1
|
Bergstrom EN, Abbasi A, Díaz-Gay M, Galland L, Ladoire S, Lippman SM, Alexandrov LB. Deep Learning Artificial Intelligence Predicts Homologous Recombination Deficiency and Platinum Response From Histologic Slides. J Clin Oncol 2024; 42:3550-3560. [PMID: 39083703 PMCID: PMC11469627 DOI: 10.1200/jco.23.02641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/28/2024] [Indexed: 08/02/2024] Open
Abstract
PURPOSE Cancers with homologous recombination deficiency (HRD) can benefit from platinum salts and poly(ADP-ribose) polymerase inhibitors. Standard diagnostic tests for detecting HRD require molecular profiling, which is not universally available. METHODS We trained DeepHRD, a deep learning platform for predicting HRD from hematoxylin and eosin (H&E)-stained histopathological slides, using primary breast (n = 1,008) and ovarian (n = 459) cancers from The Cancer Genome Atlas (TCGA). DeepHRD was compared with four standard HRD molecular tests using breast (n = 349) and ovarian (n = 141) cancers from multiple independent data sets, including platinum-treated clinical cohorts with RECIST progression-free survival (PFS), complete response (CR), and overall survival (OS) endpoints. RESULTS DeepHRD predicted HRD from held-out H&E-stained breast cancer slides in TCGA with an AUC of 0.81 (95% CI, 0.77 to 0.85). This performance was confirmed in two independent primary breast cancer cohorts (AUC, 0.76 [95% CI, 0.71 to 0.82]). In an external platinum-treated metastatic breast cancer cohort, samples predicted as HRD had higher complete CR (AUC, 0.76 [95% CI, 0.54 to 0.93]) with 3.7-fold increase in median PFS (14.4 v 3.9 months; P = .0019) and hazard ratio (HR) of 0.45 (P = .0047). There were no significant differences in nonplatinum treatment outcome by predicted HRD status in three breast cancer cohorts, including CR (AUC, 0.39) and PFS (HR, 0.98, P = .95) in taxane-treated metastatic breast cancer. Through transfer learning to high-grade serous ovarian cancer, DeepHRD-predicted HRD samples had better OS after first-line (HR, 0.46; P = .030) and neoadjuvant (HR, 0.49; P = .015) platinum therapy in two cohorts. CONCLUSION DeepHRD can predict HRD in breast and ovarian cancers directly from routine H&E slides across multiple external cohorts, slide scanners, and tissue fixation variables. When compared with molecular testing, DeepHRD classified 1.8- to 3.1-fold more patients with HRD, which exhibited better OS in high-grade serous ovarian cancer and platinum-specific PFS in metastatic breast cancer.
Collapse
Affiliation(s)
- Erik N. Bergstrom
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Ammal Abbasi
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Marcos Díaz-Gay
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Loïck Galland
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
- Platform of Transfer in Biological Oncology, Centre Georges-François Leclerc, Dijon, France
- University of Burgundy-Franche Comté, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
- Platform of Transfer in Biological Oncology, Centre Georges-François Leclerc, Dijon, France
- University of Burgundy-Franche Comté, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | | | - Ludmil B. Alexandrov
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA
| |
Collapse
|
2
|
Murai T, Kasai Y, Eguchi Y, Takano S, Kita N, Torii A, Takaoka T, Tomita N, Shibamoto Y, Hiwatashi A. Fractionated Stereotactic Intensity-Modulated Radiotherapy for Large Brain Metastases: Comprehensive Analyses of Dose-Volume Predictors of Radiation-Induced Brain Necrosis. Cancers (Basel) 2024; 16:3327. [PMID: 39409947 PMCID: PMC11482639 DOI: 10.3390/cancers16193327] [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: 08/09/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The objective was to explore dosimetric predictors of brain necrosis (BN) in fractionated stereotactic radiotherapy (SRT). METHODS After excluding collinearities carefully, multivariate logistic models were developed for comprehensive analyses of dosimetric predictors in patients who received first-line fractionated SRT for brain metastases (BMs). The normal brain volume receiving an xx Gy biological dose in 2 Gy fractions (VxxEQD2) was calculated from the retrieved dose-volume parameters. RESULTS Thirty Gy/3 fractions (fr) SRT was delivered to 34 patients with 75 BMs (median target volume, 3.2 cc), 35 Gy/5 fr to 30 patients with 57 BMs (6.4 cc), 37.5 Gy/5 fr to 28 patients with 47 BMs (20.2 cc), and 40 Gy/10 fr to 20 patients with 37 BMs (24.3 cc), according to protocols, depending on the total target volume (p < 0.001). After excluding the three-fraction groups, the incidence of symptomatic BN was significantly higher in patients with a larger V50EQD2 (adjusted odds ratio: 1.07, p < 0.02), V55EQD2 (1.08, p < 0.01), or V60EQD2 (1.09, p < 0.01) in the remaining five- and ten-fraction groups. The incidence of BN was also significantly higher in cases with V55EQD2 > 30 cc or V60EQD2 > 20 cc (p < 0.05). These doses correspond to 28 or 30 Gy/5 fr and 37 or 40 Gy/10 fr, respectively. CONCLUSIONS In five- or ten-fraction SRT, larger V55EQD2 or V60EQD2 are BN risk predictors. These biologically high doses may affect BN incidence. Thus, the planning target volume margin should be minimized as much as possible.
Collapse
Affiliation(s)
- Taro Murai
- Department of Radiation Oncology, Shonan Kamakura General Hospital, 1370-1 Okamoto, Kamakura 247-8533, Kanagawa, Japan
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| | - Yuki Kasai
- Department of Radiology, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8602, Aichi, Japan; (Y.K.); (Y.E.)
| | - Yuta Eguchi
- Department of Radiology, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8602, Aichi, Japan; (Y.K.); (Y.E.)
| | - Seiya Takano
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| | - Nozomi Kita
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| | - Akira Torii
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| | - Taiki Takaoka
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| | - Natsuo Tomita
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| | - Yuta Shibamoto
- Narita Memorial Proton Center, 78 Shirakawa-cho, Toyohashi 441-8021, Aichi, Japan;
| | - Akio Hiwatashi
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya 467-8601, Aichi, Japan; (S.T.); (N.K.); (A.T.); (T.T.); (N.T.); (A.H.)
| |
Collapse
|
3
|
Evans RT, Gillon-Zhang E, Brown JN, Knudsen KE, King C, Green AS, Silva AL, Mordaka JM, Palmer RN, Tomassini A, Collazos A, Xyrafaki C, Turner I, Ho CH, Nugent D, Jose J, Andreazza S, Potts ND, von Bargen K, Gray ER, Stolarek-Januszkiewicz M, Cooke A, Reddi HV, Balmforth BW, Osborne RJ. ASPYRE-Lung: validation of a simple, fast, robust and novel method for multi-variant genomic analysis of actionable NSCLC variants in FFPE tissue. Front Oncol 2024; 14:1420162. [PMID: 39386190 PMCID: PMC11461167 DOI: 10.3389/fonc.2024.1420162] [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: 04/19/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Genomic variant testing of tumors is a critical gateway for patients to access the full potential of personalized oncology therapeutics. Current methods such as next-generation sequencing are costly and challenging to interpret, while PCR assays are limited in the number of variants they can cover. We developed ASPYRE® (Allele-Specific PYrophosphorolysis REaction) technology to address the urgent need for rapid, accessible and affordable diagnostics informing actionable genomic target variants of a given cancer. The targeted ASPYRE-Lung panel for non-small cell carcinoma covers 114 variants in 11 genes (ALK, BRAF, EGFR, ERBB2, KRAS, RET, ROS1, MET & NTRK1/2/3) to robustly inform clinical management. The assay detects single nucleotide variants, insertions, deletions, and gene fusions from tissue-derived DNA and RNA simultaneously. Methods We tested the limit of detection, specificity, analytical accuracy and analytical precision of ASPYRE-Lung using FFPE lung tissue samples from patients with non-small cell lung carcinoma, variant-negative FFPE tissue from healthy donors, and FFPE-based contrived samples with controllable variant allele fractions. Results The sensitivity of ASPYRE-Lung was determined to be ≤ 3% variant allele fraction for single nucleotide variants and insertions or deletions, 100 copies for fusions, and 200 copies for MET exon 14 skipping. The specificity was 100% with no false positive results. The analytical accuracy test yielded no discordant calls between ASPYRE-Lung and expected results for clinical samples (via orthogonal testing) or contrived samples, and results were replicable across operators, reagent lots, runs, and real-time PCR instruments with a high degree of precision. Conclusions The technology is simple and fast, requiring only four reagent transfer steps using standard laboratory equipment (PCR and qPCR instruments) with analysis via a cloud-based algorithm. The ASPYRE-Lung assay has the potential to be transformative in facilitating access to rapid, actionable molecular profiling of tissue for patients with non-small cell carcinoma.
Collapse
Affiliation(s)
| | | | | | | | - Candace King
- Biofidelity Inc., Morrisville, NC, United States
| | | | | | | | | | | | | | | | | | - Chau Ha Ho
- Biofidelity Ltd., Cambridge, United Kingdom
| | | | - Jinsy Jose
- Biofidelity Ltd., Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Bestvina CM, Waters D, Morrison L, Emond B, Lafeuille MH, Hilts A, Mujwara D, Lefebvre P, He A, Vanderpoel J. Impact of next-generation sequencing vs polymerase chain reaction testing on payer costs and clinical outcomes throughout the treatment journeys of patients with metastatic non-small cell lung cancer. J Manag Care Spec Pharm 2024:1-12. [PMID: 39259000 DOI: 10.18553/jmcp.2024.24137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
BACKGROUND For patients with metastatic non-small cell lung cancer (mNSCLC), next-generation sequencing (NGS) biomarker testing has been associated with a faster time to appropriate targeted therapy and more comprehensive testing relative to polymerase chain reaction (PCR) testing. However, the impact on payer costs and clinical outcomes during patients' treatment journeys has not been fully characterized. OBJECTIVE To assess the costs and clinical outcomes of NGS vs PCR biomarker testing among patients with newly diagnosed de novo mNSCLC from a US payers' perspective. METHODS A Markov model assessed costs and clinical outcomes of NGS vs PCR testing from the start of testing up to 3 years after. Patients entered the model after receiving biomarker test results and then initiated first-line (1L) targeted or nontargeted therapy (immunotherapy and/or chemotherapy) depending on actionable mutation detection. A few patients with an actionable mutation were not detected by PCR and inappropriately initiated 1L nontargeted therapy. At each 1-month cycle, patients could remain on treatment with 1L, progress to second line or later (2L+), or die. Literature-based inputs included the rates of progression-free survival (PFS) and overall survival (OS), targeted and nontargeted therapy costs, total costs of testing, and medical costs of 1L, 2L+, and death. Per patient average PFS and OS as well as cumulative costs were reported for NGS and PCR testing. RESULTS In a modeled population of 100 patients (75% commercial and 25% Medicare), 45.9% of NGS and 40.0% of PCR patients tested positive for an actionable mutation. Relative to PCR, NGS was associated with $7,386 in savings per patient (NGS = $326,154; PCR = $333,540) at 1 year, driven by lower costs of testing, including estimated costs of delayed care and nontargeted therapy initiation before receiving test results (NGS = $8,866; PCR = $16,373). Treatment costs were similar (NGS = $305,644; PCR = $305,283). In the PCR cohort, the per patient costs of inappropriate 1L nontargeted therapy owing to undetected mutations were $6,455, $6,566, and $6,569 over the first 1, 2, and 3 years, respectively. Relative to PCR testing, NGS was associated with $4,060 in savings at 2 years and $1,092 at 3 years. Patients who initiated 1L targeted therapy had an additional 5.4, 8.8, and 10.4 months of PFS and an additional 1.4, 3.6, and 5.3 months of OS over the first 1, 2, and 3 years, respectively, relative to those who inappropriately initiated 1L nontargeted therapy. CONCLUSIONS In this Markov model, as early as year 1, and over 3 years following biomarker testing, patients with newly diagnosed de novo mNSCLC undergoing NGS testing are projected to have cost savings and longer PFS and OS relative to those tested with PCR.
Collapse
Affiliation(s)
| | - Dexter Waters
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| | | | | | | | | | | | | | - Andy He
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| | - Julie Vanderpoel
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| |
Collapse
|
5
|
Megahed AI, Zheng D, Chen LH, Haque R, McGary EC. Half of oncologists fail to use ordered NGS results to guide their first-line treatment decision in advanced NSCLC: A retrospective study in a community-based integrated healthcare system. Heliyon 2024; 10:e36308. [PMID: 39262959 PMCID: PMC11388373 DOI: 10.1016/j.heliyon.2024.e36308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/13/2024] Open
Abstract
Purpose Next generation sequencing (NGS) testing is used to identify driver mutation(s) in non-small cell lung cancer (NSCLC) that are amenable to targeted therapy, resulting in superior outcomes and improved tolerability. We characterized how clinicians in a large integrated healthcare system utilized NGS testing to inform first line treatment decisions in patients with stage IV NSCLC shortly after diagnosis. Methods We conducted a cross-sectional study of 964 patients within an integrated healthcare system, Kaiser Permanente Southern California (KPSC), who were diagnosed with stage IV NSCLC and completed NGS testing (Strata Oncology) between May 2019 to June 2021. Treatment start dates were used to divide patients into those who started treatment before or after NGS results, or those who did not receive treatment after NGS results. Patients harboring alterations in seven genes (EGFR, ALK, ROS-1, BRAF, KRAS, RET, and MET) were considered candidates for targeted first line therapy. Results First line treatment was initiated in half (52 %; n = 284) of all treated patients prior to NGS results. Just under half (48 %; n = 137) of these patients were found to have a targetable mutation by NGS, of whom 59 % received first line chemotherapy and/or immunotherapy, rather than targeted therapy. Nearly 27 % of the sample never received treatment, of which 31 % had a targetable mutation, and may have been candidates for targeted therapy. Not undergoing first line treatment was correlated with older age, higher comorbidity index, smoking history, and the lack of an identifiable driver mutation. Conclusion NGS tests results were not exclusively used to inform first line treatment decisions in most patients with stage IV NSCLC, and most patients with a targetable mutation were not treated with targeted therapy. Possible explanations include lengthy turnaround times for NGS testing and the availability of timelier but less accurate single gene testing.
Collapse
Affiliation(s)
- Ahmed I Megahed
- Kaiser Permanente Southern California, Research & Evaluation, Pasadena, CA, USA
| | - Dominick Zheng
- Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - Lie Hong Chen
- Kaiser Permanente Southern California, Research & Evaluation, Pasadena, CA, USA
| | - Reina Haque
- Kaiser Permanente Southern California, Research & Evaluation, Pasadena, CA, USA
- Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - Eric C McGary
- Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| |
Collapse
|
6
|
Werner R, Crosbie R, Dorney M, Connolly A, Collins D, Hand CK, Burke L. Implementation of an ISO 15189 accredited next generation sequencing service for cell-free total nucleic acid (cfTNA) analysis to facilitate driver mutation reporting in blood: the experience of a clinical diagnostic laboratory. J Clin Pathol 2024:jcp-2024-209514. [PMID: 38914446 DOI: 10.1136/jcp-2024-209514] [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: 03/04/2024] [Accepted: 06/11/2024] [Indexed: 06/26/2024]
Abstract
AIMS Next generation sequencing (NGS) on tumour tissue is integral to the delivery of personalised medicine and targeted therapy. NGS on liquid biopsy, a much less invasive technology, is an emerging clinical tool that has rapidly expanded clinical utility. Gene mutations in cell-free total nucleic acids (cfTNA) circulating in the blood are representative of whole tumour biology and can reveal different mutations from different tumour sites, thus addressing tumour heterogeneity challenges. METHODS The novel Ion Torrent Genexus NGS system with automated sample preparation, onboard library preparation, templating, sequencing, data analysis and Oncomine Reporter software was used. cfTNA extracted from plasma was verified with the targeted pan-cancer (~50 genes) Oncomine Precision Assay (OPA). Assessment criteria included analytical sensitivity, specificity, limits of detection (LOD), accuracy, repeatability, reproducibility and the establishment of performance metrics. RESULTS An ISO 15189 accredited, minimally invasive cfTNA NGS diagnostic service has been implemented. High sensitivity (>83%) and specificity between plasma and tissue were observed. A sequencing LOD of 1.2% was achieved when the depth of coverage was >22 000×. A reduction (>68%) in turnaround time (TAT) of liquid biopsy results was achieved: 5 days TAT for in-house analysis from sample receipt to a final report issued to oncologists as compared with >15 days from reference laboratories. CONCLUSION Tumour-derived somatic variants can now be reliably assessed from plasma to provide minimally invasive tumour profiling. Successful implementation of this accredited service resulted in:Appropriate molecular profiling of patients where tumour tissue is unavailable or inaccessible.Rapid TAT of plasma NGS results.
Collapse
Affiliation(s)
- Reiltin Werner
- Pathology Department, Cork University Hospital, Cork, Ireland
- Department of Pathology, School of Medicine, University College Cork College of Medicine and Health, Cork, Ireland
| | - Ruth Crosbie
- Pathology Department, Cork University Hospital, Cork, Ireland
| | - Mairead Dorney
- Pathology Department, Cork University Hospital, Cork, Ireland
| | - Amy Connolly
- Pathology Department, Cork University Hospital, Cork, Ireland
| | | | - Collette K Hand
- Department of Pathology, School of Medicine, University College Cork College of Medicine and Health, Cork, Ireland
| | - Louise Burke
- Pathology Department, Cork University Hospital, Cork, Ireland
- Department of Pathology, School of Medicine, University College Cork College of Medicine and Health, Cork, Ireland
| |
Collapse
|
7
|
Zhou S, Shen C, Wang Y, Zhao Z, Che G. Values of circulating tumor DNA for non-small cell lung cancer patients receiving neoadjuvant therapy, progress and challenges: a narrative review. J Thorac Dis 2024; 16:4742-4755. [PMID: 39144303 PMCID: PMC11320285 DOI: 10.21037/jtd-24-265] [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: 02/18/2024] [Accepted: 05/24/2024] [Indexed: 08/16/2024]
Abstract
Background and Objective The value of circulating tumor DNA (ctDNA) in neoadjuvant therapy (NAT) for lung cancer remains controversial. Therefore, we conducted a review to further investigate the role of ctDNA in non-small cell lung cancer (NSCLC) patients undergoing NAT for individualized management. Methods A search of online databases (PubMed, Embase, Web of Science, Science Direct, and Cochrane Library) was conducted to evaluate the value of ctDNA in predicting relapse, risk stratification, and efficacy of NAT in NSCLC. Only articles published in English within the last 25 years, between January 1st, 1998 and November 30th, 2023, were included. Additionally, the application of ctDNA in NSCLC is briefly reviewed. Key Content and Findings ctDNA is a non-invasive and dynamic method that plays an important role in future treatment guidance. Additionally, ctDNA successfully predicted the effect of neoadjuvant immunotherapy before surgery, and positive testing was strongly correlated with a lower major pathological response or complete pathological response rate. Sequential testing of ctDNA may serve as a secondary indicator to guide the adjustment of treatment programs. However, the application of this method has been limited by false negative results, a lack of objective indicators, and high costs. These issues must be addressed by researchers. Conclusions ctDNA has strong potential in NAT, based on positive preliminary studies. However, its widespread use is limited by the high cost of testing. Further research is needed to explore its value in risk stratification and treatment guidance in the future.
Collapse
Affiliation(s)
- Sicheng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Shen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Wang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Ziyi Zhao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Bredella MA, Fintelmann FJ, Iafrate AJ, Dagogo-Jack I, Dreyer KJ, Louis DN, Brink JA, Lennerz JK. Administrative Alignment for Integrated Diagnostics Leads to Shortened Time to Diagnose and Service Optimization. Radiology 2024; 312:e240335. [PMID: 39078305 PMCID: PMC11294756 DOI: 10.1148/radiol.240335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/04/2024] [Indexed: 07/31/2024]
Affiliation(s)
- Miriam A. Bredella
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - Florian J. Fintelmann
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - A. John Iafrate
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - Ibiayi Dagogo-Jack
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - Keith J. Dreyer
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - David N. Louis
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - James A. Brink
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| | - Jochen K. Lennerz
- From the Department of Radiology, NYU Langone Health Grossman School
of Medicine, 227 E 30th St, Translational Research Building 743, New York, NY
10016 (M.A.B.); Departments of Radiology (M.A.B., F.J.F., K.J.D., J.A.B.) and
Pathology (A.J.I., D.N.L., J.K.L.), Massachusetts General Hospital, Harvard
Medical School, Boston, Mass; Center for Integrated Diagnostics, Massachusetts
General Hospital, Harvard Medical School, Boston, Mass (A.J.I., J.K.L.);
Department of Thoracic Oncology, Massachusetts General Hospital Cancer Center,
Harvard Medical School, Boston, Mass (I.D.J.); Departments of Radiology (K.J.D.,
J.A.B.) and Pathology (D.N.L.), Brigham and Women’s Hospital, Harvard
Medical School, Boston, Mass; Data Science Office, Mass General Brigham Health
System, Boston, Mass (K.J.D.); and BostonGene, Waltham, Mass (J.K.L.)
| |
Collapse
|
9
|
Wiedower JA, Forbes SP, Tsai LJ, Liao J, Raez LE. Real-world clinical and economic outcomes for patients with advanced non-small cell lung cancer enrolled in a clinical trial following comprehensive genomic profiling via liquid biopsy. J Manag Care Spec Pharm 2024; 30:660-671. [PMID: 38950156 PMCID: PMC11220364 DOI: 10.18553/jmcp.2024.30.7.660] [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 Oncology clinical trial enrollment is strongly recommended for patients with cancer who are not eligible for established and approved therapies. Many trials are specific to biomarker-targeted therapies, which are typically managed as specialty pharmacy services. Comprehensive genomic profiling (CGP) of advanced cancers has been shown to detect biomarkers, guide targeted treatment, improve outcomes, and result in the clinical trial enrollment of patients, which is modeled to offset pharmacy costs experienced by US payers, yet payer policy coverage remains inconsistent. A common concern limiting coverage of CGP by payers is the potential of identifying biomarkers beyond guideline-recommended treatments, which creates a perception that insurance companies are being positioned to "pay for research." However, these biomarkers can increase clinical trial eligibility, and specialty pharmacy management may have an interest in maximizing the clinical trial enrollment of members. OBJECTIVE To investigate if clinical trial enrollment following liquid biopsy CGP for non-small cell lung cancer (NSCLC) is clinically and/or economically impactful from a payer claims perspective. METHODS Clinical and economic outcomes were studied using a real-world clinical genomic database (including payer claims data) from patients with NSCLC who enrolled in clinical trials immediately following liquid biopsy CGP (using Guardant360) and matched NSCLC patient controls also tested with liquid biopsy CGP. RESULTS Real-world overall survival was significantly (log-rank P < 0.0001) better for patients enrolled in clinical trials with similar costs of care, albeit with more outpatient encounters among those enrolled compared with matched controls. CONCLUSIONS The results, together with previous analyses, suggest that, in addition to the clinical benefits associated with targeted therapies directed by CGP and other testing approaches, payers and specialty pharmacy managers may consider clinical trial direction and enrollment as a clinical and economic benefit of liquid biopsy CGP and adopt this into coverage decision frameworks and formularies.
Collapse
Affiliation(s)
- Julie A. Wiedower
- Department of Nursing, Clemson University, SC
- Guardant Health, Redwood City, CA
| | | | | | | | - Luis E. Raez
- Thoracic Oncology Program, Memorial Cancer Institute/Memorial Healthcare System, Florida Atlantic University, Pembroke Pines
| |
Collapse
|
10
|
Imyanitov EN, Preobrazhenskaya EV, Orlov SV. Current status of molecular diagnostics for lung cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:742-765. [PMID: 38966170 PMCID: PMC11220319 DOI: 10.37349/etat.2024.00244] [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: 01/17/2024] [Accepted: 04/08/2024] [Indexed: 07/06/2024] Open
Abstract
The management of lung cancer (LC) requires the analysis of a diverse spectrum of molecular targets, including kinase activating mutations in EGFR, ERBB2 (HER2), BRAF and MET oncogenes, KRAS G12C substitutions, and ALK, ROS1, RET and NTRK1-3 gene fusions. Administration of immune checkpoint inhibitors (ICIs) is based on the immunohistochemical (IHC) analysis of PD-L1 expression and determination of tumor mutation burden (TMB). Clinical characteristics of the patients, particularly age, gender and smoking history, significantly influence the probability of finding the above targets: for example, LC in young patients is characterized by high frequency of kinase gene rearrangements, while heavy smokers often have KRAS G12C mutations and/or high TMB. Proper selection of first-line therapy influences overall treatment outcomes, therefore, the majority of these tests need to be completed within no more than 10 working days. Activating events in MAPK signaling pathway are mutually exclusive, hence, fast single-gene testing remains an option for some laboratories. RNA next-generation sequencing (NGS) is capable of detecting the entire repertoire of druggable gene alterations, therefore it is gradually becoming a dominating technology in LC molecular diagnosis.
Collapse
Affiliation(s)
- Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Clinical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
- I.V. Kurchatov Complex for Medical Primatology, National Research Centre “Kurchatov Institute”, 354376 Sochi, Russia
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia
- Department of Clinical Genetics, St.-Petersburg State Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Sergey V. Orlov
- I.V. Kurchatov Complex for Medical Primatology, National Research Centre “Kurchatov Institute”, 354376 Sochi, Russia
- Department of Oncology, I.P. Pavlov St.-Petersburg State Medical University, 197022 St.-Petersburg, Russia
| |
Collapse
|
11
|
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
|
12
|
Yorio J, Lofgren KT, Lee JK, Tolba K, Oxnard GR, Schrock AB, Huang RS, Brisbin L. Association of Timely Comprehensive Genomic Profiling With Precision Oncology Treatment Use and Patient Outcomes in Advanced Non-Small-Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2300292. [PMID: 38452312 PMCID: PMC10939592 DOI: 10.1200/po.23.00292] [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: 06/09/2023] [Revised: 12/11/2023] [Accepted: 01/04/2024] [Indexed: 03/09/2024] Open
Abstract
PURPOSE Timely biomarker testing remains out of reach for many patients with advanced non-small-cell lung cancer (aNSCLC). Here, we studied the quality-of-care implications of closing the gap in timely receipt of comprehensive genomic profiling (CGP) to inform first-line (1L) decisions. METHODS Using a real-world clinicogenomic database, we studied testing and 1L treatment patterns in aNSCLC after the approval of pembrolizumab in combination with pemetrexed and carboplatin (May 10, 2017). To estimate the association of timely CGP results with therapy selection and patient outcomes, we identified patients with no previous genomic testing beyond PD-L1 immunohistochemistry and dichotomized patients by whether CGP results were available before or after 1L therapy initiation. RESULTS In total, 2,694 patients were included in the 1L therapy decision impact assessment. Timely CGP increased matched targeted therapy use by 14 percentage points (17% with CGP v 2.8% without) and precision immune checkpoint inhibitor (ICPI) use by 14 percentage points (18% with CGP v 3.9% without). Receipt of timely CGP resulted in an estimated 31 percentage point decrease in ICPI use among ALK/EGFR/RET/ROS1-positive patients at an expected per-patient reduction in ineffective ICPI therapy cost of $13,659.37 with timely CGP to inform 1L treatment selection. Patient benefit of CGP extended to real-world time to therapy discontinuation (median time to therapy discontinuation: 3.9 v 10 months [hazard ratio, HR, 0.54 [95% CI, 0.42 to 0.70]; P = 1.9E-06; adjusted hazard ratio [aHR], 0.50 [95% CI, 0.38 to 0.67]; P = 2.0E-06) in 1L driver-positive patients. This effect was not significant for real-world overall survival (median overall survival: 32 v 29 months [HR, 1.2 [95% CI, 0.84 to 1.67]; P = .33; aHR, 1.4 [95% CI, 0.92 to 1.99]; P = .12). CONCLUSION Timely CGP is associated with the quality of patient care as measured by 1L matched targeted therapy use, time to therapy discontinuation, and avoidance of ineffective, costly ICPIs.
Collapse
|
13
|
Bestvina CM, Waters D, Morrison L, Emond B, Lafeuille MH, Hilts A, Lefebvre P, He A, Vanderpoel J. Cost of genetic testing, delayed care, and suboptimal treatment associated with polymerase chain reaction versus next-generation sequencing biomarker testing for genomic alterations in metastatic non-small cell lung cancer. J Med Econ 2024; 27:292-303. [PMID: 38391239 DOI: 10.1080/13696998.2024.2314430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
AIMS To assess US payers' per-patient cost of testing associated with next-generation sequencing (NGS) versus polymerase chain reaction (PCR) biomarker testing strategies among patients with metastatic non-small cell lung cancer (mNSCLC), including costs of testing, delayed care, and suboptimal treatment initiation. METHODS A decision tree model considered biomarker testing for genomic alterations using either NGS, sequential PCR testing, or hotspot panel PCR testing. Literature-based model inputs included time-to-test results, costs for testing/medical care, costs of delaying care, costs of immunotherapy [IO]/chemotherapy [CTX] initiation prior to receiving test results, and costs of suboptimal treatment initiation after test results (i.e. costs of first-line IO/CTX in patients with actionable mutations that were undetected by PCR that would have been identified with NGS). The proportion of patients testing positive for a targetable alteration, time to appropriate therapy initiation, and per-patient costs were estimated for NGS and PCR strategies combined. RESULTS In a modeled cohort of 1,000,000 members (25% Medicare, 75% commercial), an estimated 1,119 had mNSCLC and received testing. The proportion of patients testing positive for a targetable alteration was 45.9% for NGS and 40.0% for PCR testing. Mean per-patient costs were lowest for NGS ($8,866) compared to PCR ($18,246), with lower delayed care costs of $1,301 for NGS compared to $3,228 for PCR, and lower costs of IO/CTX initiation prior to receiving test results (NGS: $2,298; PCR:$5,991). Cost savings, reaching $10,496,220 at the 1,000,000-member plan level, were driven by more rapid treatment with appropriate therapy for patients tested with NGS (2.1 weeks) compared to PCR strategies (5.2 weeks). LIMITATIONS Model inputs/assumptions were based on published literature or expert opinion. CONCLUSIONS NGS testing was associated with greater cost savings versus PCR, driven by more rapid results, shorter time to appropriate therapy initiation, and minimized use of inappropriate therapies while awaiting and after test results.
Collapse
Affiliation(s)
- Christine M Bestvina
- University of Chicago Comprehensive Cancer Center; Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Dexter Waters
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
| | | | | | | | | | | | - Andy He
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
| | - Julie Vanderpoel
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
| |
Collapse
|
14
|
Kiełbowski K, Żychowska J, Becht R. Anaplastic lymphoma kinase inhibitors-a review of anticancer properties, clinical efficacy, and resistance mechanisms. Front Pharmacol 2023; 14:1285374. [PMID: 37954850 PMCID: PMC10634320 DOI: 10.3389/fphar.2023.1285374] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Fusions and mutations of anaplastic lymphoma kinase (ALK), a tyrosine kinase receptor, have been identified in several neoplastic diseases. Rearranged ALK is a driver of tumorigenesis, which activates various signaling pathway associated with proliferation and survival. To date, several agents that target and inhibit ALK have been developed. The most studied ALK-positive disease is non-small cell lung cancer, and three generations of ALK tyrosine kinase inhibitors (TKIs) have been approved for the treatment of metastatic disease. Nevertheless, the use of ALK-TKIs is associated with acquired resistance (resistance mutations, bypass signaling), which leads to disease progression and may require a substitution or introduction of other treatment agents. Understanding of the complex nature and network of resistance mutations may allow to introduce sequential and targeted therapies. In this review, we aim to summarize the efficacy and safety profile of ALK inhibitors, describe off-target anticancer effects, and discuss resistance mechanisms in the context of personalized oncology.
Collapse
Affiliation(s)
| | | | - Rafał Becht
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
15
|
Hess LM, Peterson P, Sugihara T, Bhandari NR, Krein PM, Sireci A. Initial versus early switch to targeted therapy during first-line treatment among patients with biomarker-positive advanced or metastatic non-small cell lung cancer in the United States. Cancer Treat Res Commun 2023; 37:100761. [PMID: 37717466 DOI: 10.1016/j.ctarc.2023.100761] [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: 07/13/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVES This study compared outcomes between patients with biomarker-positive advanced/metastatic non-small cell lung cancer (a/mNSCLC) who initiated treatment with targeted therapy versus those who initiated chemotherapy-based treatment and switched to targeted therapy during the first ∼3 cycles (defined as the first 56 days) of first-line treatment. MATERIALS AND METHODS This was an observational study of patients with a/mNSCLC who received targeted therapy from a nationwide electronic health record (EHR)-derived de-identified database. Outcomes were compared between those who initiated targeted therapy versus those who switched from chemotherapy to a targeted agent. Time-to-event outcomes were evaluated using Kaplan-Meier method; Cox proportional hazards models (adjusted for baseline covariates) were used to compare outcomes between groups. RESULTS Of the 4,244 patients in this study, 3,107 (73.2%) initiated the first line with targeted therapy and 346 (8.2%) switched to targeted therapy. Patients who received initial targeted therapy were significantly more likely to be non-smokers, treated in an academic practice setting, and of slightly older age (all p < 0.05). Patients who received initial targeted therapy also had a significantly longer time to start of first-line treatment (35.8 vs 25.3 days, p < 0.001). No significant differences were observed for clinical outcomes between groups. CONCLUSION In both unadjusted and adjusted analyses, there were no differences in the clinical outcomes observed among patients with a/mNSCLC in this study. This study found that initiating chemotherapy with an early switch to targeted therapy (within 56 days) of receiving biomarker positive results may be an acceptable strategy for a patient for whom immediate care is needed.
Collapse
|
16
|
Normanno N, Machado JC, Pescarmona E, Buglioni S, Navarro L, Esposito Abate R, Ferro A, Mensink R, Lambiase M, Lespinet-Fabre V, Calgua B, Jermann PM, Ilié M, Hofman P. European Real-World Assessment of the Clinical Validity of a CE-IVD Panel for Ultra-Fast Next-Generation Sequencing in Solid Tumors. Int J Mol Sci 2023; 24:13788. [PMID: 37762091 PMCID: PMC10531166 DOI: 10.3390/ijms241813788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Molecular profiling of solid tumors facilitates personalized, targeted therapeutic interventions. The ability to perform next-generation sequencing (NGS), especially from small tissue samples, in a short turnaround time (TAT) is essential to providing results that enable rapid clinical decisions. This multicenter study evaluated the performance of a CE in vitro diagnostic (IVD) assay, the Oncomine Dx Express Test, on the Ion Torrent Genexus System for detecting DNA and RNA variants in solid tumors. Eighty-two archived formalin-fixed paraffin embedded (FFPE) tissue samples from lung, colorectal, central nervous system, melanoma, breast, gastric, thyroid, and soft tissue cancers were used to assess the presence of single nucleotide variants (SNVs), insertions and deletions (indels), copy number variations (CNVs), gene fusions, and splice variants. These clinical samples were previously characterized at the various academic centers using orthogonal methods. The Oncomine Dx Express Test showed high performance with 100% concordance with previous characterization for SNVs, indels, CNVs, gene fusions, and splice variants. SNVs and indels with allele frequencies as low as 5% were correctly identified. The test detected all the expected ALK, RET, NTRK1, and ROS1 fusion isoforms and MET exon 14-skipping splice variants. The average TAT from extracted nucleic acids to the final variant report was 18.3 h. The Oncomine Dx Express Test in combination with the Ion Torrent Genexus System is a CE-IVD-compliant, performant, and multicenter reproducible method for NGS detection of actionable biomarkers from a range of tumor samples, providing results in a short TAT that could support timely decision- making for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - José Carlos Machado
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Edoardo Pescarmona
- I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy; (E.P.); (S.B.)
| | - Simonetta Buglioni
- I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy; (E.P.); (S.B.)
| | - Lara Navarro
- Consorcio Hospital General de Valencia, 46014 Valencia, Spain;
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - Anabela Ferro
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Rob Mensink
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Matilde Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| | - Byron Calgua
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (B.C.); (P.M.J.)
| | - Philip M. Jermann
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (B.C.); (P.M.J.)
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| |
Collapse
|