1
|
Pineault L, Valencia K, Buhay J, Brown A, Ziemnik S, Kelly M, Morgante J, Datta Mitra A, Yanamura L, Gilliland M, Ferea A, Kim J. ASCP explores the cancer biomarker testing navigator as a novel role to improve laboratory operations and workflows: A special report from the ASCP Biomarker Testing Navigator Project Team. Am J Clin Pathol 2025:aqaf028. [PMID: 40329479 DOI: 10.1093/ajcp/aqaf028] [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: 08/13/2024] [Indexed: 05/08/2025] Open
Abstract
OBJECTIVE Cancer biomarker testing is a critical element in precision oncology, guiding treatment decisions and improving patient outcomes. However, the complexity and variability of biomarker testing processes present significant challenges for cancer centers, often leading to delays and inefficiencies that can compromise care quality. The American Society for Clinical Pathology explored the concept of a novel laboratory professional role: the cancer biomarker testing navigator (BTN). METHODS This study explored the feasibility and impact of the BTN role on laboratory operations and workflows through a 3-phase project consisting of a quantitative needs assessment, qualitative focus group discussions, and a short-term feasibility pilot conducted at 2 cancer centers. RESULTS The needs assessment revealed that many laboratories lack dedicated staff for coordinating biomarker testing, leading to operational inefficiencies. The roundtable discussions highlighted common challenges in biomarker testing and identified potential benefits of the BTN role, such as improved communication, better tracking of send-out tests, and enhanced task efficiency. The feasibility pilot demonstrated that BTNs could coordinate multigene next-generation sequencing panels and expedite key steps to ensure optimal preanalytical processes, reduce delays in testing, and smooth operations. The BTN role represents a feasible and beneficial addition to pathology laboratories that addresses key operational challenges in cancer biomarker testing and offers a promising solution to streamline laboratory operations, improve multidisciplinary communication, and enhance patient care coordination. CONCLUSIONS Further exploration is warranted to refine the BTN role and assess its long-term sustainability in and impact on diverse laboratory settings.
Collapse
Affiliation(s)
| | - Karla Valencia
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer Buhay
- American Society for Clinical Pathology, Chicago, IL, United States
| | - Alexandra Brown
- American Society for Clinical Pathology, Chicago, IL, United States
| | - Suzanne Ziemnik
- American Society for Clinical Pathology, Chicago, IL, United States
| | - Melissa Kelly
- American Society for Clinical Pathology, Chicago, IL, United States
| | - James Morgante
- American Society for Clinical Pathology, Chicago, IL, United States
| | - Ananya Datta Mitra
- Department of Pathology, University of California Davis Health, Sacramento, CA, United States
| | - Lindsay Yanamura
- Department of Pathology, University of California Davis Health, Sacramento, CA, United States
| | - Marie Gilliland
- Spartanburg Regional Healthcare System, Spartanburg, SC, United States
| | - Amy Ferea
- Spartanburg Regional Healthcare System, Spartanburg, SC, United States
| | - Joseph Kim
- Q Synthesis LLC, Newtown, PA, United States
| |
Collapse
|
2
|
Trisolini R, Cetoretta V, Sotgiu G, Cancellieri A, Puci M, Viscuso M, Livi V, Cani M, Scambia G, Cappuzzo F, Bria E, Novello S. Supraclavicular Lymph Node Metastases in Advanced Lung Cancer: Prevalence and Analysis of Demographic, Clinical and Molecular Characteristics. Clin Lung Cancer 2025:S1525-7304(25)00033-6. [PMID: 40037966 DOI: 10.1016/j.cllc.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND The prevalence of supraclavicular lymph nodes metastases (SNM) in advanced lung cancer has not been systematically evaluated, nor has then been a comparison of demographic, clinical, or molecular characteristics between patients with and without SNM. METHODS In this prospective cohort study, the presence of SNM was evaluated using imaging studies (CT, PET, neck ultrasonography) in patients with suspected advanced lung cancer referred for biopsy aimed at diagnosis and molecular profiling. Ultrasound-guided biopsy confirmed or excluded metastatic involvement when suspicious supraclavicular nodes were identified. We assessed the prevalence of SNM and compared the demographic, clinicopathologic and molecular characteristics of patients with and without SNM. RESULTS Among the 348 patients with advanced lung cancer, 94 (27%) had SMN. SMN was more common in small cell lung cancer (24/48, 50%) and adenocarcinoma (61/248, 24.6%) than in squamous cell carcinoma (4/35, 11.4%). Compared to patients without SMN, those with SMN were more likely to have small-cell lung cancer, N2/3 disease (97.9 vs. 83.9%, P < .0001), liver metastases (29.8% vs. 16.1% P = .006), and metastases to less common sites (33.7% vs. 14.1%, P < .0001). The prevalence of genomic alterations and PD-L1 expression did not differ between biopsy samples obtained from SNM and those from the primary tumor or other metastatic sites. CONCLUSION SNM is common in patients with advanced small-cell lung cancer and adenocarcinoma. Ultrasound-guided biopsy of SNM is a simple and relatively inexpensive method for obtaining adequate tissue samples for diagnosis and comprehensive molecular profiling.
Collapse
Affiliation(s)
- Rocco Trisolini
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy
| | - Valeria Cetoretta
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy; Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Mariangela Puci
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Marta Viscuso
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy; Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanina Livi
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy
| | - Massimiliano Cani
- Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Department of Woman, Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federico Cappuzzo
- Medical Oncology Division, IRCCS Regina Elena National Cancer Institute, Rome, Italy Medical
| | - Emilio Bria
- Oncology Division, Gemelli Isola Hospital, Catholic University of the Sacred Hearth, Rome, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy.
| |
Collapse
|
3
|
Lockhart CM, Manolakis M. Emerging trends in therapeutics and diagnostics: Perspectives on the 2024 AMCP Foundation Survey. J Manag Care Spec Pharm 2025; 31:S15-S19. [PMID: 39888854 PMCID: PMC11785362 DOI: 10.18553/jmcp.2025.31.2-a.s15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
4
|
Koch A, Reinhardt P, Elicin O, Aebersold DM, Schanne DH. Predictive biomarkers of radiotherapy- related dermatitis, xerostomia, mucositis and dysphagia in head and neck cancer: A systematic review. Radiother Oncol 2025; 203:110689. [PMID: 39706342 DOI: 10.1016/j.radonc.2024.110689] [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: 08/20/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Radiotherapy is essential for treating head and neck cancer but often leads to severe toxicity. Traditional predictors include anatomical location, tumor extent, and dosimetric data. Recently, biomarkers have been explored to better predict and understand toxicity. This review aims to summarize the current literature, assess data quality, and guide future research. METHODS Two reviewers independently screened EMBASE and PubMed for studies published between 2010 and 2023. Endpoints were dermatitis, mucositis, sticky saliva/xerostomia, and dysphagia. Statistical analysis was performed using R, and bias assessed via a modified QUIPS questionnaire. Pathway analysis was conducted using gProfiler. The study adhered to PRISMA and COSMOS-E guidelines and was registered in the PROSPERO database (#CRD42023361245). RESULTS Of 2,550 abstracts, 69 publications met the inclusion criteria. These studies involved a median of 81 patients, primarily male (75 %), with common primary tumors in the nasopharynx (32 %) and oropharynx (27 %). Most patients (84 %) had advanced disease (stage III/IV). The most frequently studied biomarkers were DNA-based single-nucleotide polymorphisms (SNPs, 59 %), salivary proteins (13 %), and bacteria (10 %). Ten statistically-significant biomarkers (all SNPs) in low-bias publications were identified, particularly in DNA repair and cell detoxification pathways. Data quality was often poor and few validation studies were present in the dataset. CONCLUSION This review provides an overview of the research landscape, highlights research gaps and provides recommendations for future research directions. We identified several potential biomarkers, particularly in DNA repair pathways, that align with current understanding of radiation-induced cell damage. However, the overall data quality was poor, with key clinical variables often missing. Overall, rigorous standardization of reporting, validation studies and multi-center collaborations to increase study power and sample sizes are necessary to build high-level evidence for clinical application.
Collapse
Affiliation(s)
- Alexander Koch
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Philipp Reinhardt
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Olgun Elicin
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Daniel H Schanne
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland; Graduate School for Health Sciences, University of Bern, Switzerland.
| |
Collapse
|
5
|
Lockhart CM, Manolakis M. Emerging trends in therapeutics and diagnostics: Perspectives on the 2024 AMCP Foundation Survey. J Manag Care Spec Pharm 2025; 31:S15-S19. [PMID: 39873563 PMCID: PMC11780270 DOI: 10.18553/jmcp.2025.31.1-b.s15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
|
6
|
Wallen ZD, Nesline MK, Tierno M, Roos A, Schnettler E, Husain H, Sathyan P, Caveney B, Eisenberg M, Severson EA, Ramkissoon SH. Genomic profiling of NSCLC tumors with the TruSight oncology 500 assay provides broad coverage of clinically actionable genomic alterations and detection of known and novel associations between genomic alterations, TMB, and PD-L1. Front Oncol 2024; 14:1473327. [PMID: 39664186 PMCID: PMC11631745 DOI: 10.3389/fonc.2024.1473327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction Matching patients to an effective targeted therapy or immunotherapy is a challenge for advanced and metastatic non-small cell lung cancer (NSCLC), especially when relying on assays that test one marker at a time. Unlike traditional single marker tests, comprehensive genomic profiling (CGP) can simultaneously assess NSCLC tumors for hundreds of genomic biomarkers and markers for immunotherapy response, leading to quicker and more precise matches to therapeutics. Methods In this study, we performed CGP on 7,606 patients with advanced or metastatic NSCLC using the Illumina TruSight Oncology 500 (TSO 500) CGP assay to show its coverage and utility in detecting known and novel features of NSCLC. Results Testing revealed distinct genomic profiles of lung adenocarcinoma and squamous cell carcinomas and detected variants with a current targeted therapy or clinical trial in >72% of patient tumors. Known associations between genomic alterations and immunotherapy markers were observed including significantly lower TMB levels in tumors with therapy-associated alterations and significantly higher PD-L1 levels in tumors with ALK, MET, BRAF, or ROS1 driver mutations. Co-occurrence analysis followed by network analysis with gene module detection revealed known and novel co-occurrences between genomic alterations. Further, certain modules of genes with co-occurring genomic alterations had dose-dependent relationships with histology and increasing or decreasing levels of PD-L1 and TMB, suggesting a complex relationship between PD-L1, TMB, and genomic alterations in these gene modules. Discussion This study is the largest clinical study to date utilizing the TSO 500. It provides an opportunity to further characterize the landscape of NSCLC using this newer technology and show its clinical utility in detecting known and novel facets of NSCLC to inform treatment decision-making.
Collapse
Affiliation(s)
| | | | | | | | | | - Hatim Husain
- Moores Cancer Center at UC San Diego Health, La Jolla, CA, United States
| | | | | | | | | | - Shakti H. Ramkissoon
- Labcorp Oncology, Durham, NC, United States
- Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Department of Pathology, Winston-Salem, NC, United States
| |
Collapse
|
7
|
Vakkalagadda CV, Dressler DB, Sun Z, Fuchs J, Liu Y, Silberman P, Ragam A, Kircher S, Patel JD, Mohindra NA. The Impact of Next-Generation Sequencing Workflows on Outcomes in Advanced Lung Cancer: A Retrospective Analysis at One Academic Health System. Cancers (Basel) 2024; 16:3654. [PMID: 39518093 PMCID: PMC11545180 DOI: 10.3390/cancers16213654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Broad-based molecular testing with next-generation sequencing (NGS) is now the standard of care in advanced non-small cell lung cancer (NSCLC). Two approaches to molecular testing are (1) reflexive testing at pathologic NSCLC confirmation, often using an in-house molecular panel, and (2) send-out testing to private vendors, ordered by a clinician. This study explored the outcomes with reflex versus send-out testing. METHODS A retrospective chart review was conducted of patients diagnosed with de novo stage IV NSCLC in 2019 and 2020 at three hospitals in the same system, one academic hospital (Northwestern Memorial Hospital, or NMH) utilizing reflex, in-house NGS, and two community-based hospitals (Central DuPage Hospital, or CDH, and Delnor, or D) sending out tissue samples for testing. The outcomes assessed were the time from biopsy to results, biopsy to treatment, the incidence of first-line targetable mutations and the use of first-line targeted therapies, and overall survival. RESULTS In total, 191 patients met the inclusion criteria, 85 at NMH, 106 at CDH + D, and in total, 131 in 2019 and 60 in 2020. The time to results was significantly shorter with reflexive NGS when compared with send-out testing; the time to treatment was also shorter but not statistically significant. At CDH + D, the time to results was significantly shorter with a limited panel than with comprehensive testing, but the time to treatment was similar. NGS testing rates were 95% at NMH and 84.5% at CDH + D (p = 0.009), with 31.0% at NMH receiving 1L targeted therapies versus 20.8% at CDH + D (p = 0.08). In 2019, the median time from biopsy to treatment was 35 days at NMH and 38 days at CDH and Delnor; in 2020, time to treatment was 26 days and 37 days, respectively. Overall survival trended longer in 2020 relative to 2019 independent of site. CONCLUSION Reflexive NGS testing is associated with a shorter time to actionable results and higher rates of first-line targetable mutations than send-out testing. In practices with send-out testing, limited panels had slightly faster turnaround times but no difference in time to treatment. If resources allow, reflexive NGS should be considered in healthcare systems for patients with NSCLC.
Collapse
Affiliation(s)
- Chetan V. Vakkalagadda
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Danielle B. Dressler
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
- Division of Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Zequn Sun
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
| | - Joseph Fuchs
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
| | - Yingzhe Liu
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
| | - Philip Silberman
- Enterprise Data Warehouse, Clinical and Translational Sciences Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Avanthi Ragam
- Northwestern Medicine Delnor Hospital, Geneva, IL 60134, USA
| | - Sheetal Kircher
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
| | - Jyoti D. Patel
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
| | - Nisha A. Mohindra
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (N.A.M.)
| |
Collapse
|
8
|
D’Agnano V, Perrotta F, Stella GM, Pagliaro R, De Rosa F, Cerqua FS, Schiattarella A, Grella E, Masi U, Panico L, Bianco A, Iadevaia C. Molecular Diagnostic Yield and Safety Profile of Ultrasound-Guided Lung Biopsies: A Cross-Sectional Study. Cancers (Basel) 2024; 16:2860. [PMID: 39199631 PMCID: PMC11352358 DOI: 10.3390/cancers16162860] [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: 07/10/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND The recent advances in precision oncology for lung cancer treatment has focused attention on the importance of obtaining appropriate specimens for tissue diagnosis as well as comprehensive molecular profiling. CT scan-guided biopsies and bronchoscopy are currently the main procedures employed for tissue sampling. However, growing evidence suggests that ultrasound-guided biopsies may represent an effective as well as safe approach in this diagnostic area. This study explores the safety and the diagnostic yield for cancer molecular profiling in ultrasound-guided percutaneous lung lesion biopsies (US-PLLB). METHODS One hundred consecutive patients with suspected lung cancer, between January 2021 and May 2024, who had ultrasound-guided lung biopsies have been retrospectively analyzed. Molecular profiling was conducted with next-generation sequencing Genexus using Oncomine precision assay or polymerase chain reaction according to specimen quality. Qualitative immunohistochemical assay of programmed death ligand 1 (PD-L1) expression was evaluated by the Dako PD-L1 immunohistochemistry 22C3 pharmDx assay. The co-primary endpoints were the molecular diagnostic yield and the safety profile of US-guided lung biopsies. RESULTS From January 2021 to May 2024, 100 US-guided lung biopsies were carried out and 95 were considered for inclusion in the study. US-PLLB provided informative tissue for a histological evaluation in 93 of 95 patients with an overall diagnostic accuracy of 96.84% [Sensitivity: 92.63%; Specificity: 96.84%; PPV: 100%; NPV: 100%]. Sixty-Six patients were diagnosed with NSCLC (69.47%) and were considered for molecular diagnostic yield evaluation and PD-L1 testing. Four patients had malignant lymphoid lesions. US-PLLB was not adequate to achieve a final diagnosis in three patients (3.16%). Complete molecular profiling and PD-L1 evaluation were achieved in all patients with adenocarcinoma (molecular diagnostic yield: 100%). PD-L1 evaluation was achieved in 28 of 29 patients (96.55%) with either SCC or NOS lung cancer. The overall complication rate was 9.47% (n = 9). Six patients (6.31%) developed pneumothorax, while three patients (3.16%) suffered mild haemoptysis without desaturation. CONCLUSIONS According to our findings, US-guided lung biopsy is a safe, minimally invasive procedure in patients with suspected lung malignancies, providing an excellent diagnostic yield for both comprehensive molecular profiling and PD-L1 testing. In addition, our results suggest that US-guided biopsy may also be an effective diagnostic approach in patients with suspected lung lymphoma.
Collapse
Affiliation(s)
- Vito D’Agnano
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| | - Giulia Maria Stella
- Unit of Respiratory Diseases, Department of Medical Sciences and Infective Diseases, IRCCS Policlinico San Matteo Foundation, University of Pavia Medical School, 27100 Pavia, Italy
| | - Raffaella Pagliaro
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| | - Filippo De Rosa
- Unit of Pathology Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.D.R.); (L.P.)
| | - Francesco Saverio Cerqua
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| | - Angela Schiattarella
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| | - Edoardo Grella
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
| | - Umberto Masi
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
| | - Luigi Panico
- Unit of Pathology Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.D.R.); (L.P.)
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (F.P.); (R.P.); (A.S.); (E.G.); (U.M.); (A.B.)
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| | - Carlo Iadevaia
- U.O.C. Clinica Pneumologica L. Vanvitelli, Monaldi Hospital, A.O. dei Colli, 80131 Naples, Italy; (F.S.C.); (C.I.)
| |
Collapse
|
9
|
Abbass IM, Sheinson DM, Shah A, Gondos A, Ogale S. Cost-effectiveness of large-panel next-generation sequencing in guiding first-line treatment decisions for patients with nonsquamous advanced non-small cell lung cancer. J Manag Care Spec Pharm 2024; 30:649-659. [PMID: 38950160 PMCID: PMC11217867 DOI: 10.18553/jmcp.2024.30.7.649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
BACKGROUND Clinical practice guidelines recommend broad-panel genomic profiling to identify actionable genomic alterations for patients with advanced non-small cell lung cancer (aNSCLC). OBJECTIVE To assess the cost-effectiveness of large-panel next-generation sequencing (LP-NGS) compared with current empirical single-gene test (SGT) patterns to inform first-line treatment decisions for patients with aNSCLC from a US commercial payer perspective, accounting for the effect of testing turnaround time and time to treatment initiation. METHODS We developed a discrete-event simulation model to estimate the impact of LP-NGS vs SGT for patients with nonsquamous aNSCLC. Discrete events and timing included testing patterns, receipt of the initial test result, treatment initiation (targeted vs nontargeted therapies), switching, retesting, rebiopsies, clinical trial participation, progression on therapy, and death. LP-NGS and SGT cohorts each comprised 100,000 adults with aNSCLC simulated over a 5-year postdiagnosis period, assumed to have the same distribution of genomic alterations. The model predicted the proportion of patients receiving appropriate first-line therapy according to clinical practice guidelines. Economic outcomes included expected life-years gained, quality-adjusted life-years, and the total costs of care over 5 years. Sensitivity and scenario analyses explored the robustness of the base-case model results. RESULTS In the base-case model, LP-NGS was likely to identify more alterations than SGT. Total 5-year costs per patient were $539,658 for LP-NGS and $544,550 for SGT (net difference, $4,892 lower costs per patient for LP-NGS), which is likely to be cost-effective 95.1% of the time. The most influential model parameters on the 5-year total costs of care were preprogression nondrug medical costs on nontargeted therapy, NGS turnaround time, and clinical trial participation. CONCLUSIONS This study suggests that LP-NGS to guide first-line treatment decisions is clinically more appropriate (more likely to identify alterations and subsequently allocate patients to clinically appropriate therapy) and provides a dominant cost-effectiveness treatment strategy over 5 years for patients with newly diagnosed aNSCLC in the United States.
Collapse
Affiliation(s)
| | | | - Anuj Shah
- Genentech Inc, South San Francisco, CA
- Gilead Sciences, Foster City, CA
| | - Adam Gondos
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | |
Collapse
|
10
|
Nesline MK, Subbiah V, Previs RA, Strickland KC, Ko H, DePietro P, Biorn MD, Cooper M, Wu N, Conroy J, Pabla S, Zhang S, Wallen ZD, Sathyan P, Saini K, Eisenberg M, Caveney B, Severson EA, Ramkissoon S. The Impact of Prior Single-Gene Testing on Comprehensive Genomic Profiling Results for Patients with Non-Small Cell Lung Cancer. Oncol Ther 2024; 12:329-343. [PMID: 38502426 PMCID: PMC11187032 DOI: 10.1007/s40487-024-00270-x] [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: 12/29/2023] [Accepted: 02/29/2024] [Indexed: 03/21/2024] Open
Abstract
INTRODUCTION Tissue-based broad molecular profiling of guideline-recommended biomarkers is advised for the therapeutic management of patients with non-small cell lung cancer (NSCLC). However, practice variation can affect whether all indicated biomarkers are tested. We aimed to evaluate the impact of common single-gene testing (SGT) on subsequent comprehensive genomic profiling (CGP) test outcomes and results in NSCLC. METHODS Oncologists who ordered SGT for guideline-recommended biomarkers in NSCLC patients were prospectively contacted (May-December 2022) and offered CGP (DNA and RNA sequencing), either following receipt of negative SGT findings, or instead of SGT for each patient. We describe SGT patterns and compare CGP completion rates, turnaround time, and recommended biomarker detection for NSCLC patients with and without prior negative SGT results. RESULTS Oncologists in > 80 community practices ordered CGP for 561 NSCLC patients; 135 patients (27%) first had negative results from 30 different SGT combinations; 84% included ALK, EGFR and PD-L1, while only 3% of orders included all available SGTs for guideline-recommended genes. Among patients with negative SGT results, CGP was attempted using the same tissue specimen 90% of the time. There were also significantly more CGP order cancellations due to tissue insufficiency (17% vs. 7%), DNA sequencing failures (13% vs. 8%), and turnaround time > 14 days (62% vs. 29%) than among patients who only had CGP. Forty-six percent of patients with negative prior SGT had positive CGP results for recommended biomarkers, including targetable genomic variants in genes beyond ALK and EGFR, such as ERBB2, KRAS (non-G12C), MET (exon 14 skipping), NTRK2/3, and RET . CONCLUSION For patients with NSCLC, initial use of SGT increases subsequent CGP test cancellations, turnaround time, and the likelihood of incomplete molecular profiling for guideline-recommended biomarkers due to tissue insufficiency.
Collapse
Affiliation(s)
- Mary K Nesline
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA.
| | - Vivek Subbiah
- Sarah Cannon Research Institute, Early-Phase Drug Development, Nashville, TN, 37203, USA
| | - Rebecca A Previs
- Labcorp Oncology, Durham, NC, 27560, USA
- Duke Cancer Institute, Department of Obstetrics & Gynecology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Kyle C Strickland
- Labcorp Oncology, Durham, NC, 27560, USA
- Duke Cancer Institute, Department of Pathology, Duke University Medical Center, Durham, NC, 27710, USA
- Department of Gynecologic Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Heidi Ko
- Labcorp Oncology, Durham, NC, 27560, USA
| | - Paul DePietro
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | | | | | - Nini Wu
- Cardinal Health, Dublin, OH, 43017, USA
| | - Jeffrey Conroy
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Sarabjot Pabla
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Shengle Zhang
- Labcorp Oncology, 700 Ellicott Street, Buffalo, NY, 14203, USA
| | | | | | | | | | | | | | - Shakti Ramkissoon
- Labcorp Oncology, Durham, NC, 27560, USA
- Department of Pathology, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| |
Collapse
|
11
|
Stoeckle JH, Poland SG, Maynard H, Roman SD, Mettman D, Makarov DV, Sherman S, Becker DJ. Trial of Electronic Medical Record Integrated Next-Generation Sequencing Ordering in Veterans Affairs Cancer Care. JCO Precis Oncol 2024; 8:e2300463. [PMID: 38691812 DOI: 10.1200/po.23.00463] [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: 08/23/2023] [Revised: 01/18/2024] [Accepted: 03/15/2024] [Indexed: 05/03/2024] Open
Abstract
PURPOSE Previous studies document underuse of next-generation sequencing (NGS). We examined the impact to oncology care for veterans of incorporating NGS ordering into the Veterans Affairs (VA) electronic medical record (EMR) at two New York City VA Medical Centers. METHODS We identified patients with non-small cell lung cancer and prostate cancer with oncology clinic visits and NGS testing indications between January and December 2021. Patients were divided into external ordering (EO) with visits before we implemented an EMR ordering system for NGS in July 2021, and internal ordering (IO) with visits after this date. The primary outcome was proportion of NGS testing performed in EO versus IO groups. Secondary outcomes were time between metastatic disease diagnosis to receipt of test by vendor, time of metastatic diagnosis to result, and proportion of testing by race. RESULTS A total of 168 patients were identified, 116 EO and 52 IO patients. Between IO and EO periods, testing significantly increased from 52% to 87% (P ≤ .01); it was conducted more quickly, with time from metastatic diagnosis to sample receipt by the NGS vendor improving to median 37 days from 299 days (P = .03); and the time from documented metastatic disease to a test result improved to median 56 days from 309 days (P = .03). The proportion of tissue received by the vendor was not significantly different between the two groups. There were no significant differences in testing according to self-reported race. CONCLUSION Integration of NGS ordering in the EMR led to increased proportion and speed of testing for a vulnerable patient population served by the country's largest health system.
Collapse
Affiliation(s)
| | - Sarah G Poland
- New York University School of Medicine, NYU Langone Health, New York, NY
| | - Hannah Maynard
- New York University School of Medicine, NYU Langone Health, New York, NY
| | | | - Daniel Mettman
- New York University School of Medicine, NYU Langone Health, New York, NY
- VA NY Harbor Healthcare System, New York, NY
| | - Danil V Makarov
- New York University School of Medicine, NYU Langone Health, New York, NY
- VA NY Harbor Healthcare System, New York, NY
| | - Scott Sherman
- New York University School of Medicine, NYU Langone Health, New York, NY
- VA NY Harbor Healthcare System, New York, NY
| | - Daniel J Becker
- New York University School of Medicine, NYU Langone Health, New York, NY
- VA NY Harbor Healthcare System, New York, NY
| |
Collapse
|
12
|
Utzig M, Hoffmann H, Reinmuth N, Schütte W, Langer T, Lobitz J, Rückher J, Wesselmann S. Development and Update of Guideline-based Quality Indicators in Lung Cancer. Pneumologie 2024; 78:250-261. [PMID: 38081218 DOI: 10.1055/a-2204-4879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
BACKGROUND In 2022, an update of the German lung cancer guideline, first published in 2010 and revised in 2018, was released. This article aims to show the process of updating, developing, and implementing guideline-based quality indicators (QI) into the certification system for lung cancer centers (LCC). METHODS A multidisciplinary and interprofessional working group revised the guideline QIs from 2018 using the strong recommendations of the guideline update, a systematic review for QIs, and the results of the implemented QIs from LCC. RESULTS For 4 out of 8 indicators from the 2018 guideline, the LCC showed an improved implementation of the requirements in the last 3 years (2018-2020). For 3 indicators, the median of the results was constant at a very high level (≥96% or 100%). Only the "adjuvant cisplatin-based chemotherapy" indicator showed declining values between 2018 and 2020. The target values and plausibility limits were well achieved by LCC. After updating the guideline, one QI from 2018 was not included in the new QI set due to the small denominator population. Based on the new strong recommendations, 8 new QIs were defined. From the QI set of the guideline update, 13 of 15 indicators (7 since 2018 and 6 from 2022 on) were adopted into the certification program. CONCLUSIONS The guideline recommendations are implemented by LCC at a high level. The process presented confirms the successful implementation of the so-called quality cycle in oncology. The QIs developed by the German Guideline Program in Oncology (GGPO) are adopted by the certification program. The implementation of the QI is measured in LCC, evaluated by the German Cancer Society (DKG), and reflected back to the GGPO. The "real world" data have led to the deletion of one QI and show a high implementation of most QIs in LCC.
Collapse
Affiliation(s)
- Martin Utzig
- Zertifizierung, Deutsche Krebsgesellschaft e.V., Berlin, Germany
| | - Hans Hoffmann
- Division of Thoraxchirurgie, Klinikum rechts der Isar der Technischen Universität München, München, Germany
| | - Niels Reinmuth
- Thorakale Onkologie, Asklepios Fachkliniken München-Gauting, Gauting, Germany
| | - Wolfgang Schütte
- Klinik für Innere Medizin II, Krankenhaus Martha-Maria Halle-Dölau gGmbH, Halle, Germany
| | - Thomas Langer
- Leitlinienprogramm Onkologie, Deutsche Krebsgesellschaft e.V., Berlin, Germany
| | - Jessica Lobitz
- Wissensmanagement/Infonetz Krebs, Deutsche Krebsgesellschaft e.V., Berlin, Germany
| | - Johannes Rückher
- Zertifizierung, Deutsche Krebsgesellschaft e.V., Berlin, Germany
| | | |
Collapse
|
13
|
Fielding D, Lakis V, Dalley AJ, Chittoory H, Newell F, Koufariotis LT, Patch AM, Kazakoff S, Bashirzadeh F, Son JH, Ryan K, Steinfort D, Williamson JP, Bint M, Pahoff C, Nguyen PT, Twaddell S, Arnold D, Grainge C, Pattison A, Fairbairn D, Gune S, Christie J, Holmes O, Leonard C, Wood S, Pearson JV, Lakhani SR, Waddell N, Simpson PT, Nones K. Evaluation of Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration (EBUS-TBNA) Samples from Advanced Non-Small Cell Lung Cancer for Whole Genome, Whole Exome and Comprehensive Panel Sequencing. Cancers (Basel) 2024; 16:785. [PMID: 38398180 PMCID: PMC10887389 DOI: 10.3390/cancers16040785] [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: 12/22/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) is often the only source of tumor tissue from patients with advanced, inoperable lung cancer. EBUS-TBNA aspirates are used for the diagnosis, staging, and genomic testing to inform therapy options. Here we extracted DNA and RNA from 220 EBUS-TBNA aspirates to evaluate their suitability for whole genome (WGS), whole exome (WES), and comprehensive panel sequencing. For a subset of 40 cases, the same nucleic acid extraction was sequenced using WGS, WES, and the TruSight Oncology 500 assay. Genomic features were compared between sequencing platforms and compared with those reported by clinical testing. A total of 204 aspirates (92.7%) had sufficient DNA (100 ng) for comprehensive panel sequencing, and 109 aspirates (49.5%) had sufficient material for WGS. Comprehensive sequencing platforms detected all seven clinically reported tier 1 actionable mutations, an additional three (7%) tier 1 mutations, six (15%) tier 2-3 mutations, and biomarkers of potential immunotherapy benefit (tumor mutation burden and microsatellite instability). As expected, WGS was more suited for the detection and discovery of emerging novel biomarkers of treatment response. WGS could be performed in half of all EBUS-TBNA aspirates, which points to the enormous potential of EBUS-TBNA as source material for large, well-curated discovery-based studies for novel and more effective predictors of treatment response. Comprehensive panel sequencing is possible in the vast majority of fresh EBUS-TBNA aspirates and enhances the detection of actionable mutations over current clinical testing.
Collapse
Affiliation(s)
- David Fielding
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; (A.J.D.); (H.C.); (S.R.L.); (P.T.S.)
- Department of Thoracic Medicine, The Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia; (F.B.); (J.H.S.); (K.R.)
| | - Vanessa Lakis
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Andrew J. Dalley
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; (A.J.D.); (H.C.); (S.R.L.); (P.T.S.)
| | - Haarika Chittoory
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; (A.J.D.); (H.C.); (S.R.L.); (P.T.S.)
| | - Felicity Newell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Lambros T. Koufariotis
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Ann-Marie Patch
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Stephen Kazakoff
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Farzad Bashirzadeh
- Department of Thoracic Medicine, The Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia; (F.B.); (J.H.S.); (K.R.)
| | - Jung Hwa Son
- Department of Thoracic Medicine, The Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia; (F.B.); (J.H.S.); (K.R.)
| | - Kimberley Ryan
- Department of Thoracic Medicine, The Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia; (F.B.); (J.H.S.); (K.R.)
| | - Daniel Steinfort
- Department of Respiratory and Sleep Medicine, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; (D.S.); (J.C.)
| | - Jonathan P. Williamson
- Department of Thoracic Medicine, Liverpool Hospital Sydney, Sydney, NSW 2170, Australia;
| | - Michael Bint
- Department of Respiratory and Sleep Medicine, Sunshine Coast University Hospital, Birtinya, QLD 4575, Australia; (M.B.); (A.P.)
| | - Carl Pahoff
- Department of Thoracic Medicine, Gold Coast University Hospital, Southport, QLD 4215, Australia;
| | - Phan Tien Nguyen
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia;
| | - Scott Twaddell
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW 2305, Australia; (S.T.); (D.A.); (C.G.)
| | - David Arnold
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW 2305, Australia; (S.T.); (D.A.); (C.G.)
| | - Christopher Grainge
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW 2305, Australia; (S.T.); (D.A.); (C.G.)
| | - Andrew Pattison
- Department of Respiratory and Sleep Medicine, Sunshine Coast University Hospital, Birtinya, QLD 4575, Australia; (M.B.); (A.P.)
| | - David Fairbairn
- Pathology Queensland, The Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia;
| | - Shailendra Gune
- NSW Health Pathology South, Liverpool Hospital, Sydney, NSW 2170, Australia;
| | - Jemma Christie
- Department of Respiratory and Sleep Medicine, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; (D.S.); (J.C.)
| | - Oliver Holmes
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Conrad Leonard
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Scott Wood
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - John V. Pearson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; (A.J.D.); (H.C.); (S.R.L.); (P.T.S.)
- Pathology Queensland, The Royal Brisbane & Women’s Hospital, Brisbane, QLD 4006, Australia;
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
| | - Peter T. Simpson
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4029, Australia; (A.J.D.); (H.C.); (S.R.L.); (P.T.S.)
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (V.L.); (F.N.); (L.T.K.); (A.-M.P.); (S.K.); (O.H.); (C.L.); (S.W.); (J.V.P.); (N.W.); (K.N.)
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4067, Australia
| |
Collapse
|
14
|
Miller EJ, Galsky MD. Precision Medicine in Urothelial Carcinoma: Current Markers to Guide Treatment and Promising Future Directions. Curr Treat Options Oncol 2023; 24:1870-1888. [PMID: 38085403 DOI: 10.1007/s11864-023-01151-7] [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] [Accepted: 11/17/2023] [Indexed: 01/11/2024]
Abstract
OPINION STATEMENT The treatment landscape for urothelial cancer has changed dramatically in the last 10 years, with the approval of several new treatments. At the same time, profiling of individual tumors has become more commonplace with widespread availability of molecular testing and immunohistochemistry. For urothelial cancer, this has led to current guidelines recommending that molecular testing be obtained in the metastatic setting, and that it be considered in the setting of locally advanced disease. Between molecular testing and immunohistochemistry testing of tumors, the only current guideline-directed application of these tests is in the identification of FGFR3 or FGFR2 alterations for use of FGFR inhibitors. While additional recurrent molecular alterations linked to the pathogenesis of urothelial cancer have been identified, the ability to successfully "drug" the pathways association with such alterations remains limited. There has been extensive research into whether expression of particular proteins might inform specific treatment approaches such as the use of PD-L1 testing to guide immune checkpoint blockade. With the integration of antibody-drug conjugates into the treatment armamentarium for urothelial cancer, ongoing research is seeking to determine whether expression of the targets of these therapies, such as Nectin 4, Trop-2, or HER2, could help to guide treatment.
Collapse
Affiliation(s)
- Eric J Miller
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai/Tisch Cancer Institute, New York, NY, 10029, USA
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai/Tisch Cancer Institute, New York, NY, 10029, USA.
| |
Collapse
|
15
|
Fujimoto K, Matsumoto Y, Imabayashi T, Uchimura K, Furuse H, Tsuchida T. Suitability of respiratory endoscopy for sampling malignant thoracic tumors for comprehensive genomic profiling. Cancer Sci 2023; 114:4401-4412. [PMID: 37732498 PMCID: PMC10637053 DOI: 10.1111/cas.15951] [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: 05/06/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Comprehensive genomic profiling (CGP) is implemented to detect actionable gene aberrations and design matched therapies. Although malignant thoracic tumors are commonly detected through respiratory endoscopy, it is questionable whether the small specimens obtained thereof are sufficient for CGP. Therefore, this study aimed to investigate the suitability of respiratory endoscopy for sampling primary and metastatic thoracic tumors for CGP. Patients whose specimens were collected through respiratory endoscopy and assessed by pathologists to determine their suitability for CGP at our institution between June 2019 and May 2022 were reviewed retrospectively. The suitability of each procedure as a sampling technique for CGP and, in the cases actually analyzed, the distribution of the detected gene aberration were assessed. In total, 122 patients were eligible for analysis; the median age was 61 (range, 29-86) years, and 71 (58.2%) patients were male. Primary intrathoracic tumors were found in 91 (74.6%) cases, including 84 (68.9%) primary lung cancers; the remaining thoracic metastases of extrathoracic origin included various types. The suitability rates of specimens obtained using conventional bronchoscopy with and without cryobiopsy, endobronchial ultrasound-guided transbronchial needle aspiration, and medical thoracoscopy were 82.8% (24/29), 70.4% (19/27), 72.9% (35/48), and 100% (18/18), respectively. Of the 96 cases judged suitable, 83 were subjected to CGP, and all but one were successfully analyzed. Finally, 47 (56.6%) patients had at least one actionable gene aberration and eight (9.6%) were treated with the corresponding targeted therapies. In conclusion, specimens obtained through respiratory endoscopy are suitable for CGP; medical thoracoscopy and cryobiopsy in conventional bronchoscopy are particularly useful.
Collapse
Affiliation(s)
- Kazushi Fujimoto
- Department of Endoscopy, Respiratory Endoscopy DivisionNational Cancer Center HospitalTokyoJapan
- Department of Respiratory MedicineJapanese Red Cross Medical CenterTokyoJapan
| | - Yuji Matsumoto
- Department of Endoscopy, Respiratory Endoscopy DivisionNational Cancer Center HospitalTokyoJapan
- Department of Thoracic OncologyNational Cancer Center HospitalTokyoJapan
| | - Tatsuya Imabayashi
- Department of Endoscopy, Respiratory Endoscopy DivisionNational Cancer Center HospitalTokyoJapan
| | - Keigo Uchimura
- Department of Endoscopy, Respiratory Endoscopy DivisionNational Cancer Center HospitalTokyoJapan
| | - Hideaki Furuse
- Department of Endoscopy, Respiratory Endoscopy DivisionNational Cancer Center HospitalTokyoJapan
| | - Takaaki Tsuchida
- Department of Endoscopy, Respiratory Endoscopy DivisionNational Cancer Center HospitalTokyoJapan
| |
Collapse
|
16
|
Garassino MC, Oskar S, Arunachalam A, Zu K, Kao YH, Chen C, Meng W, Pietanza MC, Zhao B, Aggarwal H. Real-World Treatment Patterns and Outcomes of First-Line Immunotherapy Among Patients With Advanced Nonsquamous NSCLC Harboring BRAF, MET, or HER2 Alterations. JTO Clin Res Rep 2023; 4:100568. [PMID: 37744307 PMCID: PMC10514206 DOI: 10.1016/j.jtocrr.2023.100568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Data on utilization and clinical outcomes of programmed cell death protein or programmed death-ligand 1 (PD-[L]1) inhibitors in NSCLC with uncommon oncogenic alterations is limited. Methods This retrospective study used a deidentified U.S. nationwide clinicogenomic database to select patients with advanced nonsquamous NSCLC without EGFR, ALK, or ROS1 alterations, diagnosed from January 1, 2016 to September 30, 2020, who initiated first-line therapy. Our objectives were to summarize characteristics and treatment patterns for patients with four little-studied genomic alterations or driver-negative NSCLC. We estimated Kaplan-Meier real-world time on treatment (rwTOT) and time to next treatment for patients receiving PD-(L)1 inhibitors. The data cutoff was September 30, 2021. Results Of the 3971 eligible patients, 84 (2%) had NSCLC with BRAF V600E mutation, 117 (3%) had MET exon 14 skipping mutation, 130 (3%) had MET amplification, 91 (2%) had ERBB2 activation mutation, and 691 patients (17%) had driver-negative NSCLC. Patient characteristics differed among cohorts as expected. The most common first-line regimen in each cohort was a PD-(L)1 inhibitor as monotherapy or in combination with chemotherapy. The median rwTOT with anti-PD-(L)1 monotherapy was 4.6 months in the driver-negative cohort and ranged from 2.9 months (ERBB2 mutation) to 7.6 months (BRAF V600E mutation). The median rwTOT with anti-PD-(L)1-chemotherapy combination was 5.2 months in the driver-negative cohort and 6 months in all but the BRAF V600E cohort (17.5 mo). The patterns of real-world time to next treatment results were similar. Conclusions Substantial use of anti-PD-(L)1 therapy and associated clinical outcomes are consistent with previous real-world findings and suggest no detriment from PD-(L)1 inhibitors for advanced nonsquamous NSCLC harboring one of these four genomic alterations relative to driver-negative NSCLC.
Collapse
Affiliation(s)
- Marina C. Garassino
- Thoracic Oncology Program, Section of Hematology Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Sabine Oskar
- Center for Observational and Real-World Evidence (CORE), Merck & Co., Inc., Rahway, New Jersey
| | - Ashwini Arunachalam
- Center for Observational and Real-World Evidence (CORE), Merck & Co., Inc., Rahway, New Jersey
| | - Ke Zu
- Epidemiology, Merck & Co., Inc., Rahway, New Jersey
| | - Yu-Han Kao
- Center for Observational and Real-World Evidence (CORE), Merck & Co., Inc., Rahway, New Jersey
| | - Cai Chen
- Data, AI and Genome Sciences (DAGS) Department, Merck & Co., Inc., Rahway, New Jersey
| | - Weilin Meng
- Center for Observational and Real-World Evidence (CORE), Merck & Co., Inc., Rahway, New Jersey
| | | | - Bin Zhao
- Clinical Research, Merck & Co., Inc., Rahway, New Jersey
| | - Himani Aggarwal
- Center for Observational and Real-World Evidence (CORE), Merck & Co., Inc., Rahway, New Jersey
| |
Collapse
|
17
|
Vrinzen CEJ, Bloemendal HJ, Jeurissen PPT. How to create value with constrained budgets in oncological care? A narrative review. Expert Rev Pharmacoecon Outcomes Res 2023; 23:989-999. [PMID: 37650221 DOI: 10.1080/14737167.2023.2253375] [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: 06/15/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
INTRODUCTION As a result of an increasing focus on patient-centered care within oncology and more pressure on the sustainability of health-care systems, the discussion on what exactly constitutes value re-appears. Policymakers seek to improve patient values; however, funding all values is not sustainable. AREAS COVERED We collect available evidence from scientific literature and reflect on the concept of value, the possible incorporation of a wide spectrum of values in reimbursement decisions, and alternative strategies to increase value in oncological care. EXPERT OPINION We state that value holds many different aspects. For reimbursement decisions, we argue that it is simply not feasible to incorporate all patient values because of the need for efficient resource allocation. We argue that we should shift the value debate from the individual perspective of patients to creating value for the cancer population at large. The different strategies we address are as follows: (1) shared decision-making; (2) biomarkers and molecular diagnostics; (3) appropriate evaluation, payment and use of drugs; (4) supportive care; (5) cancer prevention and screening; (6) monitoring late effect; (7) concentration of care and oncological networking; and (8) management of comorbidities. Important preconditions to support these strategies are strategic planning, consistent cancer policies and data availability.
Collapse
Affiliation(s)
- Cilla E J Vrinzen
- Scientific Institute for Quality of Healthcare, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Haiko J Bloemendal
- Department of Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Patrick P T Jeurissen
- Scientific Institute for Quality of Healthcare, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Čerina D, Krpina K, Jakopović M, Dedić Plavetić N, Seiwerth F, Tomić S, Radić J, Belac Lovasić I, Canjko I, Boban M, Samaržija M, Vrdoljak E. The Challenges and Opportunities of the Implementation of Comprehensive Genomic Profiling in Everyday Clinical Practice with Non-Small Cell Lung Cancer: National Results from Croatia. Cancers (Basel) 2023; 15:3395. [PMID: 37444505 DOI: 10.3390/cancers15133395] [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: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) has become the best example of precision oncology's impact on outcomes in everyday clinical practice, significantly changing the expectations of all stakeholders, including medical professionals, society, and most importantly, patients. Consequently, the implementation of the precision oncology concept in medical systems, in order to achieve optimal and proven curative effects in NSCLC, is imperative. In this study, we investigated the development, challenges, and results associated with the implementation of precision oncology in NSCLC on a national level in Croatia. We conducted a multicenter, retrospective, cross-sectional analysis on the total population of Croatian patients with metastatic lung cancer, on whose tumors specimen comprehensive genomic profiling (CGP) testing was performed during 2020 and 2021. A total of 48 patients were included in the study. CGP revealed clinically relevant genomic alterations (CRGA) in 37 patients (79%), with a median of 2 (IQR 1-3) CRGA per patient. From the panel of recommended tests, KRAS, MET, and EGFR were the most common alterations, detected in 16 (34%), 5 (11%), and 3 (6%) patients, respectively. CGP revealed additional targetable mutations in 29 (60%) patients who would not have been tested (and consequently, whose mutations would not have been detected) according to the existing everyday standard of practice in Croatia. The tumor mutational burden was reported as high (≥10 Muts/Mb) in 19 patients (40%). CGP analysis reported some kind of targeted therapy for 34 patients (72%). CGP revealed other potentially targetable mutations, and it also determined TMB to be high in a significant number of patients. In conclusion, when possible, CGP should be used as an upfront backbone diagnostic and treatment-oriented work-up in patients with NSCLC.
Collapse
Affiliation(s)
- Dora Čerina
- Department of Oncology, University Hospital Center Split, 21000 Split, Croatia
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Kristina Krpina
- Department for Respiratory Diseases Jordanovac, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Marko Jakopović
- Department for Respiratory Diseases Jordanovac, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Natalija Dedić Plavetić
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Fran Seiwerth
- Department for Respiratory Diseases Jordanovac, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Snježana Tomić
- School of Medicine, University of Split, 21000 Split, Croatia
- Department of Pathology, Citology and Forensic Medicine, University Hospital Center Split, 21000 Split, Croatia
| | - Jasna Radić
- Department of Oncology and Nuclear Medicine, Division of Medical Oncology, University Hospital for Tumors, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
| | | | - Ivana Canjko
- Department of Radiotherapy Oncology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Marijo Boban
- Department of Oncology, University Hospital Center Split, 21000 Split, Croatia
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Miroslav Samaržija
- Department for Respiratory Diseases Jordanovac, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Eduard Vrdoljak
- Department of Oncology, University Hospital Center Split, 21000 Split, Croatia
- School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
19
|
Fielding DI, Dalley AJ, Singh M, Nandakumar L, Lakis V, Chittoory H, Fairbairn D, Patch AM, Kazakoff SH, Ferguson K, Bashirzadeh F, Bint M, Pahoff C, Son JH, Hodgson A, Sharma S, Waddell N, Lakhani SR, Hartel G, Nones K, Simpson PT. Evaluating Diff-Quik cytology smears for large-panel mutation testing in lung cancer-Predicting DNA content and success with low-malignant-cellularity samples. Cancer Cytopathol 2023. [PMID: 36938641 DOI: 10.1002/cncy.22690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 03/21/2023]
Abstract
BACKGROUND Cytology smears are commonly collected during endobronchial ultrasound-guided transbronchial needle aspiration (EBUS TBNA) procedures but are rarely used for molecular testing. Studies are needed to demonstrate their great potential, in particular for the prediction of malignant cell DNA content and for utility in molecular diagnostics using large gene panels. METHODS A prospective study was performed on samples from 66 patients with malignant lymph nodes who underwent EBUS TBNA. All patients had air-dried, Diff-Quik cytology smears and formalin-fixed, paraffin-embedded cell blocks collected for cytopathology and molecular testing. One hundred eighty-five smears were evaluated by microscopy to estimate malignant cell percentage and abundance and to calculate smear size and were subjected to DNA extraction. DNA from 56 smears from 27 patients was sequenced with the TruSight Oncology 500 assay (Illumina). RESULTS Each microscopy parameter had a significant effect on the DNA yield. An algorithm was developed that predicted a >50-ng DNA yield of a smear with an area under the curve of 0.86. Fifty DNA samples (89%) with varying malignant yields were successfully sequenced. Low-malignant-cell content (<25%) and smear area (<15%) were the main reasons for failure. All standard-of-care mutations were detected in replicate smears from individual patients, regardless of malignant cell content. Tier 1/2 mutations were discovered in two cases where standard-of-care specimens were inadequate for sequencing. Smears were scored for tumor mutation burden. CONCLUSIONS Microscopy of Diff-Quik smears can triage samples for comprehensive panel sequencing, which highlights smears as an excellent alternative to traditional testing with cell blocks.
Collapse
Affiliation(s)
- David I Fielding
- Department of Thoracic Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Andrew J Dalley
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Mahendra Singh
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Lakshmy Nandakumar
- Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Vanessa Lakis
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Haarika Chittoory
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David Fairbairn
- Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Ann-Marie Patch
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Stephen H Kazakoff
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kaltin Ferguson
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Farzad Bashirzadeh
- Department of Thoracic Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Michael Bint
- Department of Thoracic Medicine, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Carl Pahoff
- Department of Respiratory Medicine, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Jung Hwa Son
- Department of Thoracic Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Alan Hodgson
- Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Sowmya Sharma
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- ACL Pathology, Bellavista, New South Wales, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Gunter Hartel
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Peter T Simpson
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
20
|
Burns L, Jani C, Radwan A, Omari OA, Patel M, Oxnard GR, Tapan U. Implementation Challenges and Disparities in Molecular Testing for Patients With Stage IV NSCLC: Perspectives from an Urban Safety-Net Hospital. Clin Lung Cancer 2023; 24:e69-e77. [PMID: 36464575 DOI: 10.1016/j.cllc.2022.11.002] [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: 06/29/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
The advent of next-generation sequencing (NGS), including both tissue assays and circulating tumor DNA (ct-DNA), has been pivotal in improving outcomes for patients with non-small cell lung cancer (NSCLC). Although molecular testing is standard of care for advanced NSCLC, challenges still exist in its implementation. This Perspective examines barriers to the widespread implementation of NGS from the vantage point of a single urban safety-net institution, with a particular focus on examining racial disparities in NGS completion. We conducted a review of patients at our institution from January 2015 through January 2022 and examined molecular testing patterns before and after the publication of updated molecular testing guidelines from the International Association for the Study of Lung Cancer (IASLC), Association for Molecular Pathology (AMP), and College of American Pathologists (CAP) in March of 2018. While NGS increased over time, we found that 43% of patients in the March 2018 through January 2022 group still did not receive NGS, and the most common reasons for the absence of testing included a lack of physician ordering and insufficient tissue on biopsy. We did not note any racial disparities in completion or time-to-adoption of NGS. Patients with squamous cell carcinoma (SCC) histology were noted to receive liquid NGS markedly less often than patients with non-squamous histology in the March 2018 through January 2022 period. Based on our own data and a review of findings from colleagues in the field, we advocate for additional physician educational programming, increased use of ct-DNA biopsy, automated (reflexive) NGS tissue testing on receipt of biopsy, and consideration for the broader molecular profiling of patients with SCC histology.
Collapse
Affiliation(s)
- Laura Burns
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA.
| | - Chinmay Jani
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA
| | - Amr Radwan
- Section of Hematology & Medical Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| | - Omar Al Omari
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA
| | - Mohini Patel
- Boston University School of Public Health and Boston Medical Center, Boston, MA
| | - Geoffrey R Oxnard
- Section of Hematology & Medical Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| | - Umit Tapan
- Section of Hematology & Medical Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| |
Collapse
|
21
|
García-Pardo M, Aparicio I, Martínez Í, Arregui M, Tirado V, Galera M, Álvarez R, Calles A. Brief Report: Clinical Outcomes Using Plasma-Based Molecular Profiling to Guide Treatment Decisions in Patients With Advanced NSCLC and Limited Access to Broad Tissue Testing. Clin Lung Cancer 2023; 24:366-370. [PMID: 36842853 DOI: 10.1016/j.cllc.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Affiliation(s)
- Miguel García-Pardo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada; Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | - Marta Arregui
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Victoria Tirado
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Mar Galera
- Hospital General Universitario Gregorio Marañón, Madrid, Spain; Fundación de Investigación Biomédica del Hospital Gregorio Marañón, Madrid, Spain
| | - Rosa Álvarez
- Hospital General Universitario Gregorio Marañón, Madrid, Spain; Fundación de Investigación Biomédica del Hospital Gregorio Marañón, Madrid, Spain
| | - Antonio Calles
- Hospital General Universitario Gregorio Marañón, Madrid, Spain; Fundación de Investigación Biomédica del Hospital Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
22
|
Harada Y, Sato A, Nakamura H, Kai K, Kitamura S, Nakamura T, Kurihara Y, Ikeda S, Sueoka E, Kimura S, Sueoka-Aragane N. Anti-cancer effect of afatinib, dual inhibitor of HER2 and EGFR, on novel mutation HER2 E401G in models of patient-derived cancer. BMC Cancer 2023; 23:77. [PMID: 36690964 PMCID: PMC9872313 DOI: 10.1186/s12885-022-10428-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/08/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Precision medicine with gene panel testing based on next-generation sequencing for patients with cancer is being used increasingly in clinical practice. HER2, which encodes the human epidermal growth factor receptor 2 (HER2), is a potentially important driver gene. However, therapeutic strategies aimed at mutations in the HER2 extracellular domain have not been clarified. We therefore investigated the effect of EGFR co-targeted therapy with HER2 on patient-derived cancer models with the HER2 extracellular domain mutation E401G, based on our previous findings that this mutation has an epidermal growth factor receptor (EGFR)-mediated activation mechanism. METHODS We generated a xenograft (PDX) and a cancer tissue-originated spheroid (CTOS) from a patient's cancer containing an amplified HER2 E401G mutation. With these platforms, we compared the efficacy of afatinib, a tyrosine kinase inhibitor having anti-HER2 and anti-EGFR activity, with two other therapeutic options: lapatinib, which has similar properties but weaker EGFR inhibition, and trastuzumab plus pertuzumab, for which evidence exists of treatment efficacy against cancers with wild-type HER2 amplification. Similar experiments were also performed with H2170, a cell line with wild-type HER2 amplification, to contrast the characteristics of these drug's efficacies against HER2 E401G. RESULTS We confirmed that PDX and CTOS retained morphological and immunohistochemical characteristics and HER2 gene profiles of the original tumor. In both PDX and CTOS, afatinib reduced tumor size more than lapatinib or trastuzumab plus pertuzumab. In addition, afatinib treatment resulted in a statistically significant reduction in HER2 copy number at the end of treatment. On the other hand, in H2170 xenografts with wild-type HER2 amplification, trastuzumab plus pertuzumab was most effective. CONCLUSIONS Afatinib, a dual inhibitor of HER2 and EGFR, showed a promising effect on cancers with amplified HER2 E401G, which have an EGFR-mediated activation mechanism. Analysis of the activation mechanisms of mutations and development of therapeutic strategies based on those mechanisms are critical in precision medicine for cancer patients.
Collapse
Affiliation(s)
- Yohei Harada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
- Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akemi Sato
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hideaki Nakamura
- Department of Transfusion Medicine, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Sho Kitamura
- Department of Pathology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Tomomi Nakamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Eisaburo Sueoka
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| |
Collapse
|
23
|
Trapani D, Crimini E, Sandoval J, Curigliano G. Next-Generation Sequencing for Advanced Breast Cancer: What the Way to Go? Cancer Treat Res 2023; 188:343-351. [PMID: 38175352 DOI: 10.1007/978-3-031-33602-7_13] [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: 01/05/2024]
Abstract
The rapid implementation of precision medicine tools in diagnosing and treating breast cancer (BC) has widened the potential therapeutic options for patients. The applications of gene sequencing, including next-generation gene sequencing (NGS), have led to numerous questions on how to validate, implement, interpret, prioritize and operationalize precision medicine tools to deliver meaningful and impactful interventions. Limited benefit has been portended with earlier experiences of NGS-driven treatment, in BC. However, the development and use of frameworks of clinical actionability of genomic alterations, for example, detected with NGS, has resulted in better patient selection, and potentially higher therapeutic value. The European Society for Medical Oncology Scale for Clinical Actionability of molecular Targets (ESCAT) is a framework that includes five tiers of clinical actionability, with tier 1 reserved for approved drugs with demonstrated benefits for targetable genomic alterations. The re-analysis of clinical studies by grouping the genomic alterations and matched drugs with ESCAT, in high vs lower tiers has demonstrated a significant benefit portended by high tiers alterations, with the availability of efficacious treatments. As a result, frameworks for actionability, like ESCAT, should be fundamental in developing and implementing NGS-driven, and broadly, precision medicine research and treatments.
Collapse
Affiliation(s)
- Dario Trapani
- Division of New Drug Development for innovative therapies, European Institute of Oncology IRCCS, Milan, Italy.
| | - Edoardo Crimini
- Division of New Drug Development for innovative therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - José Sandoval
- Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Unit of Population Epidemiology, Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Giuseppe Curigliano
- Division of New Drug Development for innovative therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Fintelmann FJ, Martin NA, Tahir I, Quinn EM, Allen TC, Joseph L, Nikolic B, Lee C. Optimizing molecular testing of lung cancer needle biopsy specimens: potential solutions from an interdisciplinary qualitative study. Respir Res 2023; 24:17. [PMID: 36650544 PMCID: PMC9847026 DOI: 10.1186/s12931-023-02321-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Molecular testing can detect actionable genomic alterations and tumor cell surface proteins in patients with non-small cell lung cancer (NSCLC). However, utilization remains suboptimal, representing missed treatment opportunities. This study aimed to identify challenges and potential solutions to obtaining percutaneous lung needle biopsy specimens for successful molecular testing in patients with advanced NSCLC. METHODS This interdisciplinary qualitative study included ten radiologists and four pathologists from academic and community settings across the United States who routinely perform and analyze percutaneous lung needle biopsies. Participants underwent semi-structured one-on-one interviews (Phase 1). Interview questionnaires were constructed based on a literature review of key lines of inquiry and conducted by professional market researchers using the theoretical domains framework. Primary barriers to molecular testing were identified using thematic analysis. Subsequently, multidisciplinary focus groups were convened to identify potential solutions (Phase 2). RESULTS Four themes emerged as barriers to molecular testing and were matched to the clinical workflow: (1) biopsy request, (2) biopsy procedure, (3) specimen analysis, and (4) communication. The nineteen potential solutions included adding a "checkbox" to indicate molecular testing in the biopsy request, leveraging pre-procedural imaging to guide biopsies, conserving tissue through appropriate allocation strategies and next generation sequencing panels instead of sequential single-gene assays, instituting reflex-molecular testing upon NSCLC diagnosis, tracking and communicating biopsy outcomes at multidisciplinary tumor boards, and improving integration of radiologists and pathologists into oncology care teams. CONCLUSIONS Potential solutions exist to increase successful molecular testing of lung needle biopsy specimens in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Florian J. Fintelmann
- grid.32224.350000 0004 0386 9924Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Nikki A. Martin
- grid.443873.f0000 0004 0422 4933LUNGevity Foundation, Bethesda, MD USA
| | - Ismail Tahir
- grid.32224.350000 0004 0386 9924Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Elissa M. Quinn
- grid.497611.c0000 0004 1794 1958Blueprint Medicines, Boston, MA USA
| | | | - Lija Joseph
- grid.461527.30000 0004 0383 4123Lowell General Hospital, Lowell, MA USA
| | - Boris Nikolic
- grid.439147.c0000 0004 0628 7583Wyoming Valley Radiology Associates, Wilkes-Barre General Hospital, Wilkes-Barre, PA USA
| | - Christopher Lee
- grid.50956.3f0000 0001 2152 9905Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA USA
| |
Collapse
|
25
|
Wu N, Ge W, Quek RG, Gleeson M, Pouliot JF, Dietz H, Jalbert JJ, Harnett J, Antonia SJ. Trends in real-world biomarker testing and overall survival in US patients with advanced non-small-cell lung cancer. Future Oncol 2022; 18:4385-4397. [PMID: 36656547 DOI: 10.2217/fon-2022-0540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background: Trends/outcomes associated with National Comprehensive Cancer Network (NCCN)-recommended biomarker testing to guide advanced non-small-cell lung cancer (aNSCLC) treatment were assessed. Methods: Patients initiating first-line aNSCLC treatment were included using a nationwide electronic health record-derived database (1/1/2015-10/31/2021). Trends in pre-first-line biomarker testing (PD-L1, major genomic aberrations), factors associated with testing and associations between testing and outcomes were assessed. Results: PD-L1/genomic aberration testing rates increased from 33% (2016) to 81% (2018), then plateaued. Certain clinical and demographic factors were associated with a greater likelihood of PD-L1 testing. Patients tested for PD-L1 or genomic aberrations had longer overall survival (OS). Conclusion: Biomarker testing may be associated with improved OS in aNSCLC, though not all patients had equal access to testing.
Collapse
Affiliation(s)
- Ning Wu
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Wenzhen Ge
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Ruben Gw Quek
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Michelle Gleeson
- Genesis Research, LLC, 111 River Street, Suite 1120, Hoboken, NJ 07030, USA
| | - Jean-Francois Pouliot
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Hilary Dietz
- Center for Cancer Immunotherapy, Duke University School of Medicine, 20 Duke Medicine Cir, Durham, NC 27710, USA
| | - Jessica J Jalbert
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - James Harnett
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Scott J Antonia
- Center for Cancer Immunotherapy, Duke University School of Medicine, 20 Duke Medicine Cir, Durham, NC 27710, USA
| |
Collapse
|