a 3-year observational analysis of patients undergoing testing with CGP assays (either FoundationOne or FoundationOne Heme) at a community oncology practice after a regional health plan implemented a medical policy that enabled coverage of CGP. METHODS A retrospective analysis of medical records was completed at the oncology practice from November 2013 to January 2017; this date range was chosen to coincide with the regional health plan's medical policy implementation of CGP. The medical policy provided coverage of CGP for patients with advanced solid and hematologic cancers. A medical record review assessed all previous and current molecular test results, matched therapy or clinical trial enrollment, and clinical outcomes (clinical benefit or disease progression). The potential cost diversion, from payer to study sponsor, for patients who enrolled in clinical trials was explored. RESULTS There were 96 patients in the community oncology practice who received CGP over the 3-year period, 86 of whom had clinically relevant genomic alterations. Of the 86, 15 patients were treated with genomically matched therapy, and 6 patients enrolled in clinical trials based on CGP results. In a subset of 32 patients who previously underwent conventional testing, most (84%) had clinically relevant genomic alterations detected by CGP that conventional testing did not identify, and a portion of these patients subsequently received treatment based on the CGP results. In the separate cost diversion analysis of 20 patients who enrolled in phase 1 clinical trials, an estimated $25,000 per-patient cost-benefit may have been accrued to the payer. CONCLUSIONS This observational analysis characterized the use of CGP in a large community oncology practice among a group of patients insured by a regional health plan. Clinical trial enrollment was facilitated by CGP use in the community setting and may have contributed to cost diversion from the payer to study sponsors. DISCLOSURES No separate study-related funding was provided by or to Priority Health, Foundation Medicine, and Cancer and Hematology Centers of West Michigan. Data analysis by Reitsma was conducted as part of an internship funded by Priority Health. Reitsma and Fox are employed by Priority Health. Anhorn, Vanden Borre, Cavanaugh, Chudnovsky, and Erlich are employed by Foundation Medicine.
Collapse
Affiliation(s)
- Mitchell Reitsma
- Priority Health, Grand Rapids, Michigan, and Oakland University William Beaumont School of Medicine, Rochester, Michigan
| | - John Fox
- Priority Health, Grand Rapids, Michigan
| | | | | | | | | | | | | |
Collapse
|
43
|
Hickmann AK, Frick M, Hadaschik D, Battke F, Bittl M, Ganslandt O, Biskup S, Döcker D. Molecular tumor analysis and liquid biopsy: a feasibility investigation analyzing circulating tumor DNA in patients with central nervous system lymphomas. BMC Cancer 2019; 19:192. [PMID: 30823914 PMCID: PMC6397454 DOI: 10.1186/s12885-019-5394-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 02/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background Central nervous system lymphomas (CNSL) is a devastating disease. Currently, a confirmatory biopsy is required prior to treatment. Objective Our investigation aims to prove the feasibility of a minimally-invasive diagnostic approach for the molecular characterization of CNSL. Methods Tissue biopsies from 6 patients with suspected CNSL were analyzed using a 649gene next-generation sequencing (NGS) tumor panel (tumor vs. reference tissue (EDTA-blood)). The individual somatic mutation pattern was used as a basis for the digital PCR analyzing circulating tumor DNA (ctDNA) from plasma and cerebrospinal fluid (CSF) samples, identifying one selected tumor mutation during this first step of the feasibility investigation. Results NGS-analysis of biopsy tissue revealed a specific somatic mutation pattern in all confirmed lymphoma samples (n = 5, NGS-sensitivity 100%) and none in the sample identified as normal brain tissue (NGS-specificity 100%). cfDNA-extraction was dependent on the extraction-kit used and feasible in 3 samples, in all of which somatic mutations were detectable (100%). Analysis of CSF-derived cfDNA was superior to plasma-derived cfDNA and routine microscopic analysis (lymphoma cells: n = 2, 40%). One patient showed a divergent molecular pattern, typical of Burkitt-Lymphoma (HIV+, serologic evidence of EBV-infection). Lumbar puncture was tolerated without complications, whereas biopsy caused 3 hemorrhages. Conclusions Our investigation provides evidence that analysis of cfDNA in central nervous system tumors is feasible using the described protocol. Molecular characterization of CNSL could be achieved by analysis of CSF-derived cfDNA. Knowledge of a tumor’s specific mutation pattern may allow initiation of targeted therapies, treatment surveillance and could lead to minimally-invasive diagnostics in the future. Electronic supplementary material The online version of this article (10.1186/s12885-019-5394-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne-Katrin Hickmann
- Department of Neurosurgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, 9600, St. Gallen, Switzerland. .,Neurosurgical Department, Klinikum Stuttgart, Stuttgart, Germany.
| | - Maximilian Frick
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Dirk Hadaschik
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Florian Battke
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany
| | - Markus Bittl
- Neurosurgical Department, Klinikum Stuttgart, Stuttgart, Germany
| | - Oliver Ganslandt
- Neurosurgical Department, Klinikum Stuttgart, Stuttgart, Germany
| | - Saskia Biskup
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,Outpatient Clinic for Human Genetics, Tübingen, Germany
| | - Dennis Döcker
- Center for Genomics and Transcriptomics (CeGaT) GmbH, Tübingen, Germany.,Outpatient Clinic for Human Genetics, Tübingen, Germany
| |
Collapse
|
44
|
Abstract
Massively parallel sequencing is emerging from research settings into clinical practice, helping the vision of precision medicine to become a reality. The most successful applications are using the tools of implementation science within the framework of the learning health-care system. This article examines the application of massively parallel sequencing to four clinical scenarios: pharmacogenomics, diagnostic testing, somatic testing for molecular tumor characterization, and population screening. For each application, it highlights an exemplar program to illustrate the enablers and challenges of implementation. International examples are also presented. These early lessons will allow other programs to account for these factors, helping to accelerate the implementation of precision medicine and health.
Collapse
Affiliation(s)
- Marc S Williams
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania 17822-2620, USA;
| |
Collapse
|
45
|
Signorovitch J, Zhou Z, Ryan J, Anhorn R, Chawla A. Budget impact analysis of comprehensive genomic profiling in patients with advanced non-small cell lung cancer. J Med Econ 2019; 22:140-150. [PMID: 30430885 DOI: 10.1080/13696998.2018.1549056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIMS Broad molecular profiling of patients with advanced non-small cell lung cancer (NSCLC) is strongly advised to optimize genomic matching with available targeted treatment options or investigational agents. Unlike conventional molecular diagnostic testing, or smaller hotspot panels, comprehensive genomic profiling (CGP) identifies genomic alterations across hundreds of clinically relevant cancer genes from a single tissue specimen. The present study sought to estimate the budget impact of increased use of CGP using a 324-gene panel (FoundationOne) vs non-CGP (represented by a mix of conventional molecular diagnostic testing and smaller NGS hotspot panels) and the number needed to test with CGP to gain 1 life year. MATERIALS AND METHODS A decision analytic model was developed to assess the budget impact of increased CGP in advanced NSCLC from a US private payer perspective. Model inputs were based on published literature (epidemiology and treatment outcomes), real-world data (testing and rates, medical service costs), list prices for CGP and anti-cancer drugs, and assumptions for clinical trial participation. RESULTS Among 2 million covered lives, 532 had advanced NSCLC; 266 underwent molecular diagnostic testing. An increase in CGP among those tested, from 2% to 10%, was associated with $0.02 per member per month budget impact, of which $0.013 was attributable to costs of prolonged drug treatment and survival and $0.005 to testing cost. Approximately 12 patients would need to be tested with CGP to add 1 life year. LIMITATIONS The model incorporated certain assumptions to account for inputs with a limited evidence profile and simplify the possible post-CGP treatments. CONCLUSIONS An increase in CGP utilization from 2% to 10% among patients with advanced NSCLC undergoing molecular diagnostic testing was associated with a modest budget impact, most of which was attributable to increased use of more effective treatments and prolonged survival.
Collapse
Affiliation(s)
| | - Zhou Zhou
- a Analysis Group, Inc. , Boston , MA , USA
| | - Jason Ryan
- b Foundation Medicine, Inc. , Cambridge , MA , USA
| | | | | |
Collapse
|
46
|
Thompson MA, Godden JJ, Wham D, Ruggeri A, Mullane MP, Wilson A, Virani S, Weissman SM, Ramczyk B, Vanderwall P, Weese JL. Coordinating an Oncology Precision Medicine Clinic Within an Integrated Health System: Lessons Learned in Year One. J Patient Cent Res Rev 2019; 6:36-45. [PMID: 31414022 PMCID: PMC6676755 DOI: 10.17294/2330-0698.1639] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Precision medicine is a term describing strategies to promote health and prevent and treat disease based on an individual's genetic, molecular, and lifestyle characteristics. Oncology precision medicine (OPM) is a cancer treatment approach targeting cancer-specific genetic and molecular alterations. Implementation of an OPM clinical program optimally involves the support and collaboration of multiple departments, including administration, medical oncology, pathology, interventional radiology, genetics, research, and informatics. In this review, we briefly introduce the published evidence regarding OPM's potential effect on patient outcomes and discuss what we have learned over the first year of operating an OPM program within an integrated health care system (Aurora Health Care, Milwaukee, WI) comprised of multiple hospitals and clinics. We also report our experience implementing a specific OPM software platform used to embed molecular panel data into patients' electronic medical records.
Collapse
Affiliation(s)
- Michael A. Thompson
- Aurora Research Institute, Aurora Health Care, Milwaukee, WI
- Aurora Cancer Care, Aurora Health Care, Milwaukee, WI
| | | | - Deborah Wham
- Aurora Cancer Care, Aurora Health Care, Milwaukee, WI
| | | | | | - Amanda Wilson
- Pathology, Aurora St. Luke’s Medical Center, Milwaukee, WI
| | | | - Scott M. Weissman
- Aurora Cancer Care, Aurora Health Care, Milwaukee, WI
- Chicago Genetic Consultants, LLC, Northbrook, IL
| | | | | | | |
Collapse
|
47
|
Nadauld LD, Ford JM, Pritchard D, Brown T. Strategies For Clinical Implementation: Precision Oncology At Three Distinct Institutions. Health Aff (Millwood) 2019; 37:751-756. [PMID: 29733728 DOI: 10.1377/hlthaff.2017.1575] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Despite rapid advances in molecular diagnostics and targeted therapeutics, the adoption of precision medicine into clinical oncology workflows has been slow. Questions about clinical utility, inconsistent reimbursement for molecular diagnostics, and limited access to targeted therapies are some of the major hurdles that have hampered clinical adoption. Despite these challenges, providers have invested in precision medicine programs in an ongoing search for innovative care models to deliver improved patient outcomes and achieve economic gains. We describe the precision oncology medicine programs implemented by an integrated delivery system, a community care center, and an academic medical center, to demonstrate the approaches and challenges associated with clinical implementation efforts designed to advance this treatment paradigm. Payer policies that include coverage for broad genomic testing panels would support the broader application of precision medicine, deepen research benefits, and bring targeted therapies to more patients with advanced cancer.
Collapse
Affiliation(s)
- Lincoln D Nadauld
- Lincoln D. Nadauld ( ) is executive director for precision genomics and precision medicine at Intermountain Healthcare, in Salt Lake City, Utah
| | - James M Ford
- James M. Ford is a professor of medicine and genetics in the Division of Oncology, Stanford Medicine, Stanford University, in California
| | - Daryl Pritchard
- Daryl Pritchard is vice president for science policy at the Personalized Medicine Coalition, in Washington, D.C
| | - Thomas Brown
- Thomas Brown is executive director of the Swedish Cancer Institute, in Seattle, Washington
| |
Collapse
|
48
|
McKenzie AJ, H Dilks H, Jones SF, Burris H. Should next-generation sequencing tests be performed on all cancer patients? Expert Rev Mol Diagn 2019; 19:89-93. [PMID: 30618301 DOI: 10.1080/14737159.2019.1564043] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
| | - Holli H Dilks
- a Sarah Cannon Research Institute , Nashville , Tennessee , USA
| | - Suzanne F Jones
- a Sarah Cannon Research Institute , Nashville , Tennessee , USA
| | - Howard Burris
- a Sarah Cannon Research Institute , Nashville , Tennessee , USA
| |
Collapse
|
49
|
Demeure MJ. The Role of Precision Medicine in the Diagnosis and Treatment of Patients with Rare Cancers. Cancer Treat Res 2019; 178:81-108. [PMID: 31209842 DOI: 10.1007/978-3-030-16391-4_3] [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] [Indexed: 06/09/2023]
Abstract
Rare cancers pose unique challenges for patients and their physicians arising from a lack of information regarding the best therapeutic options. Very often, a lack of clinical trial data leads physicians to choose treatments based on small case series or case reports. Precision medicine based on genomic analysis of tumors may allow for selection of better treatments with greater efficacy and less toxicity. Physicians are increasingly using genetics to identify patients at high risk for certain cancers to allow for early detection or prophylactic interventions. Genomics can be used to inform prognosis and more accurately establish a diagnosis. Genomic analysis may also expose therapeutic targets for which drugs are currently available and approved for use in other cancers. Notable successes in the treatment of previously refractory cancers have resulted. New more advanced sequencing technologies, tools for interpretation, and an increasing array of targeted drugs offer additional hope, but challenges remain.
Collapse
Affiliation(s)
- Michael J Demeure
- Hoag Family Cancer Institute, Newport Beach, CA, USA.
- Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
50
|
Burris HA, Saltz LB, Yu PP. Assessing the Value of Next-Generation Sequencing Tests in a Dynamic Environment. Am Soc Clin Oncol Educ Book 2018; 38:139-146. [PMID: 30231307 DOI: 10.1200/edbk_200825] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Next-generation sequencing (NGS)-based technology has lowered the cost of cancer testing for genomic alterations and is now commercially available from a growing number of diagnostic laboratories. However, laboratories vary in the methodologies underlying their tests, the types and numbers of genomic alterations covered by the test, and the clinical annotation of the sequencing findings. Determining the value of NGS tests is dependent on whether it is used to support clinical trials or as a part of routine clinical care at a time when both the investigational drug pipeline and the list of U.S. Food and Drug Administration-approved or Compendium-listed therapeutics is in a high state of flux. Reimbursement policy for NGS testing by the Centers for Medicare & Medicaid is evolving as the value of NGS testing becomes more clearly defined for specific clinical situations. Patient care and clinical decisions-making are dependent on the oncologist's knowledge of when NGS testing has value. Here, we review principles and practice for NGS testing in this dynamic confluence of technology, cancer biology, and health care policy.
Collapse
Affiliation(s)
- Howard A Burris
- From Sarah Cannon, Nashville, TN; Memorial Sloan Kettering Cancer Center, New York, NY; Hartford HealthCare Cancer Institute, Hartford, CT, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Leonard B Saltz
- From Sarah Cannon, Nashville, TN; Memorial Sloan Kettering Cancer Center, New York, NY; Hartford HealthCare Cancer Institute, Hartford, CT, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Peter P Yu
- From Sarah Cannon, Nashville, TN; Memorial Sloan Kettering Cancer Center, New York, NY; Hartford HealthCare Cancer Institute, Hartford, CT, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|