771 results on '"David R. Gandara"'
Search Results
52. Supplementary Figure from Circulating Tumor DNA Kinetics Predict Progression-Free and Overall Survival in EGFR TKI–Treated Patients with EGFR-Mutant NSCLC (SWOG S1403)
- Author
-
David R. Gandara, Karen Kelly, Katerina Politi, Fred R. Hirsch, Zenta Walther, Mary Ann Melnick, Roy S. Herbst, Sarah B. Goldberg, James Moon, Mary W. Redman, Jieling Miao, and Philip C. Mack
- Abstract
Supplementary Figure from Circulating Tumor DNA Kinetics Predict Progression-Free and Overall Survival in EGFR TKI–Treated Patients with EGFR-Mutant NSCLC (SWOG S1403)
- Published
- 2023
- Full Text
- View/download PDF
53. Data from Design of a Phase III Clinical Trial with Prospective Biomarker Validation: SWOG S0819
- Author
-
David R. Gandara, Fred R. Hirsch, Roy S. Herbst, John J. Crowley, and Mary W. Redman
- Abstract
The role of cetuximab in the treatment of advanced non–small cell lung cancer (NSCLC) is currently unclear. The molecular target of cetuximab, epidermal growth factor receptor (EGFR), as measured by FISH, has shown potential as a predictive biomarker for cetuximab efficacy in NSCLC. SWOG S0819 is a phase III trial evaluating both the value of cetuximab in this setting and EGFR FISH as a predictive biomarker. This work describes the decision process for determining the design and interim monitoring plan for S0819. Six possible designs were evaluated in terms of their properties and the hypotheses that can be addressed within the design constraints. A subgroup-focused, multiple-hypothesis design was selected for S0819 that incorporates coprimary endpoints to assess cetuximab in both the overall study population and among EGFR FISH-positive (FISH+) patients, with the sample size determined based on evaluation in the EGFR FISH+ group. The chosen interim monitoring plan specifies interim evaluations of both efficacy and futility in the EGFR FISH+ group alone. The futility-monitoring plan to determine early stopping in the EGFR FISH-nonpositive group is based on evaluation within the positive group, the entire study population, and the nonpositive group. SWOG S0819 uses a design that addresses both the biomarker-driven and general-efficacy objectives of this study. Clin Cancer Res; 18(15); 4004–12. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
54. Supplementary Table 3 from Investigation of Metabolomic Blood Biomarkers for Detection of Adenocarcinoma Lung Cancer
- Author
-
Suzanne Miyamoto, Karen Kelly, Oliver Fiehn, David T. Cooke, Ken Y. Yoneda, David R. Gandara, Sandra L. Taylor, Brian C. DeFelice, Kyoungmi Kim, and Johannes F. Fahrmann
- Abstract
Excel - 12 MB, Supplemental Table S3. Pearson correlations between circulating metabolites in serum and plasma during ADC1 and ADC2 (Excel Spreadsheet, four tabs in the following order: ADC1 plasma, ADC1 serum, ADC2 plasma, and ADC2 serum).
- Published
- 2023
- Full Text
- View/download PDF
55. Supplementary Table 1 from Aflibercept (VEGF Trap) in Inoperable Stage III or Stage IV Melanoma of Cutaneous or Uveal Origin
- Author
-
John M. Kirkwood, Alice Chen, David R. Gandara, Janice Shipe-Spotloe, Christopher Ruel, Scott Christensen, Kim A. Margolin, Paul Frankel, and Ahmad A. Tarhini
- Abstract
PDF file - 16K
- Published
- 2023
- Full Text
- View/download PDF
56. Data from The Landscape of Actionable Genomic Alterations in Cell-Free Circulating Tumor DNA from 21,807 Advanced Cancer Patients
- Author
-
AmirAli Talasaz, Richard B. Lanman, Darya I. Chudova, Helmy Eltoukhy, Arthur M. Baca, Rebecca J. Nagy, Justin I. Odegaard, Philip C. Mack, David R. Gandara, Reza Mokhtari, James V. Vowles, Stefanie A. Mortimer, Stephen R. Fairclough, Kimberly C. Banks, and Oliver A. Zill
- Abstract
Purpose: Cell-free DNA (cfDNA) sequencing provides a noninvasive method for obtaining actionable genomic information to guide personalized cancer treatment, but the presence of multiple alterations in circulation related to treatment and tumor heterogeneity complicate the interpretation of the observed variants.Experimental Design: We describe the somatic mutation landscape of 70 cancer genes from cfDNA deep-sequencing analysis of 21,807 patients with treated, late-stage cancers across >50 cancer types. To facilitate interpretation of the genomic complexity of circulating tumor DNA in advanced, treated cancer patients, we developed methods to identify cfDNA copy-number driver alterations and cfDNA clonality.Results: Patterns and prevalence of cfDNA alterations in major driver genes for non–small cell lung, breast, and colorectal cancer largely recapitulated those from tumor tissue sequencing compendia (The Cancer Genome Atlas and COSMIC; r = 0.90–0.99), with the principal differences in alteration prevalence being due to patient treatment. This highly sensitive cfDNA sequencing assay revealed numerous subclonal tumor-derived alterations, expected as a result of clonal evolution, but leading to an apparent departure from mutual exclusivity in treatment-naïve tumors. Upon applying novel cfDNA clonality and copy-number driver identification methods, robust mutual exclusivity was observed among predicted truncal driver cfDNA alterations (FDR = 5 × 10−7 for EGFR and ERBB2), in effect distinguishing tumor-initiating alterations from secondary alterations. Treatment-associated resistance, including both novel alterations and parallel evolution, was common in the cfDNA cohort and was enriched in patients with targetable driver alterations (>18.6% patients).Conclusions: Together, these retrospective analyses of a large cfDNA sequencing data set reveal subclonal structures and emerging resistance in advanced solid tumors. Clin Cancer Res; 24(15); 3528–38. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
57. Table S7-12 from The Landscape of Actionable Genomic Alterations in Cell-Free Circulating Tumor DNA from 21,807 Advanced Cancer Patients
- Author
-
AmirAli Talasaz, Richard B. Lanman, Darya I. Chudova, Helmy Eltoukhy, Arthur M. Baca, Rebecca J. Nagy, Justin I. Odegaard, Philip C. Mack, David R. Gandara, Reza Mokhtari, James V. Vowles, Stefanie A. Mortimer, Stephen R. Fairclough, Kimberly C. Banks, and Oliver A. Zill
- Abstract
Mutual exclusivity statistics for cfDNA alterations in lung adenocarcinoma, breast cancer, and colorectal cancer
- Published
- 2023
- Full Text
- View/download PDF
58. Supplementary Tables S1-S3 from Phase I Trial of Arginine Deprivation Therapy with ADI-PEG 20 Plus Docetaxel in Patients with Advanced Malignant Solid Tumors
- Author
-
Primo N. Lara, Chong-Xian Pan, David R. Gandara, Thomas Semrad, I-Yeh Gong, Karen Kelly, Mrinal P. Dutia, Tianhong Li, Nichole Mahaffey, Taiwo Akande, Monica Diaz, John S. Bomalaski, James A. Thomson, and Benjamin K. Tomlinson
- Abstract
Supplementary Tables S1-S3. Table S1: The complete dose escalation schema for ADI-PEG 20 in combination with docetaxel. Table S2: Details the completed pharmacodynamic data for individual patients. Includes concentrations of arginine and citrulline over time and the titers of antibodies to ADI-PEG 20. Table S3: Highlights response against tumor type in patients where response was able to be assessed.
- Published
- 2023
- Full Text
- View/download PDF
59. Data from Circulating Tumor DNA Kinetics Predict Progression-Free and Overall Survival in EGFR TKI–Treated Patients with EGFR-Mutant NSCLC (SWOG S1403)
- Author
-
David R. Gandara, Karen Kelly, Katerina Politi, Fred R. Hirsch, Zenta Walther, Mary Ann Melnick, Roy S. Herbst, Sarah B. Goldberg, James Moon, Mary W. Redman, Jieling Miao, and Philip C. Mack
- Abstract
Purpose:Dynamic changes in circulating tumor DNA (ctDNA) are under investigation as an early indicator of treatment outcome.Experimental Design:Serial plasma ctDNA (baseline, 8 weeks, and at progression) was prospectively incorporated into the SWOG S1403 clinical trial of afatinib ± cetuximab in tyrosine kinase inhibitor—naïve, EGFR mutation tissue–positive non–small cell lung cancer.Results:EGFR mutations were detected in baseline ctDNA in 77% (82/106) of patients, associated with the presence of brain and/or liver metastases and M1B stage. Complete clearance of EGFR mutations in ctDNA by 8 weeks was associated with a significantly decreased risk of progression, compared with those with persistent ctDNA at Cycle 3 Day 1 [HR, 0.23; 95% confidence interval (CI), 0.12–0.45; P < 0.0001], with a median progression-free survival (PFS) of 15.1 (95% CI, 10.6–17.5) months in the group with clearance of ctDNA versus 4.6 (1.7–7.5) months in the group with persistent ctDNA. Clearance was also associated with a decreased risk of death (HR, 0.44; 95% CI, 0.21–0.90), P = 0.02; median overall survival (OS): 32.6 (23.5–not estimable) versus 15.6 (4.9–28.3) months.Conclusions:Plasma clearance of mutant EGFR ctDNA at 8 weeks was highly and significantly predictive of PFS and OS, outperforming RECIST response for predicting long-term benefit.
- Published
- 2023
- Full Text
- View/download PDF
60. Supplement 3 from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Author
-
Hsing-Jien Kung, Philip C. Mack, David R. Gandara, Primo N. Lara, Pierre Y. Desprez, Tim P. Green, Christopher P. Evans, Yoshihiro Izumiya, Phillip R. Purnell, Clifford G. Tepper, and Oliver Gautschi
- Abstract
Supplement 3 from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Published
- 2023
- Full Text
- View/download PDF
61. Data from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Author
-
Hsing-Jien Kung, Philip C. Mack, David R. Gandara, Primo N. Lara, Pierre Y. Desprez, Tim P. Green, Christopher P. Evans, Yoshihiro Izumiya, Phillip R. Purnell, Clifford G. Tepper, and Oliver Gautschi
- Abstract
Deregulated activation of the Src tyrosine kinase and heightened Id1 expression are independent mediators of aggressive tumor biology. The present report implicates Src signaling as a critical regulator of Id1 gene expression. Microarray analyses showed that Id family genes were among the most highly down-regulated by incubation of A549 lung carcinoma cells with the small-molecule Src inhibitor AZD0530. Id1 transcript and protein levels were potently reduced in a dose-dependent manner concomitantly with the reduction of activated Src levels. These effects were conserved across a panel of lung, breast, prostate, and colon cancer cell lines and confirmed by the ability of PP2, Src siRNA, and Src-blocking peptides to suppress Id1 expression. PP2, AZD0530, and dominant-negative Src abrogated Id1 promoter activity, which was induced by constitutively active Src. The Src-responsive region of the Id1 promoter was mapped to a region 1,199 to 1,360 bps upstream of the translation start site and contained a Smad-binding element. Src was also required for bone morphogenetic protein-2 (BMP-2)–induced Id1 expression and promoter activity, was moderately activated by BMP-2, and complexed with Smad1/5. Conversely, Src inhibitors blocked Smad1/5 nuclear translocation and binding to the Src-responsive region of the Id1 promoter. Consistent with a role for Src and Id1 in cancer cell invasion, Src inhibitors and Id1 siRNA decreased cancer cell invasion, which was increased by Id1 overexpression. Taken together, these results reveal that Src positively interacts with the BMP-Smad-Id pathway and provide new ways for targeted inhibition of Id1. [Cancer Res 2008;68(7):2250–8]
- Published
- 2023
- Full Text
- View/download PDF
62. Supplement 2 from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Author
-
Hsing-Jien Kung, Philip C. Mack, David R. Gandara, Primo N. Lara, Pierre Y. Desprez, Tim P. Green, Christopher P. Evans, Yoshihiro Izumiya, Phillip R. Purnell, Clifford G. Tepper, and Oliver Gautschi
- Abstract
Supplement 2 from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Published
- 2023
- Full Text
- View/download PDF
63. Supplement 1 from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Author
-
Hsing-Jien Kung, Philip C. Mack, David R. Gandara, Primo N. Lara, Pierre Y. Desprez, Tim P. Green, Christopher P. Evans, Yoshihiro Izumiya, Phillip R. Purnell, Clifford G. Tepper, and Oliver Gautschi
- Abstract
Supplement 1 from Regulation of Id1 Expression by Src: Implications for Targeting of the Bone Morphogenetic Protein Pathway in Cancer
- Published
- 2023
- Full Text
- View/download PDF
64. Comparison of SP142 and 22C3 Immunohistochemistry PD-L1 Assays for Clinical Efficacy of Atezolizumab in Non–Small Cell Lung Cancer: Results From the Randomized OAK Trial
- Author
-
Fred R. Hirsch, Sarah M. Paul, Keunchil Park, C. Matheny, Achim Rittmeyer, Jing Yi, Wei Zou, Marcus Ballinger, Mark McCleland, Fabrice Barlesi, Keith M. Kerr, Shirish M. Gadgeel, Hartmut Koeppen, David R. Gandara, David S. Shames, and Namrata Bhatia
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Population ,Antineoplastic Agents ,Subgroup analysis ,Docetaxel ,Antibodies, Monoclonal, Humanized ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,Progression-free survival ,education ,Lung cancer ,Immune Checkpoint Inhibitors ,Retrospective Studies ,education.field_of_study ,business.industry ,Hazard ratio ,medicine.disease ,Immunohistochemistry ,Confidence interval ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
BACKGROUND This phase III OAK trial (NCT02008227) subgroup analysis (data cutoff, January 9, 2019) evaluated the predictive value of 2 PD-L1 IHC tests (VENTANA SP142 and Dako 22C3) for benefit from atezolizumab versus docetaxel by programmed death ligand 1 (PD-L1) status in patients with previously treated metastatic non-small cell lung cancer. METHODS PD-L1 expression was assessed prospectively with SP142 on tumor cells (TC) and tumor-infiltrating immune cells (IC) and retrospectively with 22C3 using a tumor proportion score (TPS) based on TC membrane staining. Efficacy was assessed in the 22C3 biomarker-evaluable population (22C3-BEP) (n = 577; 47.1% of SP142-intention-to-treat population) and non-22C3-BEP (n = 648) in PD-L1 subgroups (high, low, and negative) and according to selection by 1 or both assays. RESULTS In the 22C3-BEP, overall survival benefits with atezolizumab versus docetaxel were observed across PD-L1 subgroups; benefits were greatest in SP142-defined PD-L1-high (TC3 or IC3: hazard ratio [HR], 0.39 [95% confidence interval (CI), 0.25-0.63]) and 22C3-defined PD-L1-high (TPS ≥ 50%: HR, 0.56 [95% CI, 0.38-0.82]) and low (TPS, 1% to < 50%: HR, 0.55 [95% CI, 0.37-0.82]) groups. Progression-free survival improved with increasing PD-L1 expression for both assays. SP142 and 22C3 assays identified overlapping and unique patient populations in PD-L1-high, positive, and negative subgroups. Overall survival and progression-free survival benefits favored atezolizumab over docetaxel in double PD-L1-positive and negative groups; patients with both SP142- and 22C3-positive tumors derived the greatest benefit. CONCLUSIONS Despite different scoring algorithms and differing sensitivity levels, the SP142 and 22C3 assays similarly predicted atezolizumab benefit at validated PD-L1 thresholds in patients with non-small cell lung cancer.
- Published
- 2022
- Full Text
- View/download PDF
65. Convergence of Precision Oncology and Liquid Biopsy in Non-Small Cell Lung Cancer
- Author
-
Ebaa Al-Obeidi, Jonathan W. Riess, Umberto Malapelle, Christian Rolfo, David R. Gandara, Al-Obeidi, Ebaa, Riess, Jonathan W, Malapelle, Umberto, Rolfo, Christian, and Gandara, David R
- Subjects
Tumor mutational burden (TMB) ,Oncology ,Liquid biopsy ,Next-generation sequencing (NGS) ,Non-small cell lung cancer (NSCLC) ,Hematology ,Minimal residual disease (MRD) ,Circulating tumor DNA (ctDNA) - Abstract
This review article illuminates the role of liquid biopsy in the continuum of care for non-small cell lung cancer (NSCLC). We discuss its current application in advanced-stage NSCLC at the time of diagnosis and at progression. We highlight research showing that concurrent testing of blood and tissue yields faster, more informative, and cheaper answers than the standard stepwise approach. We also describe future applications for liquid biopsy including treatment response monitoring and testing for minimal residual disease. Lastly, we discuss the emerging role of liquid biopsy for screening and early detection.
- Published
- 2023
66. Patient Knowledge and Expectations About Return of Genomic Results in a Biomarker-Driven Master Protocol Trial (SWOG S1400GEN)
- Author
-
Karen Kelly, Kate Watabayashi, Vassiliki A. Papadimitrakopoulou, Monica Yee, Meghna S. Trivedi, Dawn L. Hershman, Scott D. Ramsey, David R. Gandara, Judy Johnson, Roy S. Herbst, Deborah M Delaney, Parth D. Shah, Donald L. Patrick, Paul Litwin, Joshua A. Roth, Katherine D. Crew, Joseph M. Unger, Mary W. Redman, and Stacy W. Gray
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Sociodemographic Factors ,MEDLINE ,Pilot Projects ,ORIGINAL CONTRIBUTIONS ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Protocol (science) ,Motivation ,Oncology (nursing) ,business.industry ,Health Policy ,Genomics ,Clinical trial ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,business ,Biomarkers - Abstract
PURPOSE: Biomarker-driven master protocols represent a new paradigm in oncology clinical trials, but their complex designs and wide-ranging genomic results returned can be difficult to communicate to participants. The objective of this pilot study was to evaluate patient knowledge and expectations related to return of genomic results in the Lung Cancer Master Protocol (Lung-MAP). METHODS: Eligible participants with previously treated advanced non–small-cell lung cancer were recruited from patients enrolled in Lung-MAP. Participants completed a 38-item telephone survey ≤ 30 days from Lung-MAP consent. The survey assessed understanding about the benefits and risks of Lung-MAP participation and knowledge of the potential uses of somatic testing results returned. Descriptive statistics and odds ratios for associations between demographic factors and correct responses to survey items were assessed. RESULTS: From August 1, 2017, to June 30, 2019, we recruited 207 participants with a median age of 67, 57.3% male, and 94.2% White. Most participants “strongly/somewhat agreed” with statements that they “received enough information to understand” Lung-MAP benefits (82.6%) and risks (69.5%). In items asking about potential uses of Lung-MAP genomic results, 87.0% correctly indicated that the results help to select cancer treatment, but < 20% correctly indicated that the results are not used to confirm cancer diagnosis, would not reveal risk of developing diseases besides cancer, and would not indicate if family members had increased cancer risk. There were no associations between sociodemographic factors and proportions providing correct responses. CONCLUSION: In a large National Clinical Trials Network biomarker-driven master protocol, most participants demonstrated incorrect knowledge and expectations about the uses of genomic results provided in the study despite most indicating that they had enough information to understand benefits and risks.
- Published
- 2021
- Full Text
- View/download PDF
67. Liquid biopsy from research to clinical practice: focus on non-small cell lung cancer
- Author
-
Christian Rolfo, Edward M. Pickering, Valeria Denninghoff, Umberto Malapelle, Andrés F. Cardona, Alfredo Addeo, Kenneth J. O'Byrne, Pier Vitale Nuzzo, Diego de Miguel-Pérez, Alessandro Russo, Ignacio Duran, David R. Gandara, María José Serrano, Eloisa Jantus-Lewintre, Giancarlo Troncone, Luis E. Raez, Massimo Cristofanilli, Pasquale Pisapia, Oscar Arrieta, Beatriz Bellosillo, Patrick Pauwels, Malapelle, Umberto, Pisapia, Pasquale, Addeo, Alfredo, Arrieta, Oscar, Bellosillo, Beatriz, Cardona, Andres F, Cristofanilli, Massimo, de Miguel-Perez, Diego, Denninghoff, Valeria, Durán, Ignacio, Jantus-Lewintre, Eloísa, Nuzzo, Pier Vitale, O'Byrne, Ken, Pauwels, Patrick, Pickering, Edward M, Raez, Luis E, Russo, Alessandro, Serrano, Maria José, Gandara, David R, Troncone, Giancarlo, and Rolfo, Christian
- Subjects
medicine.medical_specialty ,Lung Neoplasms ,Context (language use) ,NSCLC ,Pathology and Forensic Medicine ,Carcinoma, Non-Small-Cell Lung ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,In patient ,Precision Medicine ,Liquid biopsy ,Intensive care medicine ,Lung cancer ,Molecular Biology ,Predictive biomarker ,liquid biopsy ,business.industry ,biomarkers ,ctDNA ,medicine.disease ,CTC ,Clinical Practice ,biomarker ,Molecular Medicine ,Personalized medicine ,Non small cell ,business - Abstract
INTRODUCTION: In the current era of personalized medicine, liquid biopsy has acquired a relevant importance in patient management of advanced stage non-small cell lung cancer (NSCLC). As a matter of fact, liquid biopsy may supplant the problem of inadequate tissue for molecular testing. The term 'liquid biopsy' refers to a number of different biological fluids, but is most clearly associated with plasma-related platforms. It must be taken into account that pre-analytical processing and the selection of the appropriate technology according to the clinical context may condition the results obtained. In addition, novel clinical applications beyond the evaluation of the molecular status of predictive biomarkers are currently under investigation. AREAS COVERED: This review summarizes the available evidence on pre-analytical issues and different clinical applications of liquid biopsies in NSCLC patients. EXPERT OPINION: Liquid biopsy should be considered not only as a valid alternative but as complementary to tissue-based molecular approaches. Careful attention should be paid to the optimization and standardization of all phases of liquid biopsy samples management in order to determine a significant improvement in either sensitivity or specificity, while significant reducing the number of 'false negative' or 'false positive' molecular results.
- Published
- 2021
- Full Text
- View/download PDF
68. NTRK1 Fusions identified by non-invasive plasma next-generation sequencing (NGS) across 9 cancer types
- Author
-
David R. Gandara, Thomas E. Stinchcombe, Caroline E. McCoach, Alison M. Schram, Emilio Paul Araujo-Mino, Timmy Nguyen, Jorge Nieva, Aditya Bardia, Lesli A. Kiedrowski, Alessandro Russo, Janakiraman Subramanian, David S. Hong, Christian Rolfo, Kristin Price, Alexander Drilon, Afshin Dowlati, Jessica J. Lin, Stephen V. Liu, Michael A. Morse, Deepa Sashital, Mohammad Mobayed, Neelima Vidula, Shahrooz Eshaghian, Scott N. Gettinger, and Sarah B. Goldberg
- Subjects
Cancer Research ,Indazoles ,Oncogene Proteins, Fusion ,Concordance ,Entrectinib ,medicine.disease_cause ,Article ,DNA sequencing ,Circulating Tumor DNA ,Neoplasms ,Biomarkers, Tumor ,medicine ,Humans ,Receptor, trkA ,Protein Kinase Inhibitors ,Genotyping ,Gene ,Neoplasm Staging ,business.industry ,Non invasive ,High-Throughput Nucleotide Sequencing ,Cancer ,medicine.disease ,Pyrimidines ,Oncology ,Benzamides ,Cancer research ,Pyrazoles ,business ,Carcinogenesis - Abstract
Background Activating fusions of the NTRK1, NTRK2 and NTRK3 genes are drivers of carcinogenesis and proliferation across a broad range of tumour types in both adult and paediatric patients. Recently, the FDA granted tumour-agnostic approvals of TRK inhibitors, larotrectinib and entrectinib, based on significant and durable responses in multiple primary tumour types. Unfortunately, testing rates in clinical practice remain quite low. Adding plasma next-generation sequencing of circulating tumour DNA (ctDNA) to tissue-based testing increases the detection rate of oncogenic drivers and demonstrates high concordance with tissue genotyping. However, the clinical potential of ctDNA analysis to identify NTRK fusion-positive tumours has been largely unexplored. Methods We retrospectively reviewed a ctDNA database in advanced stage solid tumours for NTRK1 fusions. Results NTRK1 fusion events, with nine unique fusion partners, were identified in 37 patients. Of the cases for which clinical data were available, 44% had tissue testing for NTRK1 fusions; the NTRK1 fusion detected by ctDNA was confirmed in tissue in 88% of cases. Here, we report for the first time that minimally-invasive plasma NGS can detect NTRK fusions with a high positive predictive value. Conclusion Plasma ctDNA represents a rapid, non-invasive screening method for this rare genomic target that may improve identification of patients who can benefit from TRK-targeted therapy and potentially identify subsequent on- and off-target resistance mechanisms.
- Published
- 2021
- Full Text
- View/download PDF
69. 27-gene Immuno-Oncology (IO) Score is Associated With Efficacy of Checkpoint Immunotherapy in Advanced NSCLC: A Retrospective BC Cancer Study
- Author
-
David L. Saltman, Matthew G. Varga, Tyler J. Nielsen, Nicole S. Croteau, Heather M. Lockyer, Amit L. Jain, Gregory A. Vidal, David R. Hout, Brock L. Schweitzer, Robert S. Seitz, Douglas T. Ross, and David R. Gandara
- Subjects
Pulmonary and Respiratory Medicine ,Cancer Research ,Oncology - Abstract
Immune checkpoint inhibitors (ICI) are standard of care in advanced non-small cell lung cancer (NSCLC). However, not all patients benefit, even among PD-L1 tumor proportional score (TPS) ≥50%, indicating an unmet need for additional biomarkers such as those assessing the tumor immune microenvironment (TIME). DetermaIO is a 27-gene assay that classifies TIME and has previously demonstrated association with ICI response.FFPE samples were selected from BC Cancer and West Clinic Cancer Center patients with performance status (PS) ≤2 who received at least 2 cycles of ICI monotherapy in the first (1L) or second line (2L). IO scores were generated and analyzed for association with PFS and OS.In the entire cohort (N=147), IO score was significantly associated with OS (HR=0.68, 95%CI 0.47-0.99, P = .042) and PFS (HR=0.62, 95%CI 0.43-0.88, P = .0069). In 1L treated patients (PD-L1≥50%, N=78), IO score was significantly associated with PFS (HR=0.55, 95%CI 0.32-0.94, P = .028). In exploratory analyses, IO score was associated with benefit in 1L PS2 patients for OS (HR = 0.26, 95%CI 0.091-0.74, P = .012) and PFS (HR = 0.27, 95%CI 0.098-0.72, P = .0095) which was confirmed in PFS subgroup analysis in the independent West Cancer Center study (N=13 HR=0.14, 95%CI 0.027-0.76, P = .023).These data confirm the association of DetermaIO with ICI clinical benefit in NSCLC, and expand on previous studies by demonstrating that first line treated PD-L1≥50% patients can further be stratified by IO score to identify efficacy. Exploratory analysis suggested that the IO score identifies benefit in patients with poor PS.
- Published
- 2022
70. Atezolizumab versus chemotherapy in advanced or metastatic NSCLC with high blood-based tumor mutational burden: primary analysis of BFAST cohort C randomized phase 3 trial
- Author
-
Solange Peters, Rafal Dziadziuszko, Alessandro Morabito, Enriqueta Felip, Shirish M. Gadgeel, Parneet Cheema, Manuel Cobo, Zoran Andric, Carlos H. Barrios, Masafumi Yamaguchi, Eric Dansin, Pongwut Danchaivijitr, Melissa Johnson, Silvia Novello, Michael S. Mathisen, Sarah M. Shagan, Erica Schleifman, Jin Wang, Mark Yan, Simonetta Mocci, David Voong, David A. Fabrizio, David S. Shames, Todd Riehl, David R. Gandara, Tony Mok, Institut Català de la Salut, [Peters S] Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland. [Dziadziuszko R] Medical University of Gdańsk, Gdańsk, Poland. [Morabito A] Istituto Nazionale Tumori ‘Fondazione G Pascale’, IRCCS, Naples, Italy. [Felip E] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Gadgeel SM] Henry Ford Cancer Institute/Henry Ford Health System, Detroit, MI, USA. [Cheema P] William Osler Health System, University of Toronto, Brampton, Ontario, Canada, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Lung Neoplasms ,phase 3 trial ,Therapeutics::Biological Therapy::Immunomodulation::Immunotherapy [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Marcadors tumorals ,terapéutica::terapia biológica::inmunomodulación::inmunoterapia [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,blood-based tumor ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,factores biológicos::biomarcadores::marcadores tumorales [COMPUESTOS QUÍMICOS Y DROGAS] ,Atezolizumab versus chemotherapy, NSCLC, blood-based tumor, mutational burden, phase 3 trial ,General Medicine ,Pulmons - Càncer - Immunoteràpia ,Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [DISEASES] ,neoplasias::neoplasias por localización::neoplasias torácicas::neoplasias del tracto respiratorio::neoplasias pulmonares::neoplasias de los bronquios::carcinoma broncogénico::carcinoma de pulmón de células no pequeñas [ENFERMEDADES] ,NSCLC ,Antibodies, Monoclonal, Humanized ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,General Biochemistry, Genetics and Molecular Biology ,Atezolizumab versus chemotherapy ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,Humans ,Immunotherapy ,Biological Factors::Biomarkers::Biomarkers, Tumor [CHEMICALS AND DRUGS] ,mutational burden - Abstract
Non-small-cell lung cancer; Predictive markers Càncer de pulmó de cèl·lules no petites; Marcadors predictius Cáncer de pulmón de células no pequeñas; Marcadores predictivos Tumor mutational burden (TMB) is being explored as a predictive biomarker for cancer immunotherapy outcomes in non-small cell lung cancer. BFAST (NCT03178552)—an open-label, global, multicohort trial—evaluated the safety and efficacy of first-line targeted therapies or immunotherapy in patients with unresectable Stage IIIB or IV advanced or metastatic non-small cell lung cancer who were selected for biomarker status using blood-based targeted next-generation sequencing. In the Phase 3 cohort C evaluating blood-based (b)TMB as a biomarker of atezolizumab efficacy, patients with bTMB of ≥10 (N = 471) were randomized 1:1 to receive atezolizumab or platinum-based chemotherapy per local standard of care. Cohort C did not meet its primary endpoint of investigator-assessed progression-free survival in the population with bTMB of ≥16 (hazard ratio, 0.77; 95% confidence interval: 0.59, 1.00; P = 0.053). Adverse events leading to treatment withdrawal occurred in 10% of patients in the atezolizumab arm and 20% in the chemotherapy arm. Adverse events of special interest occurred in 42% of patients in the atezolizumab arm and 26% in the chemotherapy arm. A prespecified exploratory analysis compared the bTMB clinical trial assay with the FoundationOne Liquid Companion Diagnostic assay and showed high concordance between assays. Additional exploration of bTMB to identify optimal cutoffs, confounding factors, assay improvements or cooperative biomarkers is warranted.
- Published
- 2022
71. Comprehensive characterization of 536 patient-derived xenograft models prioritizes candidatesfor targeted treatment
- Author
-
Li Chen, Jacqueline Mudd, Michael Ittmann, Carol J. Bult, Amanda R. Kirane, Jelena Randjelovic, Stephen Scott, Yige Wu, Li Ding, Vashisht G. Yennu-Nanda, Jing Wang, Christopher D. Lanier, Maihi Fujita, Emilio Cortes-Sanchez, Sienna Rocha, Susan G. Hilsenbeck, Kian-Huat Lim, Fernanda Martins Rodrigues, Jill Rubinstein, Nicholas Mitsiades, Haiyin Lin, Jayamanna Wickramasinghe, Andrew Butterfield, Bryan E. Welm, Alana L. Welm, Jose P. Zevallos, Jason Held, Nicole B. Coggins, Song Cao, Yuanxin Xi, Brenda C. Timmons, Paul Lott, David Menter, Shunqiang Li, Tina Primeau, Fei Yang, Andrea Wang-Gillam, Ramaswamy Govindan, Dali Li, Brandi Davis-Dusenbery, Sara Seepo, Michael C. Wendl, Jeffrey Grover, Brian S. White, Clifford G. Tepper, Peter N. Robinson, Michael A. Davies, Zhengtao Chu, Michael W. Lloyd, Hua Sun, Xiaoshan Zhang, Tamara Stankovic, Dylan Fingerman, Anuj Srivastava, Luis G. Carvajal-Carmona, Don L. Gibbons, Lijun Yao, Rebecca Aft, Hongyong Zhang, Ismail Meraz, John DiGiovanna, Scott Kopetz, Ling Zhao, Guadalupe Polanco-Echeverry, Feng Chen, Jeremy Hoog, Matthew A. Wyczalkowski, George Xu, John D. Minna, Yi Xu, Julie Belmar, Xiaowei Xu, Luc Girard, Dennis A. Dean, Tijana Borovski, Chong-xian Pan, Cynthia X. Ma, Alexa Morales Arana, Yize Li, Turcin Saridogan, Steven B. Neuhauser, Sandra Scherer, Vicki Chin, Rose Tipton, David R. Gandara, Sherri R. Davies, Argun Akcakanat, Rajesh Patidar, Julie K. Schwarz, Soner Koc, Gao Boning, Michael Kim, Bryce P. Kirby, Yvonne A. Evrard, Hyunsil Park, Christian Frech, Chia-Kuei Mo, Ran Zhang, Brian A. Van Tine, Jonathan W. Reiss, Min Xiao, Xing Yi Woo, Tiffany Le, Ana Estrada, Xiaofeng Zheng, Jeffrey A. Moscow, Mourad Majidi, Nadezhda V. Terekhanova, Katherine Fuh, Erkan Yuca, Timothy A. Yap, Jianhua Zhang, Matthew J. Ellis, Shannon Westin, James H. Doroshow, Vito W. Rebecca, Moon S. Chen, Coya Tapia, Reyka G Jayasinghe, Jack A. Roth, Jithesh Augustine, Ryan C. Fields, Michae T. Tetzlaff, Michael T. Lewis, Kurt W. Evans, Ralph W. deVere White, Brian J. Sanderson, May Cho, Jeffrey H. Chuang, Tiffany Wallace, Ryan Jeon, Ted Toal, Matthew H. Bailey, Bert W. O'Malley, Katherine L. Nathanson, Qin Liu, Benjamin J. Raphael, Jingqin Luo, Salma Kaochar, Huiqin Chen, Rajasekharan Somasundaram, Daniel Cui Zhou, John F. DiPersio, Andrew V. Kossenkov, Bingliang Fang, Vanessa Jensen, Simone Zaccaria, Alexey Sorokin, Ai-Hong Ma, Sidharth V. Puram, Min Jin Ha, Meenhard Herlyn, R. Jay Mashl, Kelly Gale, Bingbing Dai, Lacey E. Dobrolecki, Chieh-Hsiang Yang, and Funda Meric-Bernstam
- Subjects
endocrine system ,Science ,Druggability ,General Physics and Astronomy ,Genomics ,Computational biology ,Biology ,Genome ,digestive system ,General Biochemistry, Genetics and Molecular Biology ,Article ,Research community ,Multiple time ,medicine ,Cancer genomics ,Cancer models ,Tumor xenograft ,Multidisciplinary ,Cancer ,General Chemistry ,medicine.disease ,Pharmacogenomics ,Data integration ,hormones, hormone substitutes, and hormone antagonists - Abstract
Development of candidate cancer treatments is a resource-intensive process, with the research community continuing to investigate options beyond static genomic characterization. Toward this goal, we have established the genomic landscapes of 536 patient-derived xenograft (PDX) models across 25 cancer types, together with mutation, copy number, fusion, transcriptomic profiles, and NCI-MATCH arms. Compared with human tumors, PDXs typically have higher purity and fit to investigate dynamic driver events and molecular properties via multiple time points from same case PDXs. Here, we report on dynamic genomic landscapes and pharmacogenomic associations, including associations between activating oncogenic events and drugs, correlations between whole-genome duplications and subclone events, and the potential PDX models for NCI-MATCH trials. Lastly, we provide a web portal having comprehensive pan-cancer PDX genomic profiles and source code to facilitate identification of more druggable events and further insights into PDXs’ recapitulation of human tumors., Patient-derived xenograft models (PDX) have been extensively used to study the molecular and clinical features of cancers. Here the authors present a cohort of 536 PDX models from 25 cancers, as well as their genomic and evolutionary profiles and their suitability for clinical trials.
- Published
- 2021
72. Smoking Behavior in Patients With Early-Stage NSCLC: A Report From ECOG-ACRIN 1505 Trial
- Author
-
Stephen L. Graziano, Joan H. Schiller, Charles A. Butts, Conor E. Steuer, Steven M. Keller, Suresh S. Ramalingam, Opeyemi Jegede, Alex A. Adjei, William J. Tester, David R. Gandara, Heather A. Wakelee, and Suzanne E. Dahlberg
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Article ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Adjuvant therapy ,Humans ,Prospective Studies ,Lung cancer ,Smoking Reduction ,business.industry ,Incidence (epidemiology) ,Smoking ,Hazard ratio ,medicine.disease ,Former Smoker ,Clinical trial ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Smoking cessation ,Smoking Cessation ,business - Abstract
Introduction Smoking cessation has been reported to benefit patients even after a diagnosis of lung cancer. We studied the smoking behavior of patients who participated in a phase 3 trial of adjuvant therapy following resection of stages IB–IIIA NSCLC. Methods The ECOG-ACRIN 1505 was conducted to determine whether the addition of bevacizumab to adjuvant chemotherapy would improve overall survival (OS) for patients with early-stage NSCLC. Studying the association between smoking status and OS was a secondary end point. Patients completed a questionnaire on their smoking habits at baseline, 3, 6, 9, and 12 months. Results A total of 1501 patients were enrolled, and 99.8%, 95%, 94%, 93%, and 93% responded to the questionnaire at baseline, 3, 6, 9, and 12 months, respectively. A total of 90% reported a current or previous history of cigarette smoking. In addition, 60% of nonsmokers at enrollment reported smoking after diagnosis (before randomization); however, 1% of them reported smoking at 12 months. Furthermore, 94% of the respondents smoked none/fewer cigarettes daily at 12 months. The incidence of grades 3–5 toxicity on treatment was 68%, 76%, and 72% in never, former, and current smokers, respectively (p = 0.05). The disease-free survival for never-smokers relative to current and former smokers was (hazard ratio [HR] 0.93, p = 0.64 and HR 1.05, p = 0.72), and OS was (adjusted HR for death 0.54, p = 0.005 and adjusted HR for death 0.68, p = 0.03), respectively. Conclusions This is the first comprehensive, prospective report of smoking habits in patients with NSCLC patients from a phase III early-stage trial. There was a high rate of smoking reduction and cessation following study entry. The disease-free survival did not differ significantly between smokers and never smokers, though there were less grade 3–5 toxicities and more favorable OS in never-smokers.
- Published
- 2021
- Full Text
- View/download PDF
73. Abstract 5955: In Silico dissection of immune infiltrate signatures that are detected by DetermaIO, a predictor of response to immune therapy
- Author
-
Brian Z. Ring, Catherine T. Cronister, Robert S. Seitz, Douglas T. Ross, Brock Schweitzer, and David R. Gandara
- Subjects
Cancer Research ,Oncology - Abstract
Background: The composition of tumors includes not only malignant but also immune, stromal and other cell types. Understanding this dynamic tumor immune microenvironment (TIME) is important to guide treatment and develop novel therapies and markers. We have previously validated immuno-oncology gene expression signatures (DetermaIO (DTIO) and a 101-gene algorithm), that predict efficacy of checkpoint inhibitors (ICI) based on distinguishing immunomodulatory (IM), mesenchymal stem-like (MSL), and mesenchymal (M) phenotypes. In this study we used TCGA and clinical cohorts to identify immune infiltrate populations within these defined TIME spaces and their association with ICI treatment. Methods: We derived novel human immune infiltrate signatures from a translation of murine ImmGen cell populations and a search for conserved co-expression of immune markers across multiple tumors. In total, 20 tumors from TCGA were employed for derivation and analysis encompassing 7163 unique samples. These novel signatures were compared to published immune infiltrate signatures and then their association with ICI efficacy and each other assessed in three cohorts treated with ICI therapy, IMvigor210 and an additional bladder cohort, comprising 272 and 89 patients with censored outcome results, and a melanoma cohort (N=105). Results: The ImmGen analysis created 35 immune cell signatures and pan-tumor conserved co-expression of immune markers created eight signatures. The co-expression signatures often contained a mixed population of cell-type markers, though largely dominated by either myeloid or lymphoid markers. These signatures showed highly reproducible proportions of samples with strong expression between train and test TCGA sets. Most immune signatures had their highest representation in IM and DTIO+ tumors, however there was also consistent identification of presumptive immune infiltrate presence in MSL, M and DTIO negative cases. Two of the conserved co-expression signatures, one comprised of B-cell markers, and the other of T cell and other lymphoid markers, were associated with ICI efficacy in IMvigor210 and validated in the other “real-world” bladder cohort (B-cell: OR=0.8, p=0.022, T lymphoid: OR=0.7, p=0.005). Both signatures also had significant association with outcome in the cohort with clinical response outcomes, being strongest in patients after treatment had initiated. Conclusions: These cell-type signatures may be identifying novel immune infiltrate populations that co-exist within the tumor immune microenvironment and are potentially predictive of ICI response. The two signatures were not independent of DTIO in either cohort, suggesting that the 27-gene algorithm DTIO largely incorporates this information. This analysis begins to dissect the complex physiology of the tumor immune microenvironment that mediates response to immune therapy. Citation Format: Brian Z. Ring, Catherine T. Cronister, Robert S. Seitz, Douglas T. Ross, Brock Schweitzer, David R. Gandara. In Silico dissection of immune infiltrate signatures that are detected by DetermaIO, a predictor of response to immune therapy. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5955.
- Published
- 2023
- Full Text
- View/download PDF
74. Phase II study of the histone deacetylase inhibitor vorinostat (Suberoylanilide Hydroxamic Acid; SAHA) in recurrent or metastatic transitional cell carcinoma of the urothelium – an NCI-CTEP sponsored: California Cancer Consortium trial, NCI 6879
- Author
-
David I. Quinn, Paul Frankel, Edward M. Newman, Denice D. Tsao-Wei, Gurkamal Chatta, Primo N. Lara, Susan Groshen, Stella Khoo, John Wright, Heinz-Josef Lenz, Ana Aparicio, Przemyslaw Twardowski, and David R. Gandara
- Subjects
0301 basic medicine ,Pharmacology ,Oncology ,medicine.medical_specialty ,Bladder cancer ,medicine.drug_class ,business.industry ,Histone deacetylase inhibitor ,Phases of clinical research ,Cancer ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Stable Disease ,030220 oncology & carcinogenesis ,Internal medicine ,Toxicity ,medicine ,Clinical endpoint ,Pharmacology (medical) ,business ,Vorinostat ,medicine.drug - Abstract
Background: Until the advent of T cell check point inhibitors standard second-line therapy for patients with metastatic urothelial cancer (mUC) was undefined. Histone deacetylase inhibitors (HDACi) have anti-cancer activity in a variety of tumor models including modulation of apoptosis in bladder cancer cell lines. We evaluated the efficacy and toxicity of the HDACi vorinostat in patients with mUC failing first-line platinum-based therapy either in the adjuvant/neoadjuvant setting or for recurrent/advanced disease. Methods: Vorinostat was given orally 200 mg twice daily continuously until progression or unacceptable toxicity. The primary end point was RECIST response rate (RR); a RR > 20% was deemed interesting in a 2-stage design requiring one response in the first 12 patients to proceed to 2nd stage for a total of 37 subjects. CT or MRI scan imaging occurred every 6 weeks. Results: Fourteen patients were accrued characterized by: median age 66 years (43–84); Caucasian (79%); males (86%); and Karnofsky performance status ≥90 (50%). Accrual was terminated in the first stage as no responses were observed. Best response was stable disease (3 patients). Progression was observed in 8 patients. Two patients came off therapy prior to re-imaging and a 3rd patient died while on treatment and was not assessed for response. Median number of cycles was 2 (range 1–11). Median disease-free survival and overall survival times were 1.1 (0.8, 2.1) & 3.2 (2.1, 14.5) months, respectively. Toxicities were predominantly cytopenias and thrombocytopenic bleeding. Two pts. had grade 5 toxicity unlikely related to treatment. Two pts. had grade 4 and 6 had grade 3 toxicities observed. Two patients with stable disease remained on therapy for 6+ cycles. Conclusions: Vorinostat on this dose-schedule had limited efficacy and significant toxicity resulting in a unfavorable risk:benefit ratio in patients with mUC. NCT00363883.
- Published
- 2021
- Full Text
- View/download PDF
75. Atezolizumab Versus Docetaxel in Pretreated Patients With NSCLC: Final Results From the Randomized Phase 2 POPLAR and Phase 3 OAK Clinical Trials
- Author
-
Fabrice Barlesi, C. Matheny, Julien Mazieres, Diego Cortinovis, Wei Yu, Marcus Ballinger, Keunchil Park, David R. Gandara, S. Gadgeel, Achim Rittmeyer, and Toyoaki Hida
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Docetaxel ,Antibodies, Monoclonal, Humanized ,Fixed dose ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,Overall survival ,Humans ,Medicine ,In patient ,Adverse effect ,business.industry ,Antibodies, Monoclonal ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,business ,Previously treated ,medicine.drug - Abstract
Introduction The phase 2 POPLAR and phase 3 OAK studies of the anti–programmed death-ligand 1 (PD-L1) immunotherapy atezolizumab in patients with previously treated advanced NSCLC revealed significant improvements in survival versus docetaxel (p = 0.04 and 0.0003, respectively). Longer follow-up permits evaluation of continued benefit of atezolizumab. This study reports the final overall survival (OS) and safety findings from both trials. Methods POPLAR randomized 287 patients (atezolizumab, 144; docetaxel, 143) and OAK randomized 1225 patients (atezolizumab, 613; docetaxel, 612). The patients received atezolizumab (1200 mg fixed dose) or docetaxel (75 mg/m2) every 3 weeks. Efficacy and safety outcomes were evaluated. Results A longer OS was observed in patients receiving atezolizumab versus docetaxel in POPLAR (median OS = 12.6 mo versus 9.7 mo; hazard ratio = 0.76, 95% confidence interval [CI]: 0.58–1.00) and OAK (median OS = 13.3 versus 9.8 mo; hazard ratio = 0.78, 95% CI: 0.68–0.89). The 4-year OS rates in POPLAR were 14.8% (8.7–20.8) and 8.1% (3.2–13.0) and those in OAK were 15.5% (12.4–18.7) and 8.7% (6.2–11.3) for atezolizumab and docetaxel, respectively. Atezolizumab had improved OS benefit compared with docetaxel across all PD-L1 expression and histology groups. Most 4-year survivors in the docetaxel arms received subsequent immunotherapy (POPLAR, 50%; OAK, 65%). Of the 4-year survivors, most had Eastern Cooperative Oncology Group performance status of 0 and nonsquamous histological classification and approximately half were responders (POPLAR: atezolizumab, seven of 15; docetaxel, three of four; OAK: atezolizumab, 24 of 43; docetaxel, 11 of 26). Treatment-related grade 3/4 adverse events occurred in 27% and 16% of atezolizumab 4-year survivors in POPLAR and OAK, respectively. Conclusions Long-term follow-up suggests a consistent survival benefit with atezolizumab versus docetaxel in patients with previously treated NSCLC regardless of PD-L1 expression, histology, or subsequent immunotherapy. Atezolizumab had no new safety signals, and the safety profile was similar to that in previous studies.
- Published
- 2021
- Full Text
- View/download PDF
76. Phase 1 Trial of MLN0128 (Sapanisertib) and CB-839 HCl (Telaglenastat) in Patients With Advanced NSCLC (NCI 10327): Rationale and Study Design
- Author
-
Edward M. Newman, David R. Gandara, Paul K. Paik, Ramsey D. Badawi, Ian Lanza, Jonathan W. Riess, Wilson I. Gonsalves, Primo N. Lara, Mark Dunphy, Lorenzo Nardo, David Shackelford, Simon R. Cherry, Paul Frankel, Joel Reid, and Charles A. Kunos
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Lung Neoplasms ,Benzeneacetamides ,Squamous-cell lung cancer ,medicine.disease_cause ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,Non-Small-Cell Lung ,Lung ,Sapanisertib ,Cancer ,Benzoxazoles ,Lung Cancer ,Prognosis ,Tolerability ,5.1 Pharmaceuticals ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Adenocarcinoma ,KRAS ,Development of treatments and therapeutic interventions ,Glycolysis ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Combination therapy ,Clinical Sciences ,Oncology and Carcinogenesis ,Adenocarcinoma of Lung ,Phase I as Topic ,NRF2 ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,Thiadiazoles ,medicine ,Carcinoma ,Humans ,Clinical Trials ,Oncology & Carcinogenesis ,Lung cancer ,Glutaminolysis ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,KEAP1 ,Pyrimidines ,030104 developmental biology ,business - Abstract
Introduction There are currently no approved targeted therapies for lung squamous-cell carcinoma (LSCC) and KRAS-mutant lung adenocarcinoma (LUAD). About 30% of LSCC and 25% of KRAS-mutant LUAD exhibit hyperactive NRF2 pathway activation through mutations in NFE2L2 (the gene encoding NRF2) or its negative regulator, KEAP1. Preclinical data demonstrate that these tumors are uniquely sensitive to dual inhibition of glycolysis and glutaminolysis via mammalian target of rapamycin (mTOR) and glutaminase inhibitors. This phase 1 study was designed to assess safety and preliminary activity of the mTOR inhibitor MLN0128 (sapanisertib) in combination with the glutaminase inhibitor CB-839 HCl. Methods Phase 1 dose finding will use the queue-based variation of the 3 + 3 dose escalation scheme with the primary endpoint of identifying the recommended expansion dose. To confirm the acceptable tolerability of the recommended expansion dose, patients will subsequently enroll onto 1 of 4 expansion cohorts (n = 14 per cohort): (1) LSCC harboring NFE2L2 or (2) KEAP1 mutations, or (3) LUAD harboring KRAS/(KEAP1 or NFE2L2) coalterations, or (4) LSCC wild type for NFE2L2 and KEAP1. The primary endpoint of the dose expansion is to determine the preliminary efficacy of MLN0128/CB-839 combination therapy. Conclusion This phase 1 study will determine the recommended expansion dose and preliminary efficacy of MLN0128 and CB-839 in advanced non–small-cell lung cancer with a focus on subsets of LSCC and KRAS-mutant LUAD harboring NFE2L2 or KEAP1 mutations.
- Published
- 2021
- Full Text
- View/download PDF
77. A Genomically and Clinically Annotated Patient Derived Xenograft (PDX) Resource for Preclinical Research in Non-Small Cell Lung Cancer
- Author
-
Xing Yi Woo, Anuj Srivastava, Philip C. Mack, Joel H. Graber, Brian J. Sanderson, Michael W. Lloyd, Mandy Chen, Sergii Domanskyi, Regina Gandour-Edwards, Rebekah A. Tsai, James Keck, Mingshan Cheng, Margaret Bundy, Emily L. Jocoy, Jonathan W. Riess, William Holland, Stephen C. Grubb, James G. Peterson, Grace A. Stafford, Carolyn Paisie, Steven B. Neuhauser, R. Krishna Murthy Karuturi, Joshy George, Allen K. Simons, Margaret Chavaree, Clifford G. Tepper, Neal Goodwin, Susan D. Airhart, Primo N. Lara, Thomas H. Openshaw, Edison T. Liu, David R. Gandara, and Carol J. Bult
- Abstract
Patient-derived xenograft models (PDXs) are an effective preclinical in vivo platform for testing the efficacy of novel drug and drug combinations for cancer therapeutics. Here we describe a repository of 79 genomically and clinically annotated lung cancer PDXs available from The Jackson Laboratory that have been extensively characterized for histopathological features, mutational profiles, gene expression, and copy number aberrations. Most of the PDXs are models of non-small cell lung cancer (NSCLC), including 37 lung adenocarcinoma (LUAD) and 33 lung squamous cell carcinoma (LUSC) models. Other lung cancer models in the repository include four small cell carcinomas, two large cell neuroendocrine carcinomas, two adenosquamous carcinomas, and one pleomorphic carcinoma. Models with both de novo and acquired resistance to targeted therapies with tyrosine kinase inhibitors are available in the collection. The genomic profiles of the LUAD and LUSC PDX models are consistent with those observed in patient tumors of the same tumor type from The Cancer Genome Atlas (TCGA) and to previously characterized gene expression-based molecular subtypes. Clinically relevant mutations identified in the original patient tumors were confirmed in engrafted tumors. Treatment studies performed for a subset of the models recapitulated the responses expected based on the observed genomic profiles.SignificanceThe collection of lung cancer Patient Derived Xenograft (PDX) models maintained at The Jackson Laboratory retain both the histologic features and treatment-relevant genomic alterations observed in the originating patient tumors and show expected responses to treatment with standard-of-care agents. The models serve as a valuable preclinical platform for translational cancer research. Information and data for the models are freely available from the Mouse Models of Human Cancer database (MMHCdb, http://tumor.informatics.jax.org/mtbwi/pdxSearch.do).
- Published
- 2022
- Full Text
- View/download PDF
78. Strategies for the successful implementation of plasma-based NSCLC genotyping in clinical practice
- Author
-
Lynette M. Sholl, David R. Gandara, Christian D. Rolfo, Geoffrey R. Oxnard, Jhanelle E. Gray, and Charu Aggarwal
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Extramural ,Clinical Practice ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Routine clinical practice ,Clinical care ,business ,Genotyping - Abstract
Upfront tumour genotyping is now considered an essential step in guiding treatment decision-making in the management of patients with advanced-stage non-small-cell lung cancer (NSCLC) in light of the ever-expanding toolbox of targeted therapies and immune-checkpoint inhibitors. However, genotyping of tumour biopsy samples is not feasible for all patients and, therefore, genomic analysis of circulating tumour DNA (ctDNA) has emerged as a compelling non-invasive option. Current guidelines universally recommend genotyping and support the use of ctDNA testing in certain settings, although they often omit the detail necessary for integrating these tests into clinical care on an individual basis. In this Perspective, we describe the rationale, promise and challenges associated with ctDNA-based NSCLC genotyping and suggest a framework for the implementation of these assays into routine clinical practice. We also offer considerations for the interpretation of ctDNA genotyping results, which, particularly when using next-generation sequencing panels, can be nuanced. Through the addition of this new approach to clinical practice, we propose that oncologists might finally be able to utilize effective genotyping in nearly all patients with advanced-stage NSCLC.
- Published
- 2020
- Full Text
- View/download PDF
79. Forging Ahead in a Time of Crisis
- Author
-
David R. Gandara
- Subjects
Cancer Research ,medicine.medical_specialty ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,business.industry ,education ,Cancer ,medicine.disease ,Patient care ,Oncology ,Current management ,Family medicine ,Thoracic Oncology ,Pandemic ,medicine ,Lung cancer ,business - Abstract
ONCOLOGY® recently sat down with David R. Gandara, MD, director of the thoracic oncology program and senior advisor to the director of the University of California (UC) Davis Comprehensive Cancer Center, to discuss the impact of the pandemic on the current management and treatment of patients with lung cancer, as well as the future of education and patient care in a post–COVID-19 world.
- Published
- 2020
- Full Text
- View/download PDF
80. Spectrum of driver mutations and clinical impact of circulating tumor DNA analysis in non–small cell lung cancer: Analysis of over 8000 cases
- Author
-
David R. Gandara, Stefanie Mortimer, Amir Ali Talasaz, Jonathan W. Riess, Rebekah A. Burich, Christine E. Lee, Oliver A. Zill, Richard B. Lanman, Carin R. Espenschied, Philip C. Mack, and Kimberly C. Banks
- Subjects
Subset Analysis ,Male ,Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Adenocarcinoma of Lung ,Targeted therapy ,Circulating Tumor DNA ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,non–small cell lung cancer ,circulating cell‐free tumor DNA (ctDNA) ,Carcinoma, Non-Small-Cell Lung ,medicine ,ROS1 ,Biomarkers, Tumor ,Humans ,030212 general & internal medicine ,Liquid biopsy ,Lung cancer ,Protein Kinase Inhibitors ,Alleles ,liquid biopsy ,business.industry ,High-Throughput Nucleotide Sequencing ,Original Articles ,Oncogenes ,Sequence Analysis, DNA ,medicine.disease ,epidermal growth factor receptor (EGFR) ,ErbB Receptors ,Treatment Outcome ,Oncology ,anaplastic lymphoma kinase (ALK) ,Chest and Lung Disease ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Disease Progression ,Adenocarcinoma ,Population study ,Original Article ,Female ,Disease Site ,business ,Progressive disease ,Signal Transduction - Abstract
Background Circulating cell‐free tumor DNA (ctDNA)‐based mutation profiling, if sufficiently sensitive and comprehensive, can efficiently identify genomic targets in advanced lung adenocarcinoma. Therefore, the authors investigated the accuracy and clinical utility of a commercially available digital next‐generation sequencing platform in a large series of patients with non–small cell lung cancer (NSCLC). Methods Plasma‐based comprehensive genomic profiling results from 8388 consecutively tested patients with advanced NSCLC were analyzed. Driver and resistance mutations were examined with regard to their distribution, frequency, co‐occurrence, and mutual exclusivity. Results Somatic alterations were detected in 86% of samples. The median variant allele fraction was 0.43% (range, 0.03%‐97.62%). Activating alterations in actionable oncogenes were identified in 48% of patients, including EGFR (26.4%), MET (6.1%), and BRAF (2.8%) alterations and fusions (ALK, RET, and ROS1) in 2.3%. Treatment‐induced resistance mutations were common in this cohort, including driver‐dependent and driver‐independent alterations. In the subset of patients who had progressive disease during EGFR therapy, 64% had known or putative resistance alterations detected in plasma. Subset analysis revealed that ctDNA increased the identification of driver mutations by 65% over standard‐of‐care, tissue‐based testing at diagnosis. A pooled data analysis on this plasma‐based assay demonstrated that targeted therapy response rates were equivalent to those reported from tissue analysis. Conclusions Comprehensive ctDNA analysis detected the presence of therapeutically targetable driver and resistance mutations at the frequencies and distributions predicted for the study population. These findings add support for comprehensive ctDNA testing in patients who are incompletely tested at the time of diagnosis and as a primary option at the time of progression on targeted therapies., Circulating cell‐free tumor DNA‐based liquid biopsy using next‐generation sequencing detects a spectrum of targetable alterations at frequencies expected in patients with advanced non–small cell lung cancer. These findings support the concept of a plasma‐first algorithm at the time of progression on targeted therapy for this population.
- Published
- 2020
81. A phase I dose escalation, dose expansion and pharmacokinetic trial of gemcitabine and alisertib in advanced solid tumors and pancreatic cancer
- Author
-
Justin A. Chen, Jasmine C. Huynh, Chun-Yi Wu, Ai-Ming Yu, Karen Matsukuma, Thomas J. Semrad, David R. Gandara, Tianhong Li, Jonathan W. Riess, Kit Tam, Philip C. Mack, Anthony Martinez, Nichole Mahaffey, Karen L. Kelly, and Edward J. Kim
- Subjects
Cancer Research ,Alisertib ,Maximum Tolerated Dose ,Clinical Trials and Supportive Activities ,Adenocarcinoma ,Toxicology ,Deoxycytidine ,Pancreatic Cancer ,Phase I ,Rare Diseases ,Clinical Research ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Pharmacology (medical) ,Pharmacokinetics ,Oncology & Carcinogenesis ,Cancer ,Pharmacology ,Evaluation of treatments and therapeutic interventions ,Pharmacology and Pharmaceutical Sciences ,Azepines ,Gemcitabine ,Pancreatic Neoplasms ,Aurora kinase a ,Orphan Drug ,Pyrimidines ,Oncology ,6.1 Pharmaceuticals ,Patient Safety ,Digestive Diseases - Abstract
Purpose Aurora Kinase A (AKA) inhibition with gemcitabine represents a potentially synergistic cancer treatment strategy via mitotic catastrophe. The feasibility, safety, and preliminary efficacy of alisertib (MLN8237), an oral AKA inhibitor, with gemcitabine was evaluated in this open-label phase I trial with dose escalation and expansion. Methods Key inclusion criteria included advanced solid tumor with any number of prior chemotherapy regimens in the dose escalation phase, and advanced pancreatic adenocarcinoma with up to two prior chemotherapy regimens. Four dose levels (DLs 1–4) of alisertib (20, 30, 40, or 50 mg) were evaluated in 3 + 3 design with gemcitabine 1000 mg/m2 on days 1, 8, and 15 in 28-day cycles. Results In total, 21 subjects were treated in dose escalation and 5 subjects were treated in dose expansion at DL4. Dose-limiting toxicities were observed in 1 of 6 subjects each in DL3 and DL4. All subjects experienced treatment-related adverse events. Grade ≥ 3 treatment-related adverse events were observed in 73% of subjects, with neutropenia observed in 54%. Out of 22 subjects evaluable for response, 2 subjects (9%) had partial response and 14 subjects (64%) had stable disease. Median PFS was 4.1 months (95% CI 2.1–4.5). No significant changes in pharmacokinetic parameters for gemcitabine or its metabolite dFdU were observed with alisertib co-administration. Conclusions This trial established the recommended phase 2 dose of alisertib 50 mg to be combined with gemcitabine. Gemcitabine and alisertib are a feasible strategy with potential for disease control in multiple heavily pre-treated tumors, though gastrointestinal and hematologic toxicity was apparent.
- Published
- 2022
82. Novel Clinical Trial Designs in Pursuit of Precision Oncology: Lung-MAP As a Model
- Author
-
Christian Rolfo, Jonathan W. Riess, and David R. Gandara
- Subjects
Pulmonary and Respiratory Medicine ,Clinical Trials as Topic ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Lung ,business.industry ,medicine.disease ,Clinical trial ,Text mining ,medicine.anatomical_structure ,Oncology ,Research Design ,Precision oncology ,Neoplasms ,medicine ,Humans ,Medical physics ,Precision Medicine ,Lung cancer ,business - Published
- 2021
- Full Text
- View/download PDF
83. 466 Use of a 27-gene immuno-oncology (IO) assay to associate response to single-agent immune checkpoint inhibitor (ICI) therapy in advanced-stage NSCLC patients from a large Canadian cohort
- Author
-
Douglas T. Ross, Andrea Dickey, Frank McMahon, Nicole S. Croteau, David R. Gandara, Kim McGregor, Rob Seitz, David L. Saltman, Tyler J. Nielsen, David R. Hout, Jeremy Spille, Heather Lockyer, Brock L. Schweitzer, and Matthew Varga
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,Metastatic Urothelial Carcinoma ,medicine.diagnostic_test ,business.industry ,Immunology ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Retrospective cohort study ,medicine.disease ,Prostate cancer ,Internal medicine ,Biopsy ,medicine ,Molecular Medicine ,Immunology and Allergy ,business ,Lung cancer ,Triple-negative breast cancer ,RC254-282 - Abstract
BackgroundLung cancer is the leading cause of cancer-related deaths worldwide. The advent of ICIs specifically targeting programmed cell death protein-1 (PD-1), or its ligand (PD-L1) represents a major therapeutic advance that is now included in standard of care regimens for non-small-cell-lung cancer (NSCLC). PD-L1 expression measured by immunohistochemistry (IHC) staining is the current gold standard predictive biomarker for immune checkpoint inhibitor (ICI) therapy in NSCLC, however many factors beyond PD-L1 expression alone affect the outcome of ICI therapy. Evaluation of other factors to better inform clinical practice will reduce both the potential for adverse immune-related toxicities and expenditure on ineffective costly therapies while potentially identifying patients otherwise missed by PD-L1 staining. The 27-gene IO assay is a RT-qPCR based gene expression panel1 that was developed to classify the tumor immune microenvironment (TIME). It has been shown to be associated with response to ICI therapy in multiple tumor types including triple negative breast cancer, metastatic urothelial carcinoma, and NSCLC where the association was independent of PD-L1 status in patients treated either with monotherapy or combination therapy.2 Currently, BC Cancer measures PD-L1 status by IHC using the PD-L1 22C3 PharmDx assay and reports the tumor proportional score (TPS) to inform clinical decision. Patients with a TPS ≥ 50% may be eligible for first-line treatment with ICI monotherapy and those with < 50% TPS are eligible for second line or later ICI monotherapy. We established this retrospective study of ICI monotherapy treated NSCLC patients to assess the 27-gene IO assay as an informative biomarker for NSCLC ICI treatment decisions.MethodsThis retrospective study is utilizing the BC Cancer Study Database to select approximately 150 patients with stage IIIB or IV NSCLC treated with single-agent ICI therapy across four BC Cancer centers from 2017 forward (figure 1). Patients are selected based on availability of adequate biopsy specimens (FFPE with at least 20% tumor content), availability of PD-L1 IHC results or sufficient tissue to conduct staining, and for whom outcome data is available via chart review. RNA from patient samples is isolated from FFPE biopsies (either primary or metastatic sites) and those that yield ≥50ng RNA will be analyzed by the 27-gene IO assay 1 to derive IO scores (IO positive or IO negative) based on previously defined thresholds.3 The association between patient outcomes on ICI monotherapy and IO scores and PD-L1 IHC will be reported and compared.Abstract 466 Figure 1Schematic representation of patient workflow forReferencesSaltman, A, et al. Prostate cancer biomarkers and multiparametric MRI: is there a role for both in prostate cancer management? Ther Adv Urol 2021;13: 1756287221997186.Ranganath HJA, Smith JR, et al. One-year progression-free survival in lung cancer patients treated with immune checkpoint inhibitors is significantly associated with a novel immunomodulatory signature but not PD-L1 staining. in SITC. Journal Immunotherapy Cancer. 2019.Nielsen, TJ, et al. A novel immuno-oncology algorithm measuring tumor microenvironment to predict response to immunotherapies. Heliyon 2021;7(3):e06438.Ethics ApprovalThe University of British Columba BC Cancer Research Ethics Board Chair, Vice-Chair or second Vice-Chair, has reviewed the above described research project, including associated documentation, and finds the research project acceptable on ethical grounds for research involving human subjects. All participants have provided informed consent before taking part in the study. REB Number H20-02635.
- Published
- 2021
84. EGFR High Copy Number Together with High EGFR Protein Expression Predicts Improved Outcome for Cetuximab-based Therapy in Squamous Cell Lung Cancer: Analysis from SWOG S0819, a phase III trial of chemotherapy with or without Cetuximab in advanced NSCLC
- Author
-
Karen Kelly, James Moon, Fred R. Hirsch, Mary W. Redman, Thomas J. Semrad, Philip C. Mack, Marileila Varella-Garcia, Christopher J. Rivard, Francesco Agustoni, Roy S. Herbst, and David R. Gandara
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Cetuximab ,Gene Expression ,medicine.disease_cause ,Article ,Young Adult ,Antineoplastic Agents, Immunological ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Humans ,Lung cancer ,neoplasms ,Survival analysis ,Aged ,Aged, 80 and over ,Chemotherapy ,Proportional hazards model ,business.industry ,Middle Aged ,medicine.disease ,ErbB Receptors ,Treatment Outcome ,Immunohistochemistry ,Biomarker (medicine) ,Female ,KRAS ,business ,medicine.drug ,Forecasting - Abstract
Background The phase III S0819 trial investigated addition of cetuximab to first-line chemotherapy (CT) in NSCLC. Subgroup analyses suggested an OS benefit among patients with EGFR copy number gain in squamous cell carcinomas (SCC), (HR = 0.58 [0.39-0.86], P = .0071). A more detailed model based on EGFR FISH, EGFR IHC and KRAS mutation status was evaluated to yield a more precise predictive paradigm of cetuximab-based therapy in advanced NSCLC. Methods FISH was performed using the Colorado Scoring Criteria; H-Score was used to quantify EGFR IHC expression (cut-off ≥ 200). A Cox model was used to assess treatment effects for OS and PFS within biomarker and clinical subgroups. KRAS mutation was analyzed using Therascreen. The false discovery rate controlled for multiple comparisons. S0819 ClinicalTrials.gov Identifier: NCT00946712. Results Of 1,313 eligible patients, assay results were obtained for FISH on 976 patients (41% positive), for IHC on 945 patients (31% positive), and KRAS mutation status on 627 patients (26% positive). In SCC patients, OS was significantly improved with addition of cetuximab when both EGFR FISH and EGFR IHC were positive (N = 58), (OS HR: 0.32 [95% CI 0.18-0.59]; P = .0002, q = 0.08), median 12.6 versus 4.6 months. The results were independent of KRAS mutation status. In Non-SCC, no predictive value of EGFR IHC, EGFR FISH status and/or KRAS status was seen. Conclusions In NSCLC SCC, a combination index of EGFR FISH plus EGFR IHC results was associated with improved OS when cetuximab was added to CT, representing a potential predictive molecular paradigm for patients suitable for EGFR-antibody therapy.
- Published
- 2021
85. Phase 1 study of alisertib (MLN8237) and weekly irinotecan in adults with advanced solid tumors
- Author
-
David R. Gandara, Karen Kelly, Mili Arora, Scott D Christensen, Edward J. Kim, Tianhong Li, Thomas J. Semrad, I-Yeh Gong, and Jonathan W. Riess
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Colorectal cancer ,Toxicology ,Cohort Studies ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,80 and over ,Pharmacology (medical) ,6.2 Cellular and gene therapies ,Cancer ,Aged, 80 and over ,Azepines ,Hematology ,Pharmacology and Pharmaceutical Sciences ,Middle Aged ,Colo-Rectal Cancer ,Tolerability ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Female ,Patient Safety ,medicine.drug ,Adult ,medicine.medical_specialty ,Alisertib ,Maximum Tolerated Dose ,Clinical Trials and Supportive Activities ,Aurora A kinase ,Neutropenia ,Irinotecan ,Article ,Drug Administration Schedule ,03 medical and health sciences ,Young Adult ,Aurora kinase A ,Phase I ,Rare Diseases ,Refractory ,Pharmacokinetics ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Protein Kinase Inhibitors ,Aged ,Pharmacology ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,030104 developmental biology ,Pyrimidines ,Orphan Drug ,chemistry ,business ,Digestive Diseases - Abstract
PURPOSE: Aurora kinases are overexpressed or amplified in numerous malignancies. This study was designed to determine the safety and tolerability of the Aurora A kinase inhibitor alisertib (MLN8237) when combined with weekly irinotecan. METHODS: In this single center phase 1 study, adult patients with refractory advanced solid tumors received 100 mg/m(2) irinotecan intravenously on day 1 and 8 of a 21-day cycle. Alisertib at planned escalating dose levels of 20 – 60 mg was administered orally twice per day on days 1–3 and 8–10. Patients homozygous for UGT1A1*28 were excluded. The primary objective was the safety of alisertib when combined with irinotecan in order to determine the maximum tolerated dose (MTD). Secondary objectives included overall response rate by RECIST and pharmacokinetics in a planned expansion cohort of patients with colorectal cancer treated at the MTD. RESULTS: A total of 17 patients enrolled at 3 dose levels. Dose limiting toxicities included diarrhea, dehydration, and neutropenia. The MTD of alisertib combined with weekly irinotecan was 20 mg twice per day on days 1–3 and 8–10. One fatal cardiac arrest at the highest dose level tested was deemed possibly related to drug treatment. One partial response in 11 efficacy evaluable patients (9%) occurred in a patient with small cell lung cancer. The study was terminated prior to the planned expansion in patients with colorectal cancer. CONCLUSION: In contrast to prior results in a pediatric population, adult patients did not tolerate alisertib combined with irinotecan at clinically meaningful doses due to hematologic and gastrointestinal toxicities. The study was registered with ClinicalTrials.gov under study number NCT01923337 on August 15, 2013.
- Published
- 2021
86. Nivolumab Plus Ipilimumab vs Nivolumab for Previously Treated Patients With Stage IV Squamous Cell Lung Cancer
- Author
-
Lyudmila Bazhenova, Scott N. Gettinger, Fred R. Hirsch, Mary W. Redman, Thomas E. Stinchcombe, Susan S. Tavernier, Hui Yu, Karen Kelly, Lawrence H. Schwartz, Jeffrey D. Bradley, Roy S. Herbst, Philip C. Mack, Katherine Minichiello, Louise Highleyman, Vassiliki A. Papadimitrakopoulou, David R. Gandara, Natasha B. Leighl, Suresh S. Ramalingam, and Joseph M. Unger
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Ipilimumab ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Online First ,Lung cancer ,Lung ,Original Investigation ,business.industry ,Research ,Epithelial Cells ,medicine.disease ,Interim analysis ,Chemotherapy regimen ,Nivolumab ,Docetaxel ,Response Evaluation Criteria in Solid Tumors ,Carcinoma, Squamous Cell ,business ,Comments ,medicine.drug - Abstract
This phase 3 randomized clinical trial investigates if the addition of ipilimumab to nivolumab improves the survival outcome of patients with chemotherapy-pretreated immunotherapy-naive squamous cell lung cancer., Key Points Question Does the addition of ipilimumab to nivolumab improve survival in patients with advanced chemotherapy-pretreated immunotherapy-naive squamous cell lung cancer? Findings In this randomized clinical trial of 252 patients, the addition of ipilimumab to nivolumab did not lead to improved survival in patients with advanced chemotherapy-pretreated squamous cell carcinoma. Meaning Combination therapy with nivolumab and ipilimumab is currently only indicated as first-line therapy in patients with advanced non–small cell lung cancer., Importance Nivolumab plus ipilimumab is superior to platinum-based chemotherapy in treatment-naive advanced non–small cell lung cancer (NSCLC). Nivolumab is superior to docetaxel in advanced pretreated NSCLC. Objective To determine whether the addition of ipilimumab to nivolumab improves survival in patients with advanced, pretreated, immunotherapy-naive squamous (Sq) NSCLC. Design, Setting, and Participants The Lung Cancer Master Protocol (Lung-MAP) S1400I phase 3, open-label randomized clinical trial was conducted from December 18, 2015, to April 23, 2018, randomizing patients in a 1:1 ratio to nivolumab alone or combined with ipilimumab. The median follow-up in surviving patients was 29.5 months. The trial was conducted through the National Clinical Trials Network and included patients with advanced immunotherapy-naive SqNSCLC and a Zubrod score of 0 (asymptomatic) to 1 (symptomatic but completely ambulatory) with disease progression after standard platinum-based chemotherapy. Randomization was stratified by sex and number of prior therapies (1 vs 2 or more). Data were analyzed from May 3, 2018, to February 1, 2021. Interventions Nivolumab, 3 mg/kg intravenously every 2 weeks, with or without ipilimumab, 1 mg/kg intravenously every 6 weeks, until disease progression or intolerable toxic effects. Main Outcomes and Measures The primary end point was overall survival (OS). Secondary end points included investigator-assessed progression-free survival (IA-PFS) and response per Response Evaluation Criteria in Solid Tumors (RECIST) guidelines, version 1.1. Results Of 275 enrolled patients, 252 (mean age, 67.5 years [range 41.8-90.3 years]; 169 men [67%]; 206 White patients [82%]) were deemed eligible (125 randomized to nivolumab/ipilimumab and 127 to nivolumab). The study was closed for futility at a planned interim analysis. Overall survival was not significantly different between the groups (hazard ratio [HR], 0.87; 95% CI, 0.66-1.16; P = .34). Median survival was 10 months (95% CI, 8.0-14.4 months) in the nivolumab/ipilimumab group and 11 months (95% CI, 8.6-13.7 months) in the nivolumab group. The IA-PFS HR was 0.80 (95% CI, 0.61-1.03; P = .09); median IA-PFS was 3.8 months (95% CI, 2.7-4.4 months) in the nivolumab/ipilimumab group and 2.9 months (95% CI, 1.8-4.0 months) in the nivolumab alone group. Response rates were 18% (95% CI, 12%-25%) with nivolumab/ipilimumab and 17% (95% CI, 10%-23%) with nivolumab. Median response duration was 28.4 months (95% CI, 4.9 months to not reached) with nivolumab/ipilimumab and 9.7 months with nivolumab (95% CI, 4.2-23.1 months). Grade 3 or higher treatment-related adverse events occurred in 49 of 124 patients (39.5%) who received nivolumab/ipilimumab and in 41 of 123 (33.3%) who received nivolumab alone. Toxic effects led to discontinuation in 31 of 124 patients (25%) on nivolumab/ipilimumab and in 19 of 123 (15%) on nivolumab. Conclusions and Relevance In this phase 3 randomized clinical trial, ipilimumab added to nivolumab did not improve outcomes in patients with advanced, pretreated, immune checkpoint inhibitor–naive SqNSCLC. Trial Registration ClinicalTrials.gov Identifier: NCT02785952
- Published
- 2021
87. SWOG S1400D (NCT02965378), a Phase II Study of the Fibroblast Growth Factor Receptor Inhibitor AZD4547 in Previously Treated Patients With Fibroblast Growth Factor Pathway–Activated Stage IV Squamous Cell Lung Cancer (Lung-MAP Substudy)
- Author
-
Roy S. Herbst, Primo N. Lara, Jeffrey D. Bradley, Philip C. Hoffman, Vassiliki A. Papadimitrakopoulou, Alfred J. Newman, Jieling Miao, Mary W. Redman, Karen Kelly, Marvin J. Feldman, Hossein Borghaei, Philip C. Mack, Katherine Minichiello, David R. Gandara, and Charu Aggarwal
- Subjects
Male ,0301 basic medicine ,Oncology ,Lung Neoplasms ,Fibroblast Growth Factor ,Phases of clinical research ,Cardiorespiratory Medicine and Haematology ,Piperazines ,0302 clinical medicine ,80 and over ,Clinical endpoint ,Non-Small-Cell Lung ,Tumor ,Middle Aged ,FGFR inhibitor ,Survival Rate ,Response Evaluation Criteria in Solid Tumors ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Benzamides ,Cohort ,Female ,Type 3 ,Receptor ,Type 1 ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Clinical Sciences ,Oncology and Carcinogenesis ,Antineoplastic Agents ,and over ,03 medical and health sciences ,Internal medicine ,SWOG1400 ,medicine ,Humans ,Oncology & Carcinogenesis ,Adverse effect ,Neoplasm Staging ,Aged ,Salvage Therapy ,business.industry ,Fibroblast growth factor receptor 1 ,Carcinoma ,Gene Amplification ,medicine.disease ,LUNG-MAP ,030104 developmental biology ,Squamous Cell ,Pyrazoles ,business ,Biomarkers ,Progressive disease ,Follow-Up Studies - Abstract
Background S1400D is a biomarker-driven therapeutic substudy of Lung-MAP evaluating the fibroblast growth factor (FGF) receptor (FGFR) inhibitor AZD4547 in patients with FGF pathway-activated squamous cell. This is the first phase II trial to evaluate AZD4547 as a targeted approach in patients with previously treated FGFR-altered squamous cell NSCLC and is the first demonstration of successful implementation and conduct of a national umbrella protocol in this disease setting. Methods Eligible patients had tumoral FGFR alteration or mutation and had progressive disease after at least one line of platinum-based systemic therapy. Patients received AZD4547 80 mg twice daily orally. Primary endpoint was response by Response Evaluation Criteria in Solid Tumors version 1.1; secondary endpoints included progression-free survival, overall survival, and duration of response (DoR). Results Ninety-two patients were assigned to S1400D, 43 were enrolled, and 27 AZD4547-treated patients were evaluable. Evaluable patients were predominantly white (n = 24, 89%), median age 66 years (range, 49–88 years old), and female (n = 7, 26%). FGFR alterations included FGFR1 amplification (n = 23; 85%), FGFR3 amplification (n = 2; 7%), FGFR3 S249C (n = 2; 7%), and FGFR3 fusion (n = 1; 4%). Treatment with ADZ4547 was well tolerated; grade 3 adverse events occurred in six patients, and one patient had grade 4 sepsis. Of 27 response-evaluable patients, 1 patient with FGFR3 S249C had unconfirmed partial response with a DoR of 1.5 months and 1 patient with FGFR1 amplification had a confirmed partial response with a DoR of 2.9 months (7%, 95% confidence interval [CI]: 0%–17%). Median progression-free survival and overall survival for the AZD4547-treated cohort were 2.7 months (95% CI: 1.4– 4.5 months) and 7.5 months (95% CI: 3.7–9.3 months). Conclusions AZD4547 had an acceptable safety profile but minimal activity in this predominantly FGFR1/FGFR3–amplified cohort. Evaluation of other targeted agents in Lung-MAP is ongoing.
- Published
- 2019
- Full Text
- View/download PDF
88. Phase I trial of belinostat in combination with 13-cis-retinoic acid in advanced solid tumor malignancies: a California Cancer Consortium NCI/CTEP sponsored trial
- Author
-
Heinz Joseph Lenz, Thehang Luu, Edward M. Newman, Mihaela C. Cristea, Dean Lim, Paul Frankel, Leonard Joseph Appleman, David R. Gandara, Richard Piekarz, Brian F. Kiesel, and Jan H. Beumer
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Administration, Oral ,Phases of clinical research ,Hydroxamic Acids ,Toxicology ,Article ,Drug Administration Schedule ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Neoplasms ,Internal medicine ,Pancreatic cancer ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Pharmacology (medical) ,Infusions, Intravenous ,Isotretinoin ,Lung cancer ,Aged ,Aged, 80 and over ,Pharmacology ,Sulfonamides ,Dose-Response Relationship, Drug ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Toxicity ,Female ,Liver function ,business ,Belinostat - Abstract
PURPOSE: The reported maximum tolerated dose (MTD) of single agent belinostat is 1000 mg/m(2) given days 1-5, every 21 days. Pre-clinical evidence suggests histone deacetylase inhibitors enhance retinoic acid signaling in a variety of solid tumors. We conducted a phase I study of belinostat combined with 50-100 mg/m(2)/day 13-cis-retinoic acid (13-cRA) in patients with advanced solid tumors. METHODS: Belinostat was administered days 1-5 and 13-cRA days 1-14, every 21 days. Dose limiting toxicity (DLT) was defined as cycle 1 hematologic toxicity grade ≥3 not resolving to grade ≤1 within 1 week or non-hematologic toxicity grade ≥3 (except controlled nausea and vomiting and transient liver function abnormalities) attributable to belinostat. RESULTS: Among 51 patients, two DLTs were observed: grade 3 hypersensitivity with dizziness and hypoxia at 1700 mg/m(2)/day belinostat with 100 mg/m(2)/day 13-cRA, and grade 3 allergic reaction at 2000 mg/m(2)/day belinostat with 100 mg/m(2)/day 13-cRA. The MTD was not reached. Pharmacokinetics of belinostat may be non-linear at high doses. Ten patients had stable disease, including one with neuroendocrine pancreatic cancer for 56 cycles, one with breast cancer for 12 cycles, and one with lung cancer for 8 cycles. Partial responses included a patient with keratinizing squamous cell carcinoma of the tonsils, and a patient with lung cancer. CONCLUSIONS: The combination of belinostat 2000 mg/m(2) days 1-5 and 13-cRA 100 mg/m(2) days 1-14, every 21 days, was well-tolerated and an MTD was not reached despite doubling the established single agent MTD of belinostat.
- Published
- 2019
- Full Text
- View/download PDF
89. A phase II study of the orally administered negative enantiomer of gossypol (AT-101), a BH3 mimetic, in patients with advanced adrenal cortical carcinoma
- Author
-
Charles Erlichman, David R. Gandara, Hao Xie, Michael E. Menefee, Madeleine A. Kane, Alex A. Adjei, David I. Quinn, Jun Yin, Diane Reidy-Lagunes, Manisha H. Shah, and Keith C. Bible
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Phases of clinical research ,Gastroenterology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Adrenocortical Carcinoma ,Clinical endpoint ,Carcinoma ,Humans ,Medicine ,Pharmacology (medical) ,Response Evaluation Criteria in Solid Tumors ,Aged ,Pharmacology ,business.industry ,Gossypol ,Middle Aged ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Adrenal Cortex Neoplasms ,Hypokalemia ,Clinical trial ,030104 developmental biology ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Toxicity ,Female ,medicine.symptom ,business - Abstract
BACKGROUND: Adrenal cortical carcinoma (ACC) is a rare cancer with treatment options of limited efficacy, and poor prognosis if metastatic. AT-101 is a more potent inhibitor of B cell lymphoma 2 family apoptosis-related proteins than its racemic form, gossypol, which showed preliminary clinical activity in ACC. We thus evaluated the efficacy of AT-101 in patients with advanced ACC. METHODS: Patients with histologically confirmed metastatic, recurrent, or primarily unresectable ACC were treated with AT-101 (20 mg/day orally, 21 days out of 28-day cycles) until disease progression and/or prohibitive toxicity. The primary endpoint was objective response rate, wherein a Response Evaluation Criteria In Solid Tumors (RECIST) partial response rate of 25% would be considered promising and 10% not, with a Type I error of 10% and 90% power. In a 2-stage design, 2 responses were required of the first 21 assessable subjects to warrant complete accrual of 44 patients. Secondary endpoints included safety, progression-free survival and overall survival. RESULTS: This study accrued 29 patients between 2009 and 2011; median number of cycles was 2. Seven percent experienced grade 4 toxicity including cardiac troponin elevations and hypokalemia. None of the first 21 patients attained RECIST partial response; accordingly, study therapy was deemed ineffective and the trial was permanently closed. CONCLUSIONS: AT-101 had no meaningful clinical activity in this study in patients with advanced ACC, but demonstrated feasibility of prospective therapeutic clinical trials in this rare cancer.
- Published
- 2019
- Full Text
- View/download PDF
90. EBV-positive Primary Pulmonary Lymphoepithelioma-like Carcinoma Response to PD-L1 Blockade
- Author
-
Steffany Lim, Jonathan W. Riess, Anand Narayanan, David R. Gandara, Julie Ann S. Walby, and Friedrich D Knollmann
- Subjects
Pulmonary and Respiratory Medicine ,Lymphoepithelioma-like carcinoma ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Drug Resistance ,Adenocarcinoma of Lung ,Docetaxel ,Antibodies, Monoclonal, Humanized ,Deoxycytidine ,B7-H1 Antigen ,Atezolizumab ,PD-L1 ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Lung cancer ,Aged ,Neoplasm Staging ,biology ,Drug Substitution ,business.industry ,Immunotherapy ,medicine.disease ,Gemcitabine ,Blockade ,Treatment Outcome ,Oncology ,Monoclonal ,biology.protein ,Cancer research ,Female ,Antibody ,business - Published
- 2019
- Full Text
- View/download PDF
91. A first-in-human, phase 1 study of ASTX029, a dual-mechanism inhibitor of ERK1/2, in relapsed/refractory solid tumors
- Author
-
Patricia LoRusso, Drew W. Rasco, Geoffrey Shapiro, Alain C. Mita, Nilofer Saba Azad, Paul Swiecicki, Anthony B. El-Khoueiry, David R. Gandara, Shivaani Kummar, Hovig Tanajian, Jaruwan Taylor, Frank G Bottone, Marchi Toguchi, Chris Hindley, Danna Chan, Aram Oganesian, Harold N. Keer, Kim-Hien T Dao, Ryan J. Sullivan, and Alexander I. Spira
- Subjects
Cancer Research ,Oncology - Abstract
9085 Background: Aberrant activation of the RAS-RAF-MEK-ERK pathway is common in human cancers. This is an open-label Phase 1 study of ASTX029, a dual-mechanism extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor, in subjects with relapsed/refractory solid tumors (NCT03520075). Methods: The primary objective is to identify a recommended Phase 2 dose. Subjects with relapsed/refractory solid tumors were eligible for Phase 1A with any molecular feature and for Phase 1B if the tumor demonstrated RAS or BRAF mutations. ASTX029 was administered orally daily on a continuous basis in 21-day cycles. Phase 1A was a modified 3+3 dose-escalation design based on dose-limiting toxicity (DLT) events. Phase 1B subjects were treated at the recommended dose for expansion (RDE) based on emerging safety, pharmacokinetic (PK), and pharmacodynamic (PD) data. Disease response was evaluated by RECIST v1.1. Results: 76 subjects were treated with at least one dose of ASTX029 in Phase 1A (n = 56) and Phase 1B (n = 20). In Phase 1A, ASTX029 was evaluated from 10 mg to 280 mg daily. Two subjects experienced grade 2 central serous retinopathy (CSR) within a few days of dosing at the 280 mg daily dose level (one event was declared a DLT). Both subjects recovered to baseline within days of dose interruption. CSR is an expected AE based on the class of drugs. At the selected RDE dose level of 200 mg daily, the mean PK exposure was 109% of target exposure (13,022 ng*hr/ml), defined as the level expected to have biological activity based on mouse models. As of the data cut-off of February 7, 2022, the most frequent grade ≥2 AEs experienced by subjects (≥5%) assessed as related to ASTX029 included ocular AEs (n = 6: all Grade 2); nausea (n = 7: all Grade 2); diarrhea (n = 6: 5 Grade 2, 1 Grade 3); fatigue (n = 4: all Grade 2); rash (n = 4, 3 Grade 2, 1 Grade 3). There were 52 serious AEs, all unrelated to ASTX029 except for one subject with Grade 3 malaise. Four subjects had a partial response, including KRAS-G12A BRAF-D549N non-small cell lung cancer (NSCLC; Phase 1A: 120 mg treated 20.0 months); KRAS-G12D pancreatic cancer (Phase 1A: 200 mg treated 2.1 months); KRAS-G13D NSCLC (Phase 1B; treated 10.6 months); KRAS-G12S NSCLC (Phase 1B; treated 10.4 months and ongoing). In all, two partial responses were observed out of 3 NSCLC subjects enrolled in Phase 1B. Phospho-ERK and phospho-RSK were evaluated for PD effect on fresh tumor biopsies obtained at baseline and cycle 2. A PD effect and decreased cell proliferation (Ki-67) were observed in 6 of 9 and 3 of 8 evaluable Phase 1B samples, respectively. The most common reason for ASTX029 discontinuation was disease progression. Conclusions: This Phase 1 study of the ERK1/2 inhibitor ASTX029 has identified a dose level of 200 mg daily continuously for investigation in the Phase 2 study. PK and PD data suggest target exposures are achieved with preliminary clinical activity, especially in KRAS-mutated NSCLC. Clinical trial information: NCT03520075.
- Published
- 2022
- Full Text
- View/download PDF
92. A phase II study of talazoparib plus avelumab in patients with stage IV or recurrent nonsquamous non–small cell lung cancer bearing pathogenic STK11 genomic alterations (SWOG S1900C, LUNG-MAP sub-study, NCT04173507)
- Author
-
Ferdinandos Skoulidis, Mary Weber Redman, Jennifer Marie Suga, Tareq Al Baghdadi, John L. Villano, Sarah B. Goldberg, Liza C Villaruz, Katherine Minichiello, David R. Gandara, Roy S. Herbst, and Karen Kelly
- Subjects
Cancer Research ,Oncology - Abstract
9060 Background: Inactivating STK11 genomic alterations are prevalent in non-squamous (nsq) NSCLC and define a patient (pt) subgroup with poor prognosis and inferior response to immune checkpoint inhibitors (CPIs). PARP inhibitors (PARPi) can potentiate response to CPIs in preclinical models. We conducted a single arm Phase II study within Lung-MAP to evaluate the efficacy and safety of talazoparib in combination with avelumab in patients (pts) with previously treated nsq NSCLC harboring pathogenic STK11 genomic alterations. Methods: Eligibility: STK11 pathogenic somatic mutation or bi-allelic loss on tumor identified via LUNGMAP screening; stage IV or recurrent nsq NSCLC, receipt of one prior line of anti-PD-1/anti-PD-L1 therapy and platinum-based chemotherapy for stage IV or recurrent disease (sequentially or in combination) and disease progression > 42 days following treatment initiation, a ECOG PS of 0-1, adequate organ function and no previous PARPi exposure. Pts received talazoparib (1000 mg PO daily) plus avelumab (800 mg IV Q2W). Co-primary objectives were to evaluate the best objective response rate (ORR) and disease control rate at 12 weeks (DCR12) after study registration, assessed by RECISTv1.1. Rejection of an ORR of 10% required ≥ 8 responses or rejection of a DCR12 of 30% required ≥18 w/ disease control at 12 weeks and ≥4 responses. Results: 47 pts enrolled from January 16 - November 16, 2020; 42 pts met eligibility (50% male, 50% female). 54% of pts had PD-L1 TPS < 1%. The median TMB was 8.83 Mut/Mb and 45% of pts had KRAS mutations. 52% of the pts had received ≥2 prior lines of treatment for stage IV disease. As of the November 24, 2021 data cutoff, 3 pts remain on treatment, the ORR was 2% (n = 1) and the DCR12 was 40% (n = 17). 26 pts (62%) had SD as best objective response. One responding pt remained on treatment for > 14 mo. The median progression-free survival (39 events) was 2.7 mo (95% CI, 1.6-3.9 mo) and the median overall survival (30 events) was 7.6 mo (95% CI, 6.3-12.2 mo). There were no reported grade 5 treatment toxicities and most grade 3-4 toxicities were hematologic. Additional biomarker analysis to assess effects of key co-mutations on clinical outcomes will be presented. Conclusions: Treatment with talazoparib and avelumab did not meet the pre-specified threshold for efficacy in previously treated STK11-mutant NSCLC in this biomarker-driven Phase II study, though durable disease stabilization was observed. Further studies are required to determine optimal therapeutic approaches for this challenging subset of NSCLC pts. Funding: NIH/NCI grants U10CA180888, U10CA180819. Talazoparib was provided by Pfizer. Avelumab was provided by Pfizer, as part of an alliance between Pfizer and the healthcare business of Merck KGaA, Darmstadt, Germany (CrossRef Funder ID: 10.13039/100004755). Clinical trial information: NCT04173507.
- Published
- 2022
- Full Text
- View/download PDF
93. Osimertinib plus necitumumab in EGFR-mutant NSCLC: Final results from an ETCTN California Cancer Consortium phase I study
- Author
-
Jonathan W. Riess, Mark D. Krailo, Sukhmani Kaur Padda, Susan G. Groshen, Heather A. Wakelee, Karen L. Reckamp, Marianna Koczywas, Zofia Piotrowska, Conor Ernst Steuer, Chul Kim, Cloud P. Paweletz, Lynette M. Sholl, Grace Heavey, Jill Kolesar, Jeffrey Moscow, Pasi A. Janne, Primo \\'Lucky\\' N. Lara, Edward M. Newman, and David R. Gandara
- Subjects
Cancer Research ,Oncology - Abstract
9014 Background: Osimertinib (Osi) is standard of care in 1st line (1L) EGFR mut NSCLC and TKI resistant T790Mpos NSCLC but acquired resistance emerges; outcomes are less robust in T790Mneg, C797Xpos and EGFR exon 20 insertion (ex20ins) disease. We examined Osi with the EGFR monoclonal antibody Necitumumab (Neci) in select settings of EGFR TKI resistance. Methods: Pts were accrued to 5 expansion cohorts (ExC) at recommended phase 2 dose (RP2D) of Osi 80 mg daily and Neci 800 mg D1 + D8 of q21d cycle. ExC (18 pts/cohort): A) T790Mneg progressive disease (PD) on 1st/2nd gen TKI as last therapy, B) T790Mneg PD on 1st/2nd gen TKI and PD on 3rd gen TKI, C) T790Mpos PD on 1st/2nd gen TKI and PD on 3rd gen TKI, D) EGFR ex20ins PD on chemotherapy, E) PD on 1L osi. In ExC A-C, T790M was confirmed centrally (tissue) by ddPCR. Additional correlative studies include: tissue NGS (> 400 gene panel), EGFR FISH, plasma for PK and serial EGFR ctDNA by ddPCR. Adverse events were graded (Gr) by CTCAEv5; ORR, PFS by RECIST 1.1. Primary pre-specified efficacy endpoint ≥3/18 pts responding per cohort. Results: 101 patients accrued (100 evaluable). Efficacy is summarized in the Table. Drug related Gr 3 AEs were seen in 38% of pts, mainly rash (21%). ORR among all pts was 19% (95% CI 12-28%) that varied across cohorts (Table). In ExC A-C, 69% pts had detectable EGFR activating mutations in ctDNA, with decline in mutant allele frequency (AF) on treatment in 80% and ctDNA clearance in 33%. Conclusions: Osi/Neci is feasible and tolerable at the RP2D. EGFR ctDNA was detectable at baseline in the majority of pts with decrease in AF on treatment. Osi/Neci was active in select settings of EGFR-TKI resistance, meeting its prespecified efficacy endpoint in T790Mneg PD on 1st/2nd gen TKI as last therapy (ExC A), EGFR ex20ins post-chemo (ExC D) and PD on 1L osimertinib (ExC E). mPFS in the EGFR ex20ins cohort was within the range of current EGFR Exon 20 ins agents in development. EGFR monoclonal antibodies with osimertinib warrant further study in settings of de novo and acquired EGFR dependent resistance to EGFR-TKI. Clinical trial information: NCT02496663. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
94. Overall survival from a phase II randomized study of ramucirumab plus pembrolizumab versus standard of care for advanced non–small cell lung cancer previously treated with immunotherapy: Lung-MAP nonmatched substudy S1800A
- Author
-
Karen L. Reckamp, Mary Weber Redman, Konstantin H. Dragnev, Liza C Villaruz, Bryan A. Faller, Tareq Al Baghdadi, Susan Hines, Lu Qian, Katherine Minichiello, David R. Gandara, Karen Kelly, and Roy S. Herbst
- Subjects
Cancer Research ,Oncology - Abstract
9004 Background: Resistance to immune checkpoint inhibitor (ICI) therapy develops in most patients (pts) with advanced non-small cell lung cancer (NSCLC). Tumors that develop resistance to ICI constitute a major unmet need. Combined ICI and VEGF/VEGF receptor inhibition have shown benefit in multiple tumor types through immune modulation. We evaluated pembrolizumab and ramucirumab (P+R) in advanced, ICI-exposed NSCLC, under the aegis of Lung-MAP, a master protocol for pts with stage IV, previously treated NSCLC. Pt characteristics and treatment toxicities were presented at ASCO 2021. Methods: S1800A was a randomized phase II trial for pts ineligible for a biomarker-matched substudy with acquired resistance to ICI defined as previous ICI therapy for at least 84 days with progressive disease (PD) on or after therapy. Eligibility stipulated PD on prior platinum-based doublet therapy (sequential or in combination with ICI) and ECOG PS of 0-1. Pts were stratified by PD-L1 expression, histology, and intent to receive ramucirumab in the standard of care (SOC) arm and were randomized to P+R or SOC (investigator’s choice of docetaxel+R; docetaxel, pemetrexed, gemcitabine). With a goal of 144 total/130 eligible pts, the primary objective was to compare overall survival (OS) between the arms using a 1-sided 10% level log-rank test upon 90 deaths. Secondary endpoints included response, duration of response, investigator assessed-progression free survival and toxicity. Results: From May 17, 2019 to November 16, 2020, 166 pts were enrolled with 137 eligible (69 P+R; 68 SOC [45 +R, 23 w/o R]). Main causes for ineligibility were lack of PD on ICI or chemotherapy (6 SOC, 6 P+R), > 1 line of ICI (2 P+R), ICI discontinued due to toxicity (2 SOC), or lack of measurable disease (2 SOC, 1 P+R). OS was significantly improved with P+R (HR: 0.61 [0.38-0.97], 1-sided p-value = 0.019; median [95% CI] OS of 15.0 (13.2-17) months (mo) for P+R and 11.6 (8.5-13.8) mo in SOC arm). Progression-free survival (PFS) was not different between the arms (HR: 0.86 [0.57-1.31], 1-sided p-value=0.25; median PFS (95% CI) of 4.5 (4.0-6.9) mo for P+R and 5.2 (4.0-6.6) mo in SOC arm). ORR was not different between the arms (p=0.28). OS benefit for P+R was seen in most subgroups. Analysis of survival based on genomic alterations, tumor mutational burden and PD-L1 will be presented. Conclusions: Pembrolizumab + ramucirumab in pts with advanced NSCLC previously treated with chemotherapy and immunotherapy led to improved OS compared to SOC. Discordance of ORR and PFS from OS has been reported in prior ICI trials (Rittmeyer et al. Lancet 2017). This is the first trial in the 2nd line setting without a chemotherapy backbone to demonstrate a potential survival benefit compared to SOC regimens including docetaxel and ramucirumab using the Lung-MAP platform. Clinical trial information: NCT03971474.
- Published
- 2022
- Full Text
- View/download PDF
95. The 27-gene IO score is associated with molecular features and response to immune checkpoint inhibitors (ICI) in patients with gastric cancer
- Author
-
Matthew Gordon Varga, Catherine Cronister, Tyler J. Nielsen, Douglas Teller Ross, David Richard Hout, Robert Seitz, Kimberly McGregor, David R. Gandara, and Brock L Schweitzer
- Subjects
Cancer Research ,Oncology - Abstract
4058 Background: Gastric cancer (GC) is the 3rd leading cause of cancer-related death worldwide. Unfortunately, most gastric cancer patients are asymptomatic until the cancer has progressed to an advanced stage. ICIs have improved patient outcomes in a variety of cancers, including GC. A variety of biomarkers have been used to identify patients most likely to benefit from ICI therapies such as high PD-L1 expression, MSI-high, Epstein Barr virus (EBV) positive, or TMB. Despite these potential biomarkers, most patients with advanced GC do not respond to ICI treatment Thus, there remains an unmet clinical need for a biomarker that can better predict response to ICI therapies. Herein, we demonstrate that the 27 gene IO score, a tumor immune microenvironment (TIME) classifier is associated with the existing molecular markers of gastric cancer and with objective response to ICI therapy in a clinical cohort. Methods: RNA-seq expression data was obtained from 3 independent cohorts including TCGA (STAD), ACRG (GSE84437, GSE84426), and clinical cohort with ICI response data (PRJEB25780, PRJEB40416). The 27 gene IO algorithm was applied to all available patient data to derive IO scores. Fisher’s exact test was used to examine the associations between IO score and clinical features and molecular subtypes in each cohort. R (version 4.1.2) was used to calculate ORs with 95%CIs, and ordinal logistic regression modeling. Results: From the TCGA cohort, the IO score was associated with the molecular features of EBV, MSI, TMB, and PD-L1 (n = 135, p < 0.05 for all). Similarly, in the ACRG cohorts, the IO score was significantly associated with EBV, MSI, and PD-L1 ( n = 294, p < 0.001 for all). To determine whether the IO score was associated with response to ICIs, we examined a cohort of Korean patients with advanced stage GC curated by Kim et. al. In this cohort of 59 patients, the IO score was associated with ICI response (Fisher’s exact test, p < 0.05). When response was grouped by responders vs. non-responders (CR/PR vs SD/PD), the odds ratio for the association between IO score and response was 5.3 (95% CI: 1.3 to 23.92, p = 0.01). The linearity of continuous value of the IO score was indicative of a direct relationship between IO score and improved objective response (ordinal logistic regression, t = 2.59, p < 0.01). Conclusions: PD-L1 and TMB have shown marked levels of both spatial and temporal heterogeneity in GCs, thus there exists a need for a more comprehensive biomarker that can fully assess the TIME. The 27 gene IO score is associated with many existing biomarkers in GC and has now been shown to also be associated with response to ICIs. As such, further studies are warranted to demonstrate that the 27 gene IO score may be a more comprehensive biomarker for assessing the TIME and provide complementary data to tumor-specific biomarkers, which together could aid in clinical decision making for ICI treatment of GCs.
- Published
- 2022
- Full Text
- View/download PDF
96. Representativeness of patients enrolled in the Lung Cancer Master Protocol (Lung-MAP)
- Author
-
Riha Vaidya, Joseph M. Unger, Lu Qian, Katherine Minichiello, Roy S. Herbst, David R. Gandara, Joel W. Neal, Ticiana Leal, Jyoti D. Patel, Konstantin H. Dragnev, Saiama Naheed Waqar, Martin Joseph Edelman, Ellen V. Sigal, Stacey Adam, Shakun M. Malik, Charles David Blanke, Michael Leo LeBlanc, Karen Kelly, and Mary Weber Redman
- Subjects
Cancer Research ,Oncology - Abstract
6543 Background: A major goal of Lung-MAP, a biomarker-driven master protocol conducted within the National Clinical Trials Network of the NCI using a public-private partnership, was to improve access to novel therapeutics. Representative enrollment of patient sub-groups in clinical trials is essential for improving confidence that trial findings are valid and applicable to all patients. We examined the representativeness of patients enrolled in Lung-MAP by demographic and area-level measures compared to patients in other advanced non-small cell lung cancer (NSCLC) trials and with the US NSCLC population. Methods: We analyzed data on patients enrolled to Lung-MAP between 2014-2020 according to sex, age ( < 65 years v. ≥ 65 years), race (White v. Black v. Asian), ethnicity (Hispanic v. not Hispanic), residence (rural v. urban), insurance type (Medicaid or no insurance v. private), and neighborhood socioeconomic deprivation (quintiles of Area Deprivation Index score). Rates were compared to SWOG-led NSCLC trials conducted between 2001-2020 (date range to provide sufficient power) and, where possible, to US NSCLC population rates using Surveillance, Epidemiology, and End Results (SEER) registry data (2014-2018). Two-sided tests of proportions at the 5% level were used for all comparisons. Results: 3,556 patients enrolled to Lung-MAP were compared to 2,267 patients enrolled to SWOG-led NSCLC studies. Lung-MAP patients were more likely to be ≥ 65 years old (57.2% v. 46.7%; p
- Published
- 2022
- Full Text
- View/download PDF
97. A Phase II Study of Telisotuzumab Vedotin in Patients With c-MET-positive Stage IV or Recurrent Squamous Cell Lung Cancer (LUNG-MAP Sub-study S1400K, NCT03574753)
- Author
-
Karen Kelly, Suresh S. Ramalingam, David R. Gandara, Jeffrey D. Bradley, Lawrence H. Schwartz, Natasha B. Leighl, Philip C. Mack, Katherine Minichiello, Saloni H. Tanna, Thomas E. Stinchcombe, Vassiliki A. Papadimitrakopoulou, Roy S. Herbst, Susanne M. Arnold, Saiama N. Waqar, Fred R. Hirsch, Mary W. Redman, and Ryan S. Raddin
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Lung Neoplasms ,Phases of clinical research ,Cohort Studies ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Squamous cell carcinoma ,Monoclonal ,Clinical endpoint ,80 and over ,Non-Small-Cell Lung ,Lung ,Cancer ,Aged, 80 and over ,Lung-MAP ,Telisotuzumab vedotin ,Lung Cancer ,Antibodies, Monoclonal ,Middle Aged ,Proto-Oncogene Proteins c-met ,Progression-Free Survival ,Survival Rate ,Treatment Outcome ,Local ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Cohort ,Carcinoma, Squamous Cell ,Female ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Antibody-drug conjugate ,Clinical Trials and Supportive Activities ,Clinical Sciences ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Article ,Antibodies ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Lung cancer ,Pneumonitis ,Neoplasm Staging ,Aged ,c-MET ,business.industry ,Carcinoma ,Pneumonia ,medicine.disease ,Interim analysis ,030104 developmental biology ,Neoplasm Recurrence ,Orphan Drug ,Squamous Cell ,Neoplasm Recurrence, Local ,business - Abstract
Introduction Lung-MAP S1400K was designed to evaluate the response to telisotuzumab vedotin, an antibody-drug conjugate targeting c-MET, in patients with c-MET–positive squamous cell carcinoma (SCC). Patients and Methods Patients with previously treated SCC with c-MET–positive tumors (H score ≥ 150, Ventana SP44 assay) were enrolled into 2 cohorts: Cohort 1 (immune checkpoint inhibitor-naive) and Cohort 2 (immune checkpoint inhibitor refractory). Telisotuzumab vedotin 2.7 mg/kg was administered intravenously every 3 weeks until disease progression or unacceptable toxicity. Response assessments were performed every 6 weeks. The primary endpoint was response by Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. Secondary endpoints included progression-free survival, overall survival, response within cohort, duration of response, and toxicities. Interim analysis was planned after 20 evaluable patients, with ≥ 3 responses needed to continue enrollment. Results Forty-nine patients (14% of screened patients) were assigned to S1400K, 28 patients enrolled (15 in Cohort 1 and 13 in Cohort 2), and 23 were eligible. S1400K closed on December 21, 2018 owing to lack of efficacy. Two responses (response rate of 9%; 95% confidence interval, 0%-20%) were reported in cohort 1 (1 complete and 1 unconfirmed partial response), whereas 10 patients had stable disease, with a disease control rate of 52%. The median overall and progression-free survival was 5.6 and 2.4 months, respectively. There were 3 grade 5 events (2 pneumonitis, in Cohort 2, and 1 bronchopulmonary hemorrhage, in Cohort 1). Conclusion Telisotuzumab vedotin failed to meet the pre-specified response needed to justify continuing enrollment to S1400K. Pneumonitis was an unanticipated toxicity observed in patients with SCC.
- Published
- 2021
98. Fast progression in non–small cell lung cancer: results from the randomized phase III OAK study evaluating second-line atezolizumab versus docetaxel
- Author
-
David R. Gandara, S. Morris, A. Cardona, Julien Mazieres, Martin Reck, Marcus Ballinger, Diana Mendus, Denis Moro-Sibilot, Shirish M. Gadgeel, Achim Rittmeyer, and Solange Peters
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Lung Neoplasms ,Time Factors ,medicine.medical_treatment ,Docetaxel ,0302 clinical medicine ,Risk Factors ,Carcinoma, Non-Small-Cell Lung ,Monoclonal ,Immunology and Allergy ,Medicine ,Epidermal growth factor receptor ,Non-Small-Cell Lung ,Humanized ,Immune Checkpoint Inhibitors ,Lung ,RC254-282 ,Cancer ,Clinical/Translational Cancer Immunotherapy ,biology ,Lung Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Middle Aged ,Tubulin Modulators ,Tumor Burden ,Treatment Outcome ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Disease Progression ,Molecular Medicine ,Female ,immunotherapy ,medicine.drug ,medicine.medical_specialty ,Immunology ,Clinical Trials and Supportive Activities ,Antibodies, Monoclonal, Humanized ,Risk Assessment ,Antibodies ,03 medical and health sciences ,Atezolizumab ,Clinical Research ,Internal medicine ,Humans ,Lung cancer ,Neoplasm Staging ,Aged ,Pharmacology ,Chemotherapy ,Performance status ,business.industry ,Carcinoma ,Immunotherapy ,medicine.disease ,030104 developmental biology ,biology.protein ,business - Abstract
BackgroundTreatment-induced accelerated tumor growth is a progression pattern reported with immune checkpoint inhibitors that has never been evaluated in randomized phase III studies because it requires two pretreatment scans. This study aimed to develop clinically relevant and applicable criteria for fast progression (FP), incorporating tumor growth kinetics and early death from disease progression to analyze data from the randomized phase III OAK study.MethodsThe OAK study evaluated the efficacy and safety of atezolizumab versus docetaxel as second-line or third-line treatment for stage IIIb/IV non–small cell lung cancer. FP rates and associated baseline factors were analyzed. FP was defined as either a ≥50% increase in the sum of largest diameters (SLDs) within 6 weeks of treatment initiation or death due to cancer progression within 12 weeks (absent post-baseline scan).ResultsForty-two of 421 patients (10%) receiving atezolizumab and 37 of 402 (9%) receiving docetaxel had FP. Twenty patients with FP (48%) receiving atezolizumab versus 12 (30%) receiving docetaxel had a ≥50% SLD increase within 6 weeks. FP was significantly associated with an ECOG (Eastern Cooperative Oncology Group) performance status of 1 (vs 0), ≥3 metastatic sites at baseline, and failure of preceding first-line treatment within 6 months, but not with epidermal growth factor receptor mutation, programmed cell death 1 ligand 1 or tumor mutational burden. Overall survival in patients with FP and a ≥50% SLD increase at week 6 was similar with atezolizumab and docetaxel (unstratified HR 0.89 (95% CI 0.41 to 1.92)).ConclusionsFP rates were similar with atezolizumab and docetaxel in the OAK study, suggesting that FP may not be unique to checkpoint inhibitors, although the underlying mechanisms may differ from those of chemotherapy. Applying the FP criteria to other phase III checkpoint inhibitor trials may further elucidate the risk factors for FP.Trial registration numberNCT02008227.
- Published
- 2021
99. 1281O Atezolizumab (atezo) vs platinum-based chemo in blood-based tumour mutational burden-positive (bTMB+) patients (pts) with first-line (1L) advanced/metastatic (m)NSCLC: Results of the Blood First Assay Screening Trial (BFAST) phase III cohort C
- Author
-
M. Yamaguchi, Carlos H. Barrios, E. Felip, Z. Andric, M. Mathisen, Eric Dansin, Rafal Dziadziuszko, Shirish M. Gadgeel, Melissa Lynne Johnson, Parneet Cheema, Tony Mok, Jingjing Wang, Silvia Novello, M. Cobo Dols, David R. Gandara, P. Danchaivijitr, S.M. Shagan, Alessandro Morabito, Solange Peters, and Erica B. Schleifman
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Atezolizumab ,Screening Trial ,Internal medicine ,First line ,Cohort ,Medicine ,Hematology ,business - Published
- 2021
- Full Text
- View/download PDF
100. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer
- Author
-
Maria E. Arcila, Umberto Malapelle, Giorgio V. Scagliotti, Christopher Abbosh, Lynette M. Sholl, Daniel Tan, Ignacio I. Wistuba, Christian Rolfo, Charles Swanton, Rafal Dziadziuszko, Charu Aggarwal, Rebecca Bunn, Heather A. Wakelee, Nir Peled, Maximilian Diehn, Fabrice Barlesi, M. Wynes, Trever G. Bivona, Philip C. Mack, Janet Freeman-Daily, Lecia V. Sequist, Caroline Dive, David R. Gandara, Natasha B. Leighl, Luis E. Raez, Tony Mok, Tetsuya Mitsudomi, Chandra P. Belani, Rolfo, C., Mack, P., Scagliotti, G. V., Aggarwal, C., Arcila, M. E., Barlesi, F., Bivona, T., Diehn, M., Dive, C., Dziadziuszko, R., Leighl, N., Malapelle, U., Mok, T., Peled, N., Raez, L. E., Sequist, L., Sholl, L., Swanton, C., Abbosh, C., Tan, D., Wakelee, H., Wistuba, I., Bunn, R., Freeman-Daily, J., Wynes, M., Belani, C., Mitsudomi, T., and Gandara, D.
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Consensus ,Lung Neoplasms ,Druggability ,Consensu ,NSCLC ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,IASLC ,Liquid biopsy ,Lung cancer ,Genotyping ,Oncogene ,ctDNA ,Immunotherapy ,Oncogene addicted ,business.industry ,Cancer ,Oncogenes ,medicine.disease ,Precision medicine ,Egfr mutation ,Adenocarcinoma ,business ,Human - Abstract
Although precision medicine has had a mixed impact on the clinical management of patients with advanced-stage cancer overall, for NSCLC, and more specifically for lung adenocarcinoma, the advances have been dramatic, largely owing to the genomic complexity and growing number of druggable oncogene drivers. Furthermore, although tumor tissue is historically the "accepted standard" biospecimen for these molecular analyses, there are considerable innate limitations. Thus, liquid biopsy represents a practical alternative source for investigating tumor-derived somatic alterations. Although data are most robust in NSCLC, patients with other cancer types may also benefit from this minimally invasive approach to facilitate selection of targeted therapies. The liquid biopsy approach includes a variety of methodologies for circulating analytes. From a clinical point of view, plasma circulating tumor DNA is the most extensively studied and widely adopted alternative to tissue tumor genotyping in solid tumors, including NSCLC, first entering clinical practice for detection of EGFR mutations in NSCLC. Since the publication of the first International Association for the Study of Lung Cancer (IASLC) liquid biopsy statement in 2018, several additional advances have been made in this field, leading to changes in the therapeutic decision-making algorithm for advanced NSCLC and prompting this 2021 update. In view of the novel and impressive technological advances made in the past few years, the growing clinical application of plasma-based, next-generation sequencing, and the recent Food and Drug and Administration approval in the United States of two different assays for circulating tumor DNA analysis, IASLC revisited the role of liquid biopsy in therapeutic decision-making in a recent workshop in October 2020 and the question of "plasma first" versus "tissue first" approach toward molecular testing for advanced NSCLC. Moreover, evidence-based recommendations from IASLC provide an international perspective on when to order which test and how to interpret the results. Here, we present updates and additional considerations to the previous statement article as a consensus from a multidisciplinary and international team of experts selected by IASLC.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.