486 results on '"Philippe L Bedard"'
Search Results
2. Phase IB study of ziv-aflibercept plus pembrolizumab in patients with advanced solid tumors
- Author
-
Howard Streicher, Elad Sharon, Mariano Severgnini, Michael Manos, Anita Giobbie-Hurder, F Stephen Hodi, Andrew S Brohl, Philippe L Bedard, Kevin Tyan, Scott Rodig, Osama E Rahma, Daniel J Renouf, Emma Hathaway, and Rachel Cunningham
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background The combination of antiangiogenic agents with immune checkpoint inhibitors could potentially overcome immune suppression driven by tumor angiogenesis. We report results from a phase IB study of ziv-aflibercept plus pembrolizumab in patients with advanced solid tumors.Methods This is a multicenter phase IB dose-escalation study of the combination of ziv-aflibercept (at 2–4 mg/kg) plus pembrolizumab (at 2 mg/kg) administered intravenously every 2 weeks with expansion cohorts in programmed cell death protein 1 (PD-1)/programmed death-ligand 1(PD-L1)-naïve melanoma, renal cell carcinoma (RCC), microsatellite stable colorectal cancer (CRC), and ovarian cancer. The primary objective was to determine maximum tolerated dose (MTD) and recommended dose of the combination. Secondary endpoints included overall response rate (ORR) and overall survival (OS). Exploratory objectives included correlation of clinical efficacy with tumor and peripheral immune population densities.Results Overall, 33 patients were enrolled during dose escalation (n=3) and dose expansion (n=30). No dose-limiting toxicities were reported in the initial dose level. Ziv-aflibercept 4 mg/kg plus pembrolizumab 2 mg/kg every 2 weeks was established as the MTD. Grade ≥3 adverse events occurred in 19/33 patients (58%), the most common being hypertension (36%) and proteinuria (18%). ORR in the dose-expansion cohort was 16.7% (5/30, 90% CI 7% to 32%). Complete responses occurred in melanoma (n=2); partial responses occurred in RCC (n=1), mesothelioma (n=1), and melanoma (n=1). Median OS was as follows: melanoma, not reached (NR); RCC, 15.7 months (90% CI 2.5 to 15.7); CRC, 3.3 months (90% CI 0.6 to 3.4); ovarian, 12.5 months (90% CI 3.8 to 13.6); other solid tumors, NR. Activated tumor-infiltrating CD8 T cells at baseline (CD8+PD1+), high CD40L expression, and increased peripheral memory CD8 T cells correlated with clinical response.Conclusion The combination of ziv-aflibercept and pembrolizumab demonstrated an acceptable safety profile with antitumor activity in solid tumors. The combination is currently being studied in sarcoma and anti-PD-1-resistant melanoma.Trial registration number NCT02298959.
- Published
- 2022
- Full Text
- View/download PDF
3. First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas
- Author
-
Ravit Geva, Jennifer Mataraza, Osama Rahma, Jason John Luke, Philippe L Bedard, Sarina A Piha-Paul, Angad Singh, Tira J Tan, Darren WT Lim, Cinta Hierro, Toshikiko Doi, Alexander Lesokhin, Javier Otero, Lisa Nardi, Alexandros Xyrafas, and Xinhui Chen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background GWN323 is an IgG1 monoclonal antibody (mAb) against the glucocorticoid-induced tumor necrosis factor receptor-related protein. This first-in-human, open-label phase I/Ib study aimed to investigate the safety and tolerability and to identify the recommended doses of GWN323 with/without spartalizumab, an anti-programmed cell death receptor-1 agent, for future studies. Pharmacokinetics, preliminary efficacy and efficacy biomarkers were also assessed.Methods Patients (aged ≥18 years) with advanced/metastatic solid tumors with Eastern Cooperative Oncology Group performance status of ≤2 were included. GWN323 (10–1500 mg) or GWN323+spartalizumab (GWN323 10–750 mg+spartalizumab 100–300 mg) were administered intravenously at various dose levels and schedules during the dose-escalation phase. Dose-limiting toxicities (DLTs) were assessed during the first 21 days in a single-agent arm and 42 days in a combination arm. Adverse events (AEs) were graded per National Cancer Institute-Common Toxicity Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors V.1.1.Results Overall, 92 patients (single-agent, n=39; combination, n=53) were included. The maximum administered doses (MADs) in the single-agent and combination arms were GWN323 1500 mg every 3 weeks (q3w) and GWN323 750 mg+spartalizumab 300 mg q3w, respectively. No DLTs were observed with single-agent treatment. Three DLTs (6%, all grade ≥3) were noted with combination treatment: blood creatine phosphokinase increase, respiratory failure and small intestinal obstruction. Serious AEs were reported in 30.8% and 34.0%, and drug-related AEs were reported in 82.1% and 77.4% of patients with single-agent and combination treatments, respectively. Disease was stable in 7 patients and progressed in 26 patients with single-agent treatment. In combination arm patients, 1 had complete response (endometrial cancer); 3, partial response (rectal cancer, adenocarcinoma of colon and melanoma); 14, stable disease; and 27, disease progression. GWN323 exhibited a pharmacokinetic profile typical of mAbs with a dose-dependent increase in the pharmacokinetic exposure. Inconsistent decreases in regulatory T cells and increases in CD8+ T cells were observed in the combination arm. Gene expression analyses showed no significant effect of GWN323 on interferon-γ or natural killer-cell signatures.Conclusions GWN323, as a single agent and in combination, was well tolerated in patients with relapsed/refractory solid tumors. The MAD was 1500 mg q3w for single-agent and GWN323 750 mg+spartalizumab 300 mg q3w for combination treatments. Minimal single-agent activity and modest clinical benefit were observed with the spartalizumab combination.Trial registration number NCT02740270.
- Published
- 2021
- Full Text
- View/download PDF
4. Evaluation of liver enzyme elevations and hepatotoxicity in patients treated with checkpoint inhibitor immunotherapy.
- Author
-
Morven Cunningham, Marco Iafolla, Yada Kanjanapan, Orlando Cerocchi, Marcus Butler, Lillian L Siu, Philippe L Bedard, Kendra Ross, Bettina Hansen, Anna Spreafico, and Jordan J Feld
- Subjects
Medicine ,Science - Abstract
Background and aimsImmune checkpoint inhibitors (ICI) are increasingly used in cancer therapy. Elevated liver enzymes frequently occur in patients treated with ICI but evaluation is poorly described. We sought to better understand causes of liver enzyme elevation, investigation and management.MethodsPatients treated with anti-PD-1, PDL-1 or CTLA-4 therapy in Phase I/II clinical trials between August 2012 and December 2018 were included. Clinical records of patients with significant liver enzyme elevations were retrospectively reviewed.ResultsOf 470 ICI-treated patients, liver enzyme elevation occurred in 102 (21.6%), attributed to disease progression (56; 54.9%), other drugs/toxins (7; 6.9%), other causes (22; 21.6%) and ICI immunotoxicity (17; 16.7%; 3.6% of total cohort). Immunotoxicity was associated with higher peak ALT than other causes of enzyme elevation (N = 17; M = 217, 95% CI 145-324 for immunotoxicity, N = 103; M = 74, 95% CI 59-92 for other causes; ratio of means 0.34, 95% CI 0.19-0.60, p = ConclusionsLiver enzyme elevation is common in patients receiving ICI, but often has a cause other than immunotoxicity. A biochemical signature with higher ALT and ALT/AST ratio, a history of prior ICI exposure and other organ immunotoxicities may help to identify patients at a higher likelihood of immunotoxicity. Liver biopsy can be safely deferred in most patients. We propose an approach to diagnostic evaluation in patients with liver enzyme elevations following ICI exposure.
- Published
- 2021
- Full Text
- View/download PDF
5. An open-label, phase II multicohort study of an oral hypomethylating agent CC-486 and durvalumab in advanced solid tumors
- Author
-
Neda Stjepanovic, Amit M Oza, Anna Spreafico, Stephanie Lheureux, Marcus O Butler, Pamela S Ohashi, Kirsty Taylor, Helen Loo Yau, Ankur Chakravarthy, Ben Wang, Shu Yi Shen, Ilias Ettayebi, Charles A Ishak, Philippe L Bedard, Albiruni Abdul Razak, Aaron R Hansen, Dave Cescon, Brendan Van As, Sarah Boross-Harmer, Lisa Wang, Trevor J Pugh, Lillian L Siu, and Daniel D De Carvalho
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Purpose To evaluate whether administration of the oral DNA hypomethylating agent CC-486 enhances the poor response rate of immunologically ‘cold’ solid tumors to immune checkpoint inhibitor durvalumab.Experimental design PD-L1/PD-1 inhibitor naïve patients with advanced microsatellite stable colorectal cancer; platinum resistant ovarian cancer; and estrogen receptor positive, HER2 negative breast cancer were enrolled in this single-institution, investigator-initiated trial. Two 28 day regimens, regimen A (CC-486 300 mg QD Days 1–14 (cycles 1–3 only) in combination with durvalumab 1500 mg intravenous day 15) and regimen B (CC-486 100 mg QD days 1–21 (cycle 1 and beyond), vitamin C 500 mg once a day continuously and durvalumab 1500 mg intravenous day 15) were investigated. Patients underwent paired tumor biopsies and serial peripheral blood mononuclear cells (PBMCs) collection for immune-profiling, transcriptomic and epigenomic analyzes.Results A total of 28 patients were enrolled, 19 patients treated on regimen A and 9 on regimen B. The combination of CC-486 and durvalumab was tolerable. Regimen B, with a lower dose of CC-486 extended over a longer treatment course, showed less grade 3/4 adverse effects. Global LINE-1 methylation assessment of serial PBMCs and genome-wide DNA methylation profile in paired tumor biopsies demonstrated minimal changes in global methylation in both regimens. The lack of robust tumor DNA demethylation was accompanied by an absence of the expected ‘viral mimicry’ inflammatory response, and consequently, no clinical responses were observed. The disease control rate was 7.1%. The median progression-free survival was 1.9 months (95% CI 1.5 to 2.3) and median overall survival was 5 months (95% CI 4.5 to 10).Conclusions The evaluated treatment schedules of CC-486 in combination with durvalumab did not demonstrate robust pharmacodynamic or clinical activity in selected immunologically cold solid tumors. Lessons learned from this biomarker-rich study should inform continued drug development efforts using these agents.Trial registration number NCT02811497.
- Published
- 2020
- Full Text
- View/download PDF
6. Tumor reactive γδ T cells contribute to a complete response to PD-1 blockade in a Merkel cell carcinoma patient
- Author
-
Scott C. Lien, Dalam Ly, S. Y. Cindy Yang, Ben X. Wang, Derek L. Clouthier, Michael St. Paul, Ramy Gadalla, Babak Noamani, Carlos R. Garcia-Batres, Sarah Boross-Harmer, Philippe L. Bedard, Trevor J. Pugh, Anna Spreafico, Naoto Hirano, Albiruni R. A. Razak, and Pamela S. Ohashi
- Subjects
Science - Abstract
Abstract Immunotherapies targeting PD-1/PD-L1 are now widely used in the clinic to treat a variety of malignancies. While most of the research on T cell exhaustion and PD-1 blockade has been focused on conventional αβ T cells, the contribution of innate-like T cells such as γδ T cells to anti-PD-1/PD-L1 mediated therapy is limited. Here we show that tumor reactive γδ T cells respond to PD-1 blockade in a Merkel cell carcinoma (MCC) patient experiencing a complete response to therapy. We find clonally expanded γδ T cells in the blood and tumor after pembrolizumab treatment, and this Vγ2Vδ1 clonotype recognizes Merkel cancer cells in a TCR-dependent manner. Notably, the intra-tumoral γδ T cells in the MCC patient are characterized by higher expression of PD-1 and TIGIT, relative to conventional CD4 and CD8 T cells. Our results demonstrate that innate-like T cells could also contribute to an anti-tumor response after PD-1 blockade.
- Published
- 2024
- Full Text
- View/download PDF
7. Autoimmune PaneLs as PrEdictors of Toxicity in Patients TReated with Immune Checkpoint InhibiTors (ALERT)
- Author
-
Sofia Genta, Katherine Lajkosz, Noelle R. Yee, Pavlina Spiliopoulou, Alya Heirali, Aaron R. Hansen, Lillian L. Siu, Sam Saibil, Lee-Anne Stayner, Maryia Yanekina, Maxwell B. Sauder, Sareh Keshavarzi, Abdulazeez Salawu, Olga Vornicova, Marcus O. Butler, Philippe L. Bedard, Albiruni R. Abdul Razak, Robert Rottapel, Andrzej Chruscinski, Bryan Coburn, and Anna Spreafico
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Immune-checkpoint inhibitors (ICI) can lead to immune-related adverse events (irAEs) in a significant proportion of patients. The mechanisms underlying irAEs development are mostly unknown and might involve multiple immune effectors, such as T cells, B cells and autoantibodies (AutoAb). Methods We used custom autoantigen (AutoAg) microarrays to profile AutoAb related to irAEs in patients receiving ICI. Plasma was collected before and after ICI from cancer patients participating in two clinical trials (NCT03686202, NCT02644369). A one-time collection was obtained from healthy controls for comparison. Custom arrays with 162 autoAg were used to detect IgG and IgM reactivities. Differences of median fluorescent intensity (MFI) were analyzed with Wilcoxon sign rank test and Kruskal–Wallis test. MFI 500 was used as threshold to define autoAb reactivity. Results A total of 114 patients and 14 healthy controls were included in this study. irAEs of grade (G) ≥ 2 occurred in 37/114 patients (32%). We observed a greater number of IgG and IgM reactivities in pre-ICI collections from patients versus healthy controls (62 vs 32 p
- Published
- 2023
- Full Text
- View/download PDF
8. Impact on costs and outcomes of multi-gene panel testing for advanced solid malignancies: a cost-consequence analysis using linked administrative dataResearch in context
- Author
-
Alberto Hernando-Calvo, Paul Nguyen, Philippe L. Bedard, Kelvin K.W. Chan, Ramy R. Saleh, Deirdre Weymann, Celeste Yu, Eitan Amir, Dean A. Regier, Bishal Gyawali, Danielle Kain, Brooke Wilson, Craig C. Earle, Nicole Mittmann, Albiruni R. Abdul Razak, Wanrudee Isaranuwatchai, Peter Sabatini, Anna Spreafico, Tracy L. Stockley, Trevor J. Pugh, Christine Williams, Lillian L. Siu, and Timothy P. Hanna
- Subjects
Precision oncology ,Cost-consequence analysis ,NGS ,Molecular profiling ,Matched therapies ,Medicine (General) ,R5-920 - Abstract
Summary: Background: To date, economic analyses of tissue-based next generation sequencing genomic profiling (NGS) for advanced solid tumors have typically required models with assumptions, with little real-world evidence on overall survival (OS), clinical trial enrollment or end-of-life quality of care. Methods: Cost consequence analysis of NGS testing (555 or 161-gene panels) for advanced solid tumors through the OCTANE clinical trial (NCT02906943). This is a longitudinal, propensity score-matched retrospective cohort study in Ontario, Canada using linked administrative data. Patients enrolled in OCTANE at Princess Margaret Cancer Centre from August 2016 until March 2019 were matched with contemporary patients without large gene panel testing from across Ontario not enrolled in OCTANE. Patients were matched according to 19 patient, disease and treatment variables. Full 2-year follow-up data was available. Sensitivity analyses considered alternative matched cohorts. Main Outcomes were mean per capita costs (2019 Canadian dollars) from a public payer's perspective, OS, clinical trial enrollment and end-of-life quality metrics. Findings: There were 782 OCTANE patients with 782 matched controls. Variables were balanced after matching (standardized difference 0.40), and greater in two (ovary, biliary, both p
- Published
- 2024
- Full Text
- View/download PDF
9. Seize the engine: Emerging cell cycle targets in breast cancer
- Author
-
Jesús Fuentes‐Antrás, Philippe L. Bedard, and David W. Cescon
- Subjects
breast cancer ,cell cycle checkpoints ,clinical trials ,cyclin‐dependent kinases ,drug development ,mitotic kinases ,Medicine (General) ,R5-920 - Abstract
Abstract Breast cancer arises from a series of molecular alterations that disrupt cell cycle checkpoints, leading to aberrant cell proliferation and genomic instability. Targeted pharmacological inhibition of cell cycle regulators has long been considered a promising anti‐cancer strategy. Initial attempts to drug critical cell cycle drivers were hampered by poor selectivity, modest efficacy and haematological toxicity. Advances in our understanding of the molecular basis of cell cycle disruption and the mechanisms of resistance to CDK4/6 inhibitors have reignited interest in blocking specific components of the cell cycle machinery, such as CDK2, CDK4, CDK7, PLK4, WEE1, PKMYT1, AURKA and TTK. These targets play critical roles in regulating quiescence, DNA replication and chromosome segregation. Extensive preclinical data support their potential to overcome CDK4/6 inhibitor resistance, induce synthetic lethality or sensitise tumours to immune checkpoint inhibitors. This review provides a biological and drug development perspective on emerging cell cycle targets and novel inhibitors, many of which exhibit favourable safety profiles and promising activity in clinical trials.
- Published
- 2024
- Full Text
- View/download PDF
10. Addressing racial and ethnic disparities in AACR project GENIE
- Author
-
Shawn M. Sweeney, Jessica A. Lavery, Hannah E. Fuchs, Jocelyn A. Lee, Samantha Brown, Katherine S. Panageas, Charles L. Sawyers, and Philippe L. Bedard
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
11. Predicting response and toxicity to PD-1 inhibition using serum autoantibodies identified from immuno-mass spectrometry [version 1; peer review: 2 approved]
- Author
-
Milena Music, Marco Iafolla, Antoninus Soosaipillai, Ihor Batruch, Ioannis Prassas, Melania Pintilie, Aaron R. Hansen, Philippe L. Bedard, Stephanie Lheureux, Anna Spreafico, Albiruni Abdul Razak, Lillian L. Siu, and Eleftherios P. Diamandis
- Subjects
Research Article ,Articles ,programmed cell death protein 1 ,immune checkpoint blockade ,pembrolizumab ,response ,predictive biomarkers ,autoantibodies ,anti-thyroglobulin antibody ,anti-thyroid peroxidase antibody ,hypothyroidism ,immune-related adverse events - Abstract
Background: Validated biomarkers are needed to identify patients at increased risk of immune-related adverse events (irAEs) to immune checkpoint blockade (ICB). Antibodies directed against endogenous antigens can change after exposure to ICB. Methods: Patients with different solid tumors stratified into cohorts received pembrolizumab every 3 weeks in a Phase II trial (INSPIRE study). Blood samples were collected prior to first pembrolizumab exposure (baseline) and approximately 7 weeks (pre-cycle 3) into treatment. In a discovery analysis, autoantibody target immuno-mass spectrometry was performed in baseline and pre-cycle 3 pooled sera of 24 INSPIRE patients based on clinical benefit (CBR) and irAEs. Results: Thyroglobulin (Tg) and thyroid peroxidase (TPO) were identified as the candidate autoantibody targets. In the overall cohort of 78 patients, the frequency of CBR and irAEs from pembrolizumab was 31% and 24%, respectively. Patients with an anti-Tg titer increase ≥1.5x from baseline to pre-cycle 3 were more likely to have irAEs relative to patients without this increase in unadjusted, cohort adjusted, and multivariable models (OR=17.4, 95% CI 1.8–173.8, p=0.015). Similarly, patients with an anti-TPO titer ≥ 1.5x from baseline to pre-cycle 3 were more likely to have irAEs relative to patients without the increase in unadjusted and cohort adjusted (OR=6.1, 95% CI 1.1–32.7, p=0.035) models. Further, the cohort adjusted analysis showed patients with anti-Tg titer greater than median (10.0 IU/mL) at pre-cycle 3 were more likely to have irAEs (OR=4.7, 95% CI 1.2–17.8, p=0.024). Patients with pre-cycle 3 anti-TPO titers greater than median (10.0 IU/mL) had a significant difference in overall survival (23.8 vs 11.5 months; HR=1.8, 95% CI 1.0–3.2, p=0.05). Conclusions: Patient increase ≥1.5x of anti-Tg and anti-TPO titers from baseline to pre-cycle 3 were associated with irAEs from pembrolizumab, and patients with elevated pre-cycle 3 anti-TPO titers had an improvement in overall survival.
- Published
- 2020
- Full Text
- View/download PDF
12. A phase II trial of GSK2256098 and trametinib in patients with advanced pancreatic ductal adenocarcinoma
- Author
-
Kyaw L. Aung, Elaine McWhirter, Stephen Welch, Lisa Wang, Sophia Lovell, Lee-Anne Stayner, Saara Ali, Anne Malpage, Barbara Makepeace, Makilpriya Ramachandran, Gun Ho Jang, Steven Gallinger, Tong Zhang, Tracy L. Stockley, Sandra E. Fischer, Neesha Dhani, David Hedley, Jennifer J. Knox, Lillian L. Siu, Rachel Goodwin, and Philippe L. Bedard
- Subjects
Oncology ,Gastroenterology - Published
- 2022
- Full Text
- View/download PDF
13. Phase II Study of Afatinib in Patients With Tumors With Human Epidermal Growth Factor Receptor 2-Activating Mutations: Results From the National Cancer Institute-Molecular Analysis for Therapy Choice ECOG-ACRIN Trial (EAY131) Subprotocol EAY131-B
- Author
-
Philippe L. Bedard, Shuli Li, Kari B. Wisinski, Eddy S. Yang, Sewanti A. Limaye, Edith P. Mitchell, James A. Zwiebel, Jeffrey A. Moscow, Robert J. Gray, Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David R. Patton, P. Mickey Williams, Stanley R. Hamilton, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, and Keith T. Flaherty
- Subjects
Diarrhea ,Male ,Cancer Research ,Receptor, ErbB-2 ,Breast Neoplasms ,Stroke Volume ,Middle Aged ,Afatinib ,National Cancer Institute (U.S.) ,United States ,Ventricular Function, Left ,Oncology ,Receptors, Estrogen ,Mutation ,Quinazolines ,Humans ,Female ,In Situ Hybridization, Fluorescence - Abstract
PURPOSE National Cancer Institute–Molecular Analysis for Therapy Choice is a multicohort trial that assigns patients with advanced cancers to targeted therapies on the basis of central tumor genomic testing. Arm B evaluated afatinib, an ErbB family tyrosine kinase inhibitor, in patients with ERBB2-activating mutations. METHODS Eligible patients had selected ERBB2 single-nucleotide variants or insertions/deletions detected by the National Cancer Institute–Molecular Analysis for Therapy Choice next-generation sequencing assay. Patients had performance status ≤ 1, left ventricular ejection fraction > 50%, grade ≤ 1 diarrhea, and no prior human epidermal growth factor receptor 2 (HER2) therapy. Patients received afatinib 40 mg once daily in 28-day cycles. The primary end point was objective response rate (ORR). Secondary end points were 6-month progression-free survival, overall survival, toxicity, and molecular correlates. RESULTS A total of 59 patients were assigned and 40 were enrolled. The median age was 62 years, 78% were female, 68% had performance status = 1, and 58% had received > 3 prior therapies. The confirmed ORR was 2.7% (n = 1 of 37; 90% CI, 0.14 to 12.2), and 6-month progression-free survival was 12.0% (90% CI, 5.6 to 25.8). A confirmed partial response occurred in a patient with adenocarcinoma of extra-mammary Paget disease of skin who progressed after cycle 6. Two unconfirmed partial responses were observed (low-grade serous gynecological tract and estrogen receptor–positive/HER2-negative immunohistochemistry breast ductal carcinoma). Of 12 patients with breast cancer, 1 additional patient with lobular carcinoma (estrogen receptor–positive/HER2 fluorescent in situ hybridization) had a 51% reduction in target lesions but progressed because of a new lesion at cycle 6. The most common (> 20%) treatment-related adverse events were diarrhea (68%), mucositis (43%), fatigue (40%), acneiform rash (30%), dehydration (27%), vomiting (27%), nausea (27%), anemia (27%), and anorexia (22%). Four patients (11%) discontinued because of adverse events. CONCLUSION Although afatinib did not meet the prespecified threshold for antitumor activity in this heavily pretreated cohort, the response in a rare tumor type is notable. The safety profile of afatinib was consistent with prior studies.
- Published
- 2023
14. The GENIE BPC NSCLC cohort: a real-world repository integrating standardized clinical and genomic data for 1,846 patients with non-small cell lung cancer
- Author
-
Noura J. Choudhury, Jessica A. Lavery, Samantha Brown, Ino de Bruijn, Justin Jee, Thinh Ngoc. Tran, Hira Rizvi, Kathryn C. Arbour, Karissa Whiting, Ronglai Shen, Matthew Hellmann, Philippe L. Bedard, Celeste Yu, Natasha Leighl, Michele LeNoue-Newton, Christine Micheel, Jeremy L. Warner, Michelle S. Ginsberg, Andrew Plodkowski, Jeffrey Girshman, Peter Sawan, Shirin Pillai, Shawn M. Sweeney, Kenneth L. Kehl, Katherine S. Panageas, Nikolaus Schultz, Deb Schrag, and Gregory J. Riely
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: We describe the clinical and genomic landscape of the non-small cell lung cancer (NSCLC) cohort of the AACR Project GENIE Biopharma Collaborative (BPC). Experimental Design: 1,846 patients with NSCLC whose tumors were sequenced from 2014 to 2018 at four institutions participating in AACR GENIE were randomly chosen for curation using the PRISSMMÔ data model. Progression-free survival (PFS) and overall survival (OS) were estimated for patients treated with standard therapies. Results: In this cohort, 44% of tumors harbored a targetable oncogenic alteration, with EGFR (20%), KRAS G12C (13%) and oncogenic fusions (ALK, RET and ROS1; 5%) as the most frequent. Median OS (mOS) on first-line platinum-based therapy without immunotherapy was 17.4 mo (95% confidence interval (CI) 14.9,19.5 mo). For second-line therapies, mOS was 9.2 mo (95% CI 7.5,11.3 mo) for immune checkpoint inhibitors (ICI) and 6.4 mo (95% CI 5.1,8.1 mo) for docetaxel +/- ramucirumab. In a subset of patients treated with ICI in the second-line or later setting, median RECIST PFS (2.5 mo; 95% CI 2.2, 2.8) and median real-world PFS based on imaging reports (2.2 mo; 95% CI 1.7,2.6) were similar. In exploratory analysis of the impact of tumor mutational burden (TMB) on survival on ICI treatment in the second-line or higher setting, TMB z-score harmonized across gene panels was associated with improved OS (univariable HR 0.85, p=0.03, n=247 patients). Conclusions: The GENIE BPC cohort provides comprehensive clinico-genomic data for patients with NSCLC, which can improve understanding of real-world patient outcomes.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Information from Heterogeneity of Circulating Tumor Cell–Associated Genomic Gains in Breast Cancer and Its Association with the Host Immune Response
- Author
-
Susan J. Done, David McCready, Philippe L. Bedard, Martin J. Yaffe, John M.S. Bartlett, Naoto Hirano, Trevor D. McKee, Janina Kulka, Borbala Szekely, Christos Sotiriou, Christine Desmedt, Kela Liu, Rachel Peters, Yulia Yerofeyeva, Alison Cheung, Dan Wang, Fred Fu, Amanda Mao, Carrie Wei, Sabrina Manolescu, Eshetu G. Atenafu, Manjula Maganti, Shiyi Chen, Melanie Dawe, Ranju Nair, Zaldy Balde, and Nisha Kanwar
- Abstract
Includes Supplemental Figure S1 and Supplemental Tables S1-S7
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary File from Heterogeneity of Circulating Tumor Cell–Associated Genomic Gains in Breast Cancer and Its Association with the Host Immune Response
- Author
-
Susan J. Done, David McCready, Philippe L. Bedard, Martin J. Yaffe, John M.S. Bartlett, Naoto Hirano, Trevor D. McKee, Janina Kulka, Borbala Szekely, Christos Sotiriou, Christine Desmedt, Kela Liu, Rachel Peters, Yulia Yerofeyeva, Alison Cheung, Dan Wang, Fred Fu, Amanda Mao, Carrie Wei, Sabrina Manolescu, Eshetu G. Atenafu, Manjula Maganti, Shiyi Chen, Melanie Dawe, Ranju Nair, Zaldy Balde, and Nisha Kanwar
- Abstract
Raw cell counts of FISH gene probes for (1) primary breast tumors, (2) metastatic sites, (3) pre- and post-treatment [Hungarian sample], and (4) TNBC cases.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Methods, Supplemental Tables 1-2, Supplemental Figures 1-4 from AACR Project GENIE: Powering Precision Medicine through an International Consortium
- Author
-
Hongxin Zhang, Ahmet Zehir, Emily Yu, Thomas V. Yu, Celeste Yu, Stuart Watt, Chetna Wathoo, Lucy L. Wang, Emile E. Voest, Carl Virtanen, Victor E. Velculescu, Harm van Tinteren, Tony van de Velde, Laura J. Van 'T Veer, Eliezer M. Van Allen, Stacy B. Thomas, Jelle J. ten Hoeve, Barry S. Taylor, Shawn M. Sweeney, Thomas P. Stricker, Natalie H. Stickle, Parin Sripakdeevong, Jean Charles Soria, Gabe S. Sonke, David B. Solit, Lillian L. Siu, Priyanka Shivdasani, Kenna R Mills Shaw, Nikolaus Schultz, Deborah Schrag, Charles L. Sawyers, Mark J. Routbort, Barrett J. Rollins, Brendan Reardon, Trevor J. Pugh, Ben Ho Park, John A. Orechia, Larsson Omberg, Petra M. Nederlof, Nathanael D. Moore, Gordon Mills, Clinton Miller, Christine M. Micheel, Funda Meric-Bernstam, Gerrit A. Meijer, David S. Maxwell, Ian Maurer, Laura E. MacConaill, Zhibin Lu, David Liu, James Lindsay, Neal I. Lindeman, Mia A. Levy, Eva M. Lepisto, Michele L. LeNoue-Newton, Céline Lefebvre, Marc Ladanyi, Ritika Kundra, Priti Kumari, Walter Kinyua, Cyriac Kandoth, Suzanne Kamel-Reid, David M. Hyman, Jan Hudeček, Hugo Horlings, Stephanie Hintzen, Zachary J. Heins, Justin Guinney, Benjamin E. Gross, Christopher D. Gocke, Stu Gardos, Francisco Garcia, Jianjiong Gao, P. Andrew Futreal, Matthew D. Ducar, Raymond N. DuBois, Semih Dogan, Catherine Del Vecchio Fitz, Nancy E. Davidson, Kristen K. Dang, Debyani Chakravarty, Ethan Cerami, Fabien Calvo, Mariska Bierkens, Michael F. Berger, Philippe L. Bedard, José Baselga, Alexander S. Baras, Monica Arnedos, and Fabrice André
- Abstract
Supplemental Methods. Supplemental Table 1: ââ,¬â€¹Genomic Data Characterization by Center. Supplemental Table 2: ââ,¬â€¹Gene Panels Submitted by Each Center. Figure S1: Number of putative germline SNPs per sample, before and after uniform germline filtering. Figure S2ââ,¬â€¹. Distribution of total somatic mutation burden per sample stratified by sequencing panel. Figure S3: ââ,¬â€¹Log-scale comparison of mutation frequencies at hotspot sites between GENIE (data aggregated from all sequencing panels) and cancerhotspots.org (CHS) using a binomial test. Figure S4:ââ,¬â€¹ Comparison of mutation frequencies at hotspot sites in each GENIE sequencing panel with cancerhotspots.org (CHS) using a binomial test.
- Published
- 2023
- Full Text
- View/download PDF
18. Table S4 from AACR Project GENIE: Powering Precision Medicine through an International Consortium
- Author
-
Hongxin Zhang, Ahmet Zehir, Emily Yu, Thomas V. Yu, Celeste Yu, Stuart Watt, Chetna Wathoo, Lucy L. Wang, Emile E. Voest, Carl Virtanen, Victor E. Velculescu, Harm van Tinteren, Tony van de Velde, Laura J. Van 'T Veer, Eliezer M. Van Allen, Stacy B. Thomas, Jelle J. ten Hoeve, Barry S. Taylor, Shawn M. Sweeney, Thomas P. Stricker, Natalie H. Stickle, Parin Sripakdeevong, Jean Charles Soria, Gabe S. Sonke, David B. Solit, Lillian L. Siu, Priyanka Shivdasani, Kenna R Mills Shaw, Nikolaus Schultz, Deborah Schrag, Charles L. Sawyers, Mark J. Routbort, Barrett J. Rollins, Brendan Reardon, Trevor J. Pugh, Ben Ho Park, John A. Orechia, Larsson Omberg, Petra M. Nederlof, Nathanael D. Moore, Gordon Mills, Clinton Miller, Christine M. Micheel, Funda Meric-Bernstam, Gerrit A. Meijer, David S. Maxwell, Ian Maurer, Laura E. MacConaill, Zhibin Lu, David Liu, James Lindsay, Neal I. Lindeman, Mia A. Levy, Eva M. Lepisto, Michele L. LeNoue-Newton, Céline Lefebvre, Marc Ladanyi, Ritika Kundra, Priti Kumari, Walter Kinyua, Cyriac Kandoth, Suzanne Kamel-Reid, David M. Hyman, Jan Hudeček, Hugo Horlings, Stephanie Hintzen, Zachary J. Heins, Justin Guinney, Benjamin E. Gross, Christopher D. Gocke, Stu Gardos, Francisco Garcia, Jianjiong Gao, P. Andrew Futreal, Matthew D. Ducar, Raymond N. DuBois, Semih Dogan, Catherine Del Vecchio Fitz, Nancy E. Davidson, Kristen K. Dang, Debyani Chakravarty, Ethan Cerami, Fabien Calvo, Mariska Bierkens, Michael F. Berger, Philippe L. Bedard, José Baselga, Alexander S. Baras, Monica Arnedos, and Fabrice André
- Abstract
Table S4
- Published
- 2023
- Full Text
- View/download PDF
19. Data from Genomic and Transcriptomic Analyses of Breast Cancer Primaries and Matched Metastases in AURORA, the Breast International Group (BIG) Molecular Screening Initiative
- Author
-
Martine J. Piccart, David Venet, Matteo Benelli, Lucy R. Yates, Florentine S. Hilbers, Carmela Caballero, Andrea Vingiani, Mariana Brandão, Maite Lasa Montoya, Nadia Harbeck, David A. Cameron, Susan Knox, Sherene Loi, Philippe L. Bedard, Oskar Th Johannsson, Nancy E. Davidson, Gabriele Zoppoli, Barbro Linderholm, Sibylle Loibl, Ethel Seyll, Marta Paoli, Ásgerdur Sverrisdóttir, Richard Greil, Silvia Khodaverdi, Martina Degiorgis, Karolina Fs Larsson, Andrea Bonetti, Peter Hall, Angel Guerrero-Zotano, Hans Wildiers, Andrea Gombos, Beatriz Rojas, Dario Romagnoli, Sandrine Marreaud, Caroline Duhem, Joan Albanell, Fatima Cardoso, Marija Balic, Marco Colleoni, Jorge S. Reis-Filho, Judith M. Bliss, Giuseppe Curigliano, Elsemieke D. Scheepers, Giuseppe Viale, Evandro de Azambuja, Eva M. Ciruelos, Angelo Di Leo, Justin Ndozeng, Mark E. Robson, Einav Nili Gal-Yam, Christos Sotiriou, Debora Fumagalli, Alexandre Irrthum, Mafalda Oliveira, and Philippe Aftimos
- Abstract
AURORA aims to study the processes of relapse in metastatic breast cancer (MBC) by performing multi-omics profiling on paired primary tumors and early-course metastases. Among 381 patients (primary tumor and metastasis pairs: 252 targeted gene sequencing, 152 RNA sequencing, 67 single nucleotide polymorphism arrays), we found a driver role for GATA1 and MEN1 somatic mutations. Metastases were enriched in ESR1, PTEN, CDH1, PIK3CA, and RB1 mutations; MDM4 and MYC amplifications; and ARID1A deletions. An increase in clonality was observed in driver genes such as ERBB2 and RB1. Intrinsic subtype switching occurred in 36% of cases. Luminal A/B to HER2-enriched switching was associated with TP53 and/or PIK3CA mutations. Metastases had lower immune score and increased immune-permissive cells. High tumor mutational burden correlated to shorter time to relapse in HR+/HER2− cancers. ESCAT tier I/II alterations were detected in 51% of patients and matched therapy was used in 7%. Integration of multi-omics analyses in clinical practice could affect treatment strategies in MBC.Significance:The AURORA program, through the genomic and transcriptomic analyses of matched primary and metastatic samples from 381 patients with breast cancer, coupled with prospectively collected clinical data, identified genomic alterations enriched in metastases and prognostic biomarkers. ESCAT tier I/II alterations were detected in more than half of the patients.This article is highlighted in the In This Issue feature, p. 2659
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary methods, tables and figures. from Genomic and Transcriptomic Analyses of Breast Cancer Primaries and Matched Metastases in AURORA, the Breast International Group (BIG) Molecular Screening Initiative
- Author
-
Martine J. Piccart, David Venet, Matteo Benelli, Lucy R. Yates, Florentine S. Hilbers, Carmela Caballero, Andrea Vingiani, Mariana Brandão, Maite Lasa Montoya, Nadia Harbeck, David A. Cameron, Susan Knox, Sherene Loi, Philippe L. Bedard, Oskar Th Johannsson, Nancy E. Davidson, Gabriele Zoppoli, Barbro Linderholm, Sibylle Loibl, Ethel Seyll, Marta Paoli, Ásgerdur Sverrisdóttir, Richard Greil, Silvia Khodaverdi, Martina Degiorgis, Karolina Fs Larsson, Andrea Bonetti, Peter Hall, Angel Guerrero-Zotano, Hans Wildiers, Andrea Gombos, Beatriz Rojas, Dario Romagnoli, Sandrine Marreaud, Caroline Duhem, Joan Albanell, Fatima Cardoso, Marija Balic, Marco Colleoni, Jorge S. Reis-Filho, Judith M. Bliss, Giuseppe Curigliano, Elsemieke D. Scheepers, Giuseppe Viale, Evandro de Azambuja, Eva M. Ciruelos, Angelo Di Leo, Justin Ndozeng, Mark E. Robson, Einav Nili Gal-Yam, Christos Sotiriou, Debora Fumagalli, Alexandre Irrthum, Mafalda Oliveira, and Philippe Aftimos
- Abstract
File with the supplementary material & methods, supplementary tables and supplementary figure.
- Published
- 2023
- Full Text
- View/download PDF
21. Supplemental File 1 from AACR Project GENIE: Powering Precision Medicine through an International Consortium
- Author
-
Hongxin Zhang, Ahmet Zehir, Emily Yu, Thomas V. Yu, Celeste Yu, Stuart Watt, Chetna Wathoo, Lucy L. Wang, Emile E. Voest, Carl Virtanen, Victor E. Velculescu, Harm van Tinteren, Tony van de Velde, Laura J. Van 'T Veer, Eliezer M. Van Allen, Stacy B. Thomas, Jelle J. ten Hoeve, Barry S. Taylor, Shawn M. Sweeney, Thomas P. Stricker, Natalie H. Stickle, Parin Sripakdeevong, Jean Charles Soria, Gabe S. Sonke, David B. Solit, Lillian L. Siu, Priyanka Shivdasani, Kenna R Mills Shaw, Nikolaus Schultz, Deborah Schrag, Charles L. Sawyers, Mark J. Routbort, Barrett J. Rollins, Brendan Reardon, Trevor J. Pugh, Ben Ho Park, John A. Orechia, Larsson Omberg, Petra M. Nederlof, Nathanael D. Moore, Gordon Mills, Clinton Miller, Christine M. Micheel, Funda Meric-Bernstam, Gerrit A. Meijer, David S. Maxwell, Ian Maurer, Laura E. MacConaill, Zhibin Lu, David Liu, James Lindsay, Neal I. Lindeman, Mia A. Levy, Eva M. Lepisto, Michele L. LeNoue-Newton, Céline Lefebvre, Marc Ladanyi, Ritika Kundra, Priti Kumari, Walter Kinyua, Cyriac Kandoth, Suzanne Kamel-Reid, David M. Hyman, Jan Hudeček, Hugo Horlings, Stephanie Hintzen, Zachary J. Heins, Justin Guinney, Benjamin E. Gross, Christopher D. Gocke, Stu Gardos, Francisco Garcia, Jianjiong Gao, P. Andrew Futreal, Matthew D. Ducar, Raymond N. DuBois, Semih Dogan, Catherine Del Vecchio Fitz, Nancy E. Davidson, Kristen K. Dang, Debyani Chakravarty, Ethan Cerami, Fabien Calvo, Mariska Bierkens, Michael F. Berger, Philippe L. Bedard, José Baselga, Alexander S. Baras, Monica Arnedos, and Fabrice André
- Abstract
AACR GENIE Data Guide
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Data from Characteristics and Outcome of AKT1E17K-Mutant Breast Cancer Defined through AACR Project GENIE, a Clinicogenomic Registry
- Author
-
David M. Hyman, Deborah Schrag, Alexia Iasonos, Philippe L. Bedard, Funda Meric-Bernstam, Mia Levy, Fabrice André, Charles L. Sawyers, Ben H. Park, Seth Sheffler-Collins, Jocelyn Lee, Stuart M. Gardos, Andrew Zarski, Nikolaus Schultz, JianJiong Gao, Shawn M. Sweeney, Ritika Kundra, Benjamin E. Gross, Jan Hudecek, Hugo Horlings, Chetna Wathoo, Christine M. Micheel, Semih Dogan, Natalie Blauvelt, Michele L. Lenoue-Newton, Michael J. Hasset, Monica Arnedos, Eva M. Lepisto, Celeste Yu, Bastien Nguyen, Qin Zhou, and Lillian M. Smyth
- Abstract
Supplementary Tables and Figures
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary file - liver genes from Genomic and Transcriptomic Analyses of Breast Cancer Primaries and Matched Metastases in AURORA, the Breast International Group (BIG) Molecular Screening Initiative
- Author
-
Martine J. Piccart, David Venet, Matteo Benelli, Lucy R. Yates, Florentine S. Hilbers, Carmela Caballero, Andrea Vingiani, Mariana Brandão, Maite Lasa Montoya, Nadia Harbeck, David A. Cameron, Susan Knox, Sherene Loi, Philippe L. Bedard, Oskar Th Johannsson, Nancy E. Davidson, Gabriele Zoppoli, Barbro Linderholm, Sibylle Loibl, Ethel Seyll, Marta Paoli, Ásgerdur Sverrisdóttir, Richard Greil, Silvia Khodaverdi, Martina Degiorgis, Karolina Fs Larsson, Andrea Bonetti, Peter Hall, Angel Guerrero-Zotano, Hans Wildiers, Andrea Gombos, Beatriz Rojas, Dario Romagnoli, Sandrine Marreaud, Caroline Duhem, Joan Albanell, Fatima Cardoso, Marija Balic, Marco Colleoni, Jorge S. Reis-Filho, Judith M. Bliss, Giuseppe Curigliano, Elsemieke D. Scheepers, Giuseppe Viale, Evandro de Azambuja, Eva M. Ciruelos, Angelo Di Leo, Justin Ndozeng, Mark E. Robson, Einav Nili Gal-Yam, Christos Sotiriou, Debora Fumagalli, Alexandre Irrthum, Mafalda Oliveira, and Philippe Aftimos
- Abstract
Supplementary file with the listing of the liver genes excluded from transcriptomic analyses.
- Published
- 2023
- Full Text
- View/download PDF
24. Data from Characteristics and Outcome of AKT1E17K-Mutant Breast Cancer Defined through AACR Project GENIE, a Clinicogenomic Registry
- Author
-
David M. Hyman, Deborah Schrag, Alexia Iasonos, Philippe L. Bedard, Funda Meric-Bernstam, Mia Levy, Fabrice André, Charles L. Sawyers, Ben H. Park, Seth Sheffler-Collins, Jocelyn Lee, Stuart M. Gardos, Andrew Zarski, Nikolaus Schultz, JianJiong Gao, Shawn M. Sweeney, Ritika Kundra, Benjamin E. Gross, Jan Hudecek, Hugo Horlings, Chetna Wathoo, Christine M. Micheel, Semih Dogan, Natalie Blauvelt, Michele L. Lenoue-Newton, Michael J. Hasset, Monica Arnedos, Eva M. Lepisto, Celeste Yu, Bastien Nguyen, Qin Zhou, and Lillian M. Smyth
- Abstract
AKT inhibitors have promising activity in AKT1E17K-mutant estrogen receptor (ER)–positive metastatic breast cancer, but the natural history of this rare genomic subtype remains unknown. Utilizing AACR Project GENIE, an international clinicogenomic data-sharing consortium, we conducted a comparative analysis of clinical outcomes of patients with matched AKT1E17K-mutant (n = 153) and AKT1–wild-type (n = 302) metastatic breast cancer. AKT1-mutant cases had similar adjusted overall survival (OS) compared with AKT1–wild-type controls (median OS, 24.1 vs. 29.9, respectively; P = 0.98). AKT1-mutant cases enjoyed longer durations on mTOR inhibitor therapy, an observation previously unrecognized in pivotal clinical trials due to the rarity of this alteration. Other baseline clinicopathologic features, as well as durations on other classes of therapy, were broadly similar. In summary, we demonstrate the feasibility of using a novel and publicly accessible clincogenomic registry to define outcomes in a rare genomically defined cancer subtype, an approach with broad applicability to precision oncology.Significance:We delineate the natural history of a rare genomically distinct cancer, AKT1E17K-mutant ER-positive breast cancer, using a publicly accessible registry of real-world patient data, thereby illustrating the potential to inform drug registration through synthetic control data.See related commentary by Castellanos and Baxi, p. 490.
- Published
- 2023
- Full Text
- View/download PDF
25. Tumor-Naïve Circulating Tumor DNA as an Early Response Biomarker for Patients Treated With Immunotherapy in Early Phase Clinical Trials
- Author
-
Enrique Sanz-Garcia, Sofia Genta, Xiaoxi Chen, Qiuxiang Ou, Daniel V. Araujo, Albiruni R. Abdul Razak, Aaron R. Hansen, Anna Spreafico, Hua Bao, Xue Wu, Lillian L. Siu, and Philippe L. Bedard
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE To evaluate early circulating tumor DNA (ctDNA) kinetics using a tumor-naïve assay and correlate it with clinical outcomes in early phase immunotherapy (IO) trials. METHODS Plasma samples were analyzed using a 425-gene next-generation sequencing panel at baseline and before cycle 2 (3-4 weeks) in patients with advanced solid tumors treated with investigational IO agents. Variant allele frequency (VAF) for mutations in each gene, mean VAF (mVAF) from all mutations, and change in mVAF between both time points were calculated. Hyperprogression (HyperPD) was measured using Matos and Caramella criteria. RESULTS A total of 162 plasma samples were collected from 81 patients with 27 different tumor types. Patients were treated in 37 different IO phase I/II trials, 72% of which involved a PD-1/PD-L1 inhibitor. ctDNA was detected in 122 plasma samples (75.3%). A decrease in mVAF from baseline to precycle 2 was observed in 24 patients (37.5%) and was associated with longer progression-free survival (hazard ratio [HR], 0.43; 95% CI, 0.24 to 0.77; P < .01) and overall survival (HR, 0.54; 95% CI, 0.3 to 0.96; P = .03) compared with an increase. These differences were more marked if there was a >50% decrease in mVAF for both progression-free survival (HR, 0.29; 95% CI, 0.13 to 0.62; P < .001) and overall survival (HR, 0.23; 95% CI, 0.09 to 0.6; P = .001). No differences in mVAF changes were observed between the HyperPD and progressive disease patients. CONCLUSION A decrease in ctDNA within 4 weeks of treatment was associated with treatment outcomes in patients in early phase IO trials. Tumor-naïve ctDNA assays may be useful for identifying early treatment benefits in phase I/II IO trials.
- Published
- 2023
- Full Text
- View/download PDF
26. A phase III trial of alpelisib + trastuzumab ± fulvestrant versus trastuzumab + chemotherapy in HER2+ PIK3CA-mutated breast cancer
- Author
-
Jose Alejandro Pérez-Fidalgo, Carmen Criscitiello, Eva Carrasco, Meredith M Regan, Angelo Di Leo, Karin Ribi, Virginie Adam, and Philippe L Bedard
- Subjects
Cancer Research ,Oncology ,General Medicine ,skin and connective tissue diseases ,neoplasms - Abstract
ALPHABET is a randomized phase III trial assessing alpelisib + trastuzumab with or without fulvestrant in previously treated HER2-positive PIK3CA-mutated advanced breast cancer. Patients will be included in two cohorts according to hormone receptor (HR) status. In the experimental arms, patients in the HR-negative cohort will receive trastuzumab + alpelisib, and patients in the HR-positive cohort will receive the same treatment plus fulvestrant. Patients in the control arms will receive trastuzumab + physician’s choice chemotherapy (eribuline, capecitabine or vinorelbine). Key eligibility criteria include 1–4 previous lines of anti-HER2 therapy and prior trastuzumab emtansine. The primary end point is investigator-assessed progression-free survival. The study aims to recruit a total of 300 patients.
- Published
- 2022
- Full Text
- View/download PDF
27. Molecular profiling of advanced solid tumors and patient outcomes with genotype-matched clinical trials: the Princess Margaret IMPACT/COMPACT trial
- Author
-
Tracy L. Stockley, Amit M. Oza, Hal K. Berman, Natasha B. Leighl, Jennifer J. Knox, Frances A. Shepherd, Eric X. Chen, Monika K. Krzyzanowska, Neesha Dhani, Anthony M. Joshua, Ming-Sound Tsao, Stefano Serra, Blaise Clarke, Michael H. Roehrl, Tong Zhang, Mahadeo A. Sukhai, Nadia Califaretti, Mateya Trinkaus, Patricia Shaw, Theodorus van der Kwast, Lisa Wang, Carl Virtanen, Raymond H. Kim, Albiruni R. A. Razak, Aaron R. Hansen, Celeste Yu, Trevor J. Pugh, Suzanne Kamel-Reid, Lillian L. Siu, and Philippe L. Bedard
- Subjects
Molecular profiling ,DNA sequencing ,Clinical trials ,Solid tumors ,Precision medicine ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background The clinical utility of molecular profiling of tumor tissue to guide treatment of patients with advanced solid tumors is unknown. Our objectives were to evaluate the frequency of genomic alterations, clinical “actionability” of somatic variants, enrollment in mutation-targeted or other clinical trials, and outcome of molecular profiling for advanced solid tumor patients at the Princess Margaret Cancer Centre (PM). Methods Patients with advanced solid tumors aged ≥18 years, good performance status, and archival tumor tissue available were prospectively consented. DNA from archival formalin-fixed paraffin-embedded tumor tissue was tested using a MALDI-TOF MS hotspot panel or a targeted next generation sequencing (NGS) panel. Somatic variants were classified according to clinical actionability and an annotated report included in the electronic medical record. Oncologists were provided with summary tables of their patients’ molecular profiling results and available mutation-specific clinical trials. Enrolment in genotype-matched versus genotype-unmatched clinical trials following release of profiling results and response by RECIST v1.1 criteria were evaluated. Results From March 2012 to July 2014, 1893 patients were enrolled and 1640 tested. After a median follow-up of 18 months, 245 patients (15 %) who were tested were subsequently treated on 277 therapeutic clinical trials, including 84 patients (5 %) on 89 genotype-matched trials. The overall response rate was higher in patients treated on genotype-matched trials (19 %) compared with genotype-unmatched trials (9 %; p
- Published
- 2016
- Full Text
- View/download PDF
28. Abstract P2-13-07: Zanidatamab (ZW25), a HER2-targeted bispecific antibody, in combination with chemotherapy (chemo) for HER2-positive breast cancer (BC): Results from a phase 1 study
- Author
-
Philippe L Bedard, Seock-Ah Im, Elena Elimova, Sun Young Rha, Rachel Goodwin, Cristiano Ferrario, Keun-Wook Lee, Diana Hanna, Funda Meric-Bernstam, Jose Mayordomo, Murali Beeram, Erika Hamilton, Jorge Chaves, Melody Cobleigh, Tony Mwatha, Joseph Woolery, and Do-Youn Oh
- Subjects
Cancer Research ,Oncology - Abstract
Background: Despite the availability of multiple HER2-targeted therapies, most patients (pts) with advanced HER2-positive BC experience disease progression, and an unmet medical need remains. Zanidatamab (zani) binds to HER2 across a range of expression levels and induces formation of receptor clusters, resulting in receptor internalization and downregulation that affect signal transduction. Zani potently activates antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement-dependent cytotoxicity. In this ongoing phase 1 study (NCT02892123), data from BC pts treated with zani monotherapy have been previously reported. Here, we evaluated the safety and antitumor activity of zani in combination with chemo in HER2-positive BC pts. Methods: Pts with locally advanced and/or metastatic HER2-positive (immunohistochemistry [IHC] 3+ or IHC 2+/fluorescence in situ hybridization+) BC who received prior trastuzumab (tras), pertuzumab (pert), and T-DM1 were enrolled. Eligibility criteria included Eastern Cooperative Oncology Group performance status ≤1 and adequate organ function. Prior history of treated stable brain metastases was allowed. Zani (20 mg/kg Q2W or 30 mg/kg Q3W) was administered in combination with paclitaxel (pac), capecitabine (cap), or vinorelbine (vin). Zani + chemo was continued until disease progression or toxicity; if chemo was discontinued due to toxicity, zani treatment could be continued. Primary endpoints were safety and tolerability assessments; secondary efficacy endpoints included objective response rate (ORR) and disease control rate (DCR; complete response [CR] + partial response [PR] + stable disease [SD]) per RECIST v1.1, and progression-free survival (PFS). Clinical benefit rate (CBR) was defined as SD ≥24 weeks or best overall response of CR or PR. Results: As of 3 May 2021, 20 HER2-positive BC pts had enrolled (zani + pac, n=4; zani + cap, n=9; zani + vin, n=7). Ten pts were hormone receptor positive (data available for 19 pts), and 7 pts had a prior history of brain metastases. Median age was 53 years (range, 38-72), with median prior systemic therapies of 4.5 (range, 2-7) and median of 2 regimens in the metastatic setting. Pts received a median of 3 prior HER2 regimens; all pts received tras and T-DM1, 17 received pert, and 6 received tyrosine kinase inhibitors. In the 16 response evaluable (measurable disease and either post-baseline assessments or earlier death/progression) pts, confirmed ORR was 37.5% (with 1 additional response pending confirmation), CBR was 50%, and DCR was 81.3%. Median duration of response was not reached (range, 1.8+ to 16.8+ mo), with 4 of 6 confirmed responses ongoing. Among all pts (n=20), the most common (≥30% of pts) zani- and/or chemo-related adverse events (AEs) were diarrhea (65%), nausea (45%), peripheral neuropathy (35%), and fatigue (30%). One pt experienced grade 3 diarrhea leading to dose reduction of zani, and 1 pt discontinued zani due to AEs (abdominal pain and nausea, both grade ≤2). Four grade 4 AEs were observed in 4 (20%) pts: neutrophil count decreased (2 pts) and neutropenia (1 pt), all related to chemo, and hyponatremia (1 pt, not related to study treatment). Serious AEs were observed in 2 (10%) pts (pleural effusion, 1 pt; pneumonitis and upper respiratory tract infection, 1 pt), none related to zani. No grade 5 AEs were reported. Conclusions: Zani in combination with chemo is well tolerated, with encouraging and durable antitumor activity in heavily pretreated (including prior tras, pert, and T-DM1) pts with HER2-positive BC. These data support further investigation of zani as a novel therapeutic for these pts. The cap and vin cohorts continue to enroll, and additional cohorts are evaluating zani ± chemo in combination with tucatinib. Citation Format: Philippe L Bedard, Seock-Ah Im, Elena Elimova, Sun Young Rha, Rachel Goodwin, Cristiano Ferrario, Keun-Wook Lee, Diana Hanna, Funda Meric-Bernstam, Jose Mayordomo, Murali Beeram, Erika Hamilton, Jorge Chaves, Melody Cobleigh, Tony Mwatha, Joseph Woolery, Do-Youn Oh. Zanidatamab (ZW25), a HER2-targeted bispecific antibody, in combination with chemotherapy (chemo) for HER2-positive breast cancer (BC): Results from a phase 1 study [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-13-07.
- Published
- 2022
- Full Text
- View/download PDF
29. Data from Phase I/Ib Clinical Trial of Sabatolimab, an Anti–TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti–PD-1 Antibody, in Advanced Solid Tumors
- Author
-
Aung Naing, Luigi Manenti, Sabine Gutzwiller, Haiying Sun, Alexandros Xyrafas, Tyler A. Longmire, Catherine A. Sabatos-Peyton, Armando Santoro, Sofie Wilgenhof, F. Stephen Hodi, Chia-Chi Lin, Philippe L. Bedard, John Sarantopoulos, Patrick M. Forde, David Tai, Toshihiko Doi, Nicolas Mach, Hans Gelderblom, and Giuseppe Curigliano
- Abstract
Purpose:Sabatolimab (MBG453) and spartalizumab are mAbs that bind T-cell immunoglobulin domain and mucin domain-3 (TIM-3) and programmed death-1 (PD-1), respectively. This phase I/II study evaluated the safety and efficacy of sabatolimab, with or without spartalizumab, in patients with advanced solid tumors.Patients and Methods:Primary objectives of the phase I/Ib part were to characterize the safety and estimate recommended phase II dose (RP2D) for future studies. Dose escalation was guided by a Bayesian (hierarchical) logistic regression model. Sabatolimab was administered intravenously, 20 to 1,200 mg, every 2 or 4 weeks (Q2W or Q4W). Spartalizumab was administered intravenously, 80 to 400 mg, Q2W or Q4W.Results:Enrolled patients (n = 219) had a range of cancers, most commonly ovarian (17%) and colorectal cancer (7%); patients received sabatolimab (n = 133) or sabatolimab plus spartalizumab (n = 86). The MTD was not reached. The most common adverse event suspected to be treatment-related was fatigue (9%, sabatolimab; 15%, combination). No responses were seen with sabatolimab. Five patients receiving combination treatment had partial responses (6%; lasting 12–27 months) in colorectal cancer (n = 2), non–small cell lung cancer (NSCLC), malignant perianal melanoma, and SCLC. Of the five, two patients had elevated expression of immune markers in baseline biopsies; another three had >10% TIM-3–positive staining, including one patient with NSCLC who received prior PD-1 therapy.Conclusions:Sabatolimab plus spartalizumab was well tolerated and showed preliminary signs of antitumor activity. The RP2D for sabatolimab was selected as 800 mg Q4W (alternatively Q3W or Q2W schedules, based on modeling), with or without 400 mg spartalizumab Q4W.
- Published
- 2023
- Full Text
- View/download PDF
30. Supplementary Data from Phase I/Ib Clinical Trial of Sabatolimab, an Anti–TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti–PD-1 Antibody, in Advanced Solid Tumors
- Author
-
Aung Naing, Luigi Manenti, Sabine Gutzwiller, Haiying Sun, Alexandros Xyrafas, Tyler A. Longmire, Catherine A. Sabatos-Peyton, Armando Santoro, Sofie Wilgenhof, F. Stephen Hodi, Chia-Chi Lin, Philippe L. Bedard, John Sarantopoulos, Patrick M. Forde, David Tai, Toshihiko Doi, Nicolas Mach, Hans Gelderblom, and Giuseppe Curigliano
- Abstract
Supplementary Materials
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Table S4. from Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
José Baselga, Jerry Y. Hsu, Michael C. Wei, Thomas J. Stout, Timothy R. Wilson, Na Cui, Alison K. Cardenas, Mafalda Oliveira, Lajos Pusztai, Manish R. Patel, Philippe L. Bedard, Andrés Cervantes, Ian E. Krop, Donald A. Richards, Cristina Saura, and Maura N. Dickler
- Abstract
Exposure to taselisib
- Published
- 2023
- Full Text
- View/download PDF
32. Data from A Phase I First-in-Human Study of Nesvacumab (REGN910), a Fully Human Anti–Angiopoietin-2 (Ang2) Monoclonal Antibody, in Patients with Advanced Solid Tumors
- Author
-
Lillian L. Siu, A. Thomas DiCioccio, Bo Gao, Pamela A. Trail, Ana Kostic, Israel Lowy, Carrie M. Brownstein, Lieve Adriaens, Muralidhar Beeram, Ariceli A. Alfaro, Philippe L. Bedard, Amita Patnaik, Katherine Van Loon, Albiruni R. Abdul Razak, Anthony W. Tolcher, Robin Kate Kelley, and Kyriakos P. Papadopoulos
- Abstract
Purpose: Nesvacumab (REGN910) is a fully human immunoglobulin G1 (IgG1) monoclonal antibody that specifically binds and inactivates the Tie2 receptor ligand Ang2 with high affinity, but shows no binding to Ang1. The main objectives of this trial were to determine the safety, tolerability, dose-limiting toxicities (DLT), and recommended phase II dose (RP2D) of nesvacumab.Experimental Design: Nesvacumab was administered intravenously every two weeks with dose escalations from 1 to 20 mg/kg in patients with advanced solid tumors.Results: A total of 47 patients were treated with nesvacumab. No patients in the dose escalation phase experienced DLTs, therefore a maximum tolerated dose (MTD) was not reached. The most common nesvacumab-related adverse events were fatigue (23.4%), peripheral edema (21.3%), decreased appetite, and diarrhea (each 10.6%; all grade ≤ 2). Nesvacumab was characterized by linear kinetics and had a terminal half-life of 6.35 to 9.66 days in a dose-independent manner. Best response by RECIST 1.1 in 43 evaluable patients included 1 partial response (adrenocortical carcinoma) of 24 weeks duration. Two patients with hepatocellular carcinoma had stable disease (SD) > 16 weeks, with tumor regression and >50% decrease in α-fetoprotein. Analyses of putative angiogenesis biomarkers in serum and tumor biopsies were uninformative for treatment duration.Conclusions: Nesvacumab safety profile was acceptable at all dose levels tested. Preliminary antitumor activity was observed in patients with treatment-refractory advanced solid tumors. On the basis of cumulative safety, antitumor activity, pharmacokinetic and pharmacodynamic data, the 20 mg/kg dose was determined to be the RP2D. Clin Cancer Res; 22(6); 1348–55. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Figure 2 from Elucidating Prognosis and Biology of Breast Cancer Arising in Young Women Using Gene Expression Profiling
- Author
-
Sherene Loi, Christos Sotiriou, Martine J. Piccart, Benjamin Haibe-Kains, Michail Ignatiadis, Carmen Criscitiello, Sandeep K. Singhal, Philippe L. Bedard, Stefan Michiels, and Hatem A. Azim
- Abstract
PDF file - 78K
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Tables from Phase I Basket Study of Taselisib, an Isoform-Selective PI3K Inhibitor, in Patients with PIK3CA-Mutant Cancers
- Author
-
David M. Hyman, Michael C. Wei, Timothy R. Wilson, Maurizio Scaltriti, Heidi M. Savage, Surai Jones, John Bond, Helen Won, Lara Dunn, Jasgit C. Sachdev, Philippe L. Bedard, Raid Aljumaily, Cynthia X. Ma, Vincent Ribrag, Manish R. Patel, Anton Melnyk, Valentina Gambardella, Cristina Saura, Dejan Juric, Matthew T. Chang, and Komal Jhaveri
- Abstract
Supplementary Tables
- Published
- 2023
- Full Text
- View/download PDF
35. Supplementary Figure 1 from Elucidating Prognosis and Biology of Breast Cancer Arising in Young Women Using Gene Expression Profiling
- Author
-
Sherene Loi, Christos Sotiriou, Martine J. Piccart, Benjamin Haibe-Kains, Michail Ignatiadis, Carmen Criscitiello, Sandeep K. Singhal, Philippe L. Bedard, Stefan Michiels, and Hatem A. Azim
- Abstract
PDF file - 92K
- Published
- 2023
- Full Text
- View/download PDF
36. Table S1 from Further Investigation of the Role of ACYP2 and WFS1 Pharmacogenomic Variants in the Development of Cisplatin-Induced Ototoxicity in Testicular Cancer Patients
- Author
-
Colin J.D. Ross, Bruce C. Carleton, Karen A. Gelmon, Michael R. Hayden, Philippe L. Bedard, Christian K. Kollmannsberger, Daniel J. Renouf, Geoffrey Liu, Galen E.B. Wright, José G. Monzon, Carol Critchley, Beth Brooks, and Britt I. Drögemöller
- Abstract
Table S1 provides a summary of the genetic association results in the European only cohort
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Methods from Phase I Basket Study of Taselisib, an Isoform-Selective PI3K Inhibitor, in Patients with PIK3CA-Mutant Cancers
- Author
-
David M. Hyman, Michael C. Wei, Timothy R. Wilson, Maurizio Scaltriti, Heidi M. Savage, Surai Jones, John Bond, Helen Won, Lara Dunn, Jasgit C. Sachdev, Philippe L. Bedard, Raid Aljumaily, Cynthia X. Ma, Vincent Ribrag, Manish R. Patel, Anton Melnyk, Valentina Gambardella, Cristina Saura, Dejan Juric, Matthew T. Chang, and Komal Jhaveri
- Abstract
Supplementary Methods
- Published
- 2023
- Full Text
- View/download PDF
38. redacted protocol from Safety and Antitumor Activity of Pembrolizumab in Patients with Estrogen Receptor–Positive/Human Epidermal Growth Factor Receptor 2–Negative Advanced Breast Cancer
- Author
-
Antoinette R. Tan, Vassiliki Karantza, Dina Pietrangelo, Sanatan Saraf, Sherene Loi, Roberto Salgado, Andrea Varga, Emilie M.J. van Brummelen, Christophe Le Tourneau, Jasgit Sachdev, Philippe L. Bedard, Sarina A. Piha-Paul, Patrick A. Ott, Seock-Ah Im, Jean-Pierre Delord, and Hope S. Rugo
- Abstract
redacted protocol
- Published
- 2023
- Full Text
- View/download PDF
39. Supplementary Figures S1, S2, S3 and Tables S1, S2 from Neratinib Efficacy and Circulating Tumor DNA Detection of HER2 Mutations in HER2 Nonamplified Metastatic Breast Cancer
- Author
-
Matthew J.C. Ellis, Hussam Al-Kateb, Philippe L. Bedard, Kimberly Blackwell, Mark Pegram, Daniel F. Hayes, John Pfeifer, Alshad S. Lalani, Richard Bryce, Richard B. Lanman, Kimberly C. Banks, Caroline Bumb, Jill Anderson, Shana Thomas, Polly Ann Niravath, Carey Anders, Timothy J. Pluard, Andres Forero, Melody Cobleigh, Debu Tripathy, Christy Russell, Matthew P. Goetz, Michael Naughton, Eric Winer, Gretchen Kimmick, Melinda L. Telli, Rachel A. Freedman, Feng Gao, Ron Bose, and Cynthia X. Ma
- Abstract
Supplementary Figures S1, S2, S3 and Tables S1, S2
- Published
- 2023
- Full Text
- View/download PDF
40. Supplementary Figures 1-9 from A Phase I First-in-Human Study of Nesvacumab (REGN910), a Fully Human Anti–Angiopoietin-2 (Ang2) Monoclonal Antibody, in Patients with Advanced Solid Tumors
- Author
-
Lillian L. Siu, A. Thomas DiCioccio, Bo Gao, Pamela A. Trail, Ana Kostic, Israel Lowy, Carrie M. Brownstein, Lieve Adriaens, Muralidhar Beeram, Ariceli A. Alfaro, Philippe L. Bedard, Amita Patnaik, Katherine Van Loon, Albiruni R. Abdul Razak, Anthony W. Tolcher, Robin Kate Kelley, and Kyriakos P. Papadopoulos
- Abstract
Supplementary Figure 1 Total Nesvacumab (REGN910) and Total Ang2 vs Time (cycle 1) following IV infusion in Patients. Supplementary Figure 2 Serum levels of ESM1, SDF-1, PLGF-1 and sVCAM-1 following Nesvacumab (REGN910) treatment. Supplementary Figure 3 Correlative analysis of baseline serum Ang2, ESM-1, SDF-1 ,PIGF and cVCAM-1 and treatment duration. Supplementary Figure 4 Correlative analysis of baseline Ang2 in tumor vessels and treatment duration/response. Supplementary Figure 5 Ang2 and VEGF-A protein levels in tumor tissue pre and post treatment with Nesvacumab (REGN910). Supplementary Figure 6 Biomarker analysis of archival tumor tissue. Supplementary Figure 7 Microvascular density in tumor tissue pre and post treatment with Nesvacumab (REGN910). Supplementary Figure 8 Tumor cell proliferation in tumor tissue pre and post treatment with Nesvacumab (REGN910). Supplementary Figure 9 TIE-2 protein levels associated with tumor vessels pre and post treatment with Nesvacumab (REGN910)
- Published
- 2023
- Full Text
- View/download PDF
41. Figure S4 from Further Investigation of the Role of ACYP2 and WFS1 Pharmacogenomic Variants in the Development of Cisplatin-Induced Ototoxicity in Testicular Cancer Patients
- Author
-
Colin J.D. Ross, Bruce C. Carleton, Karen A. Gelmon, Michael R. Hayden, Philippe L. Bedard, Christian K. Kollmannsberger, Daniel J. Renouf, Geoffrey Liu, Galen E.B. Wright, José G. Monzon, Carol Critchley, Beth Brooks, and Britt I. Drögemöller
- Abstract
Figure S4 provides examples of audiogram configurations
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figure S1. from Phase II Study of Taselisib (GDC-0032) in Combination with Fulvestrant in Patients with HER2-Negative, Hormone Receptor–Positive Advanced Breast Cancer
- Author
-
José Baselga, Jerry Y. Hsu, Michael C. Wei, Thomas J. Stout, Timothy R. Wilson, Na Cui, Alison K. Cardenas, Mafalda Oliveira, Lajos Pusztai, Manish R. Patel, Philippe L. Bedard, Andrés Cervantes, Ian E. Krop, Donald A. Richards, Cristina Saura, and Maura N. Dickler
- Abstract
Participant PIK3CA-mutation central testing results
- Published
- 2023
- Full Text
- View/download PDF
43. Supplementary Table 1 from A Phase I First-in-Human Study of Nesvacumab (REGN910), a Fully Human Anti–Angiopoietin-2 (Ang2) Monoclonal Antibody, in Patients with Advanced Solid Tumors
- Author
-
Lillian L. Siu, A. Thomas DiCioccio, Bo Gao, Pamela A. Trail, Ana Kostic, Israel Lowy, Carrie M. Brownstein, Lieve Adriaens, Muralidhar Beeram, Ariceli A. Alfaro, Philippe L. Bedard, Amita Patnaik, Katherine Van Loon, Albiruni R. Abdul Razak, Anthony W. Tolcher, Robin Kate Kelley, and Kyriakos P. Papadopoulos
- Abstract
Supplementary Table 1. Summary of the Most Common (>10%) Treatment-Emergent Adverse Events
- Published
- 2023
- Full Text
- View/download PDF
44. Supplementary Figure 3 from Elucidating Prognosis and Biology of Breast Cancer Arising in Young Women Using Gene Expression Profiling
- Author
-
Sherene Loi, Christos Sotiriou, Martine J. Piccart, Benjamin Haibe-Kains, Michail Ignatiadis, Carmen Criscitiello, Sandeep K. Singhal, Philippe L. Bedard, Stefan Michiels, and Hatem A. Azim
- Abstract
PDF file - 53K
- Published
- 2023
- Full Text
- View/download PDF
45. Data from Neratinib Efficacy and Circulating Tumor DNA Detection of HER2 Mutations in HER2 Nonamplified Metastatic Breast Cancer
- Author
-
Matthew J.C. Ellis, Hussam Al-Kateb, Philippe L. Bedard, Kimberly Blackwell, Mark Pegram, Daniel F. Hayes, John Pfeifer, Alshad S. Lalani, Richard Bryce, Richard B. Lanman, Kimberly C. Banks, Caroline Bumb, Jill Anderson, Shana Thomas, Polly Ann Niravath, Carey Anders, Timothy J. Pluard, Andres Forero, Melody Cobleigh, Debu Tripathy, Christy Russell, Matthew P. Goetz, Michael Naughton, Eric Winer, Gretchen Kimmick, Melinda L. Telli, Rachel A. Freedman, Feng Gao, Ron Bose, and Cynthia X. Ma
- Abstract
Purpose: Based on promising preclinical data, we conducted a single-arm phase II trial to assess the clinical benefit rate (CBR) of neratinib, defined as complete/partial response (CR/PR) or stable disease (SD) ≥24 weeks, in HER2mut nonamplified metastatic breast cancer (MBC). Secondary endpoints included progression-free survival (PFS), toxicity, and circulating tumor DNA (ctDNA) HER2mut detection.Experimental Design: Tumor tissue positive for HER2mut was required for eligibility. Neratinib was administered 240 mg daily with prophylactic loperamide. ctDNA sequencing was performed retrospectively for 54 patients (14 positive and 40 negative for tumor HER2mut).Results: Nine of 381 tumors (2.4%) sequenced centrally harbored HER2mut (lobular 7.8% vs. ductal 1.6%; P = 0.026). Thirteen additional HER2mut cases were identified locally. Twenty-one of these 22 HER2mut cases were estrogen receptor positive. Sixteen patients [median age 58 (31–74) years and three (2–10) prior metastatic regimens] received neratinib. The CBR was 31% [90% confidence interval (CI), 13%–55%], including one CR, one PR, and three SD ≥24 weeks. Median PFS was 16 (90% CI, 8–31) weeks. Diarrhea (grade 2, 44%; grade 3, 25%) was the most common adverse event. Baseline ctDNA sequencing identified the same HER2mut in 11 of 14 tumor-positive cases (sensitivity, 79%; 90% CI, 53%–94%) and correctly assigned 32 of 32 informative negative cases (specificity, 100%; 90% CI, 91%–100%). In addition, ctDNA HER2mut variant allele frequency decreased in nine of 11 paired samples at week 4, followed by an increase upon progression.Conclusions: Neratinib is active in HER2mut, nonamplified MBC. ctDNA sequencing offers a noninvasive strategy to identify patients with HER2mut cancers for clinical trial participation. Clin Cancer Res; 23(19); 5687–95. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
46. Supplementary Figure 7 from Elucidating Prognosis and Biology of Breast Cancer Arising in Young Women Using Gene Expression Profiling
- Author
-
Sherene Loi, Christos Sotiriou, Martine J. Piccart, Benjamin Haibe-Kains, Michail Ignatiadis, Carmen Criscitiello, Sandeep K. Singhal, Philippe L. Bedard, Stefan Michiels, and Hatem A. Azim
- Abstract
PDF file - 89K
- Published
- 2023
- Full Text
- View/download PDF
47. Supplementary Figure 4 from Elucidating Prognosis and Biology of Breast Cancer Arising in Young Women Using Gene Expression Profiling
- Author
-
Sherene Loi, Christos Sotiriou, Martine J. Piccart, Benjamin Haibe-Kains, Michail Ignatiadis, Carmen Criscitiello, Sandeep K. Singhal, Philippe L. Bedard, Stefan Michiels, and Hatem A. Azim
- Abstract
PDF file - 118K
- Published
- 2023
- Full Text
- View/download PDF
48. Data from Natural History and Characteristics of ERBB2-mutated Hormone Receptor–positive Metastatic Breast Cancer: A Multi-institutional Retrospective Case–control Study from AACR Project GENIE
- Author
-
Monica Arnedos, Christine M. Micheel, Fei Ye, Alshad S. Lalani, Grace Mann, Feng Xu, Lisa D. Eli, Mia Levy, Chetna Wathoo, Celeste Yu, Semih Dogan, Lillian Smyth, Fabrice Andre, Eva M. Lepisto, Deborah Schrag, Rinaa S. Punglia, Funda Meric-Bernstam, Philippe L. Bedard, Natalie Blauvelt, David M. Hyman, Thomas Stricker, Sheau-Chiann Chen, and Michele L. LeNoue-Newton
- Abstract
Purpose:We wanted to determine the prognosis and the phenotypic characteristics of hormone receptor–positive advanced breast cancer tumors harboring an ERBB2 mutation in the absence of a HER2 amplification.Experimental Design:We retrospectively collected information from the American Association of Cancer Research-Genomics Evidence Neoplasia Information Exchange registry database from patients with hormone receptor–positive, HER2-negative, ERBB2-mutated advanced breast cancer. Phenotypic and co-mutational features, as well as response to treatment and outcome were compared with matched control cases ERBB2 wild type.Results:A total of 45 ERBB2-mutant cases were identified for 90 matched controls. The presence of an ERBB2 mutation was not associated with worse outcome determined by overall survival (OS) from first metastatic relapse. No significant differences were observed in phenotypic characteristics apart from higher lobular infiltrating subtype in the ERBB2-mutated group. ERBB2 mutation did not seem to have an impact in response to treatment or time-to-progression (TTP) to endocrine therapy compared with ERBB2 wild type. In the co-mutational analyses, CDH1 mutation was more frequent in the ERBB2-mutated group (FDR < 1). Although not significant, fewer co-occurring ESR1 mutations and more KRAS mutations were identified in the ERBB2-mutated group.Conclusions:ERBB2-activating mutation was not associated with a worse OS from time of first metastatic relapse, or differences in TTP on treatment as compared with a series of matched controls. Although not significant, differences in coexisting mutations (CDH1, ESR1, and KRAS) were noted between the ERBB2-mutated and the control group.
- Published
- 2023
- Full Text
- View/download PDF
49. Table S1, Table S2; Figure S1, Figure S2, Figure S3, Figure S4, Figure S5 from Lucitanib for the Treatment of HR+/HER2− Metastatic Breast Cancer: Results from the Multicohort Phase II FINESSE Study
- Author
-
Giuseppe Curigliano, Fabrice Andre, Nicholas C. Turner, Marie-Jeanne Pierrat, Sherene Loi, Sibylle Loibl, Theodora Goulioti, Malou Vicente, Elsemieke D. Scheepers, Laura Xuereb, Philippe L. Bedard, Philippe Aftimos, José Perez-Garcia, Amal Arahmani, Fergus Daly, Matteo Lambertini, Debora Fumagalli, Hatem A. Azim, Camille Poirot, Christine Campbell, Javier Cortes, Alex Pearson, and Rina Hui
- Abstract
Supplementary Table S1. Treatment dose, duration, interruption and reduction. Supplementary Table S2A. Performance of the FGF23 ELISA, Kainos Laboratories Inc., cat# CY4000. Supplementary Table S2B. Concentration range of FGF23 in serum and plasma samples. Supplementary Table S2C. ddPCR assays used for determination of FGFR1 CNV. Supplementary Figure S1. A) FINESSE study design. B) Supplementary Figure S1B. FINESSE CONSORT diagram. Supplementary Figure S2. FISH screening results for FGFR1, CCND1/11q and FGF3,4,19. All samples were screened for both FGFR1 and CCND1/11q. Only samples positive for CCND1/11q amplification were assessed for FGF3,4,19 amplification. Of these, 1 patient was not assessed for FGF3,4,19 at the request of the site. 3 patients assessed as FGF3,4,19 amplified by additional evalution criteria to those described in the Methods section, with >50% of tumour cells showing {greater than or equal to}5 gene signals/nucleus. Supplementary Figure S3. Progression-free survival by A) FGFR1 amplification status by FISH and B) FGFR1 expression by IHC. Supplementary Figure S4. Expression of endothelial FGF2 and Ki67 presented by trial cohort. Supplementary Figure S5. Progression free survival by A) endothelial FGF2 and B) endothelial Ki67 expression.
- Published
- 2023
- Full Text
- View/download PDF
50. Supplementary Figure 6 from Elucidating Prognosis and Biology of Breast Cancer Arising in Young Women Using Gene Expression Profiling
- Author
-
Sherene Loi, Christos Sotiriou, Martine J. Piccart, Benjamin Haibe-Kains, Michail Ignatiadis, Carmen Criscitiello, Sandeep K. Singhal, Philippe L. Bedard, Stefan Michiels, and Hatem A. Azim
- Abstract
PDF file - 52K
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.