157 results on '"Fadi Braiteh"'
Search Results
2. Association of Tumor Mutational Burden and Immune Gene Expression with Response to PD-1 Blockade by Sasanlimab Across Tumor Types and Routes of Administration
- Author
-
Alison Forgie, Melissa Lynne Johnson, Jeffrey Chou, Ira Jacobs, Xiao Wang, Siwen Hu-Lieskovan, Juneko E. Grilley-Olson, Fadi Braiteh, and Vinicius Bonato
- Subjects
Cancer Research ,medicine.drug_class ,Programmed Cell Death 1 Receptor ,Gene Expression ,Monoclonal antibody ,B7-H1 Antigen ,Interferon-gamma ,Neoplasms ,Biomarkers, Tumor ,medicine ,Humans ,Pharmacology (medical) ,Original Research Article ,Receptor ,Immune Checkpoint Inhibitors ,biology ,business.industry ,Antibodies, Monoclonal ,Cell cycle ,Acquired immune system ,Oncology ,biology.protein ,Cancer research ,Biomarker (medicine) ,Immunohistochemistry ,Antibody ,business ,CD8 - Abstract
Background Sasanlimab is a monoclonal antibody that binds to the programmed cell death receptor 1 (PD-1). Anti-PD-1 monoclonal antibodies have improved patient clinical outcomes; however, not all treated patients derive clinical benefit. Further insights on potential biomarkers beyond PD-L1 expression levels would help to identify the patients most likely to respond to treatment. Objective This study evaluated tumor biopsies from patients treated with intravenous or subcutaneous sasanlimab to identify biomarkers of response and characterize pharmacodynamic activity. Methods Anti-PD-1/PD-ligand 1 (PD-L1)-naive patients with advanced solid tumors received sasanlimab intravenously at 1, 3, or 10 mg/kg every 3 weeks (n = 23) or subcutaneously at 300 mg every 4 weeks (n = 15). Best tumor percentage change from baseline was determined by RECIST. Whole-exome DNA and RNA sequencing were performed in tumor samples collected from treated patients at protocol-defined timepoints. PD-L1 and CD8 protein expression were evaluated in tumor biopsies by immunohistochemistry. Associations with response were assessed by linear regression analysis. Results Baseline tumor mutational burden (TMB), as well as PD-L1 and CD8 expression, were significantly associated with response to sasanlimab across the multiple dose levels, routes of administration, and range of tumor types evaluated. TMB is an independent biomarker from the various tumor inflammatory genes and signatures evaluated. Gene set enrichment analysis showed that higher baseline expression levels of genes related to the interferon-γ and PD-1 signaling pathways and the cell cycle were significantly associated with response to sasanlimab across tumor types. No differences were observed between routes of administration with regard to response to sasanlimab for the biomarkers of interest (TMB, PD-L1, CD8, and interferon-γ signature). Evaluation of pharmacodynamic changes showed increased tumor expression of genes enriched in adaptive immune response pathways. Conclusions Our findings indicate an active, immunomodulatory mechanism for the anti-PD-1 antibody sasanlimab across different tumor types and routes of administration. Trial Registration ClinicalTrials.gov identifier NCT02573259; registered October 2015. Supplementary Information The online version contains supplementary material available at 10.1007/s11523-021-00833-2.
- Published
- 2021
3. Safety and Clinical Activity of Atezolizumab in Patients with Metastatic Castration-Resistant Prostate Cancer: A Phase I Study
- Author
-
Yohann Loriot, Indrani Sarkar, Daniel P. Petrylak, Joseph Kim, Paul Conkling, Thomas Powles, Susheela Carroll, Lauren C. Harshman, Jean-Pierre Delord, Edward E. Kadel, Sujata Narayanan, Marcella Fassò, David R. Shaffer, Michael S. Gordon, Sanjeev Mariathasan, Kobe C. Yuen, John D. Powderly, Fadi Braiteh, and Carol O'Hear
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,medicine ,Humans ,Enzalutamide ,In patient ,Adverse effect ,Response Evaluation Criteria in Solid Tumors ,Aged ,business.industry ,Middle Aged ,medicine.disease ,Confidence interval ,Phase i study ,Survival Rate ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,business - Abstract
Purpose: Atezolizumab [anti–programmed death-ligand 1 (anti-PD-L1)] is well tolerated and efficacious in multiple cancers, but has not been previously evaluated in metastatic castration-resistant prostate cancer (mCRPC). This study examined the safety, efficacy, and biomarkers of atezolizumab monotherapy for mCRPC. Patients and Methods: This phase Ia, open-label, dose-escalation and dose-expansion study (PCD4989g) enrolled patients with mCRPC who had progressed on sipuleucel-T or enzalutamide. Atezolizumab was given intravenously every 3 weeks until confirmed disease progression or loss of clinical benefit. Prespecified endpoints included safety, efficacy, biomarker analyses, and radiographic assessments. Results: All 35 evaluable patients [median age, 68 years (range, 45–83 years)] received atezolizumab after ≥1 prior line of therapy; 62.9% of patients had received ≥3 prior lines. Treatment-related adverse events occurred in 21 patients (60.0%), with no deaths. One patient had a confirmed partial response (PR) per RECIST 1.1, and 1 patient had a PR per immune-related response criteria. The confirmed 50% PSA response rate was 8.6% (3 patients). Median overall survival (OS) was 14.7 months [95% confidence interval (CI): 5.9–not evaluable], with a 1-year OS rate of 52.3% (95% CI: 34–70); 2-year OS was 35.9% (95% CI: 13–59). Median follow-up was 13.0 months (range, 1.2–28.1 months). Biomarker analyses showed that atezolizumab activated immune responses; however, a composite biomarker failed to reveal consistent correlations with efficacy. Conclusions: Atezolizumab was generally well tolerated in patients with mCRPC, with a safety profile consistent with other tumor types. In heavily pretreated patients, atezolizumab monotherapy demonstrated evidence of disease control; however, its limited efficacy suggests a combination approach may be needed.
- Published
- 2021
4. 116 Evaluation of bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in cervical cancer: data from phase 1 and phase 2 studies
- Author
-
William Jeffery Edenfield, Laureen S. Ojalvo, Alexander I. Spira, E Calvo Aller, Andrés Cervantes, M De Miguel, Luis Paz-Ares, TW Park-Simon, Marika Rasschaert, FL Munoz, Julius Strauss, Isabelle Dussault, Fadi Braiteh, G. Jehl, James L. Gulley, Tianhong Li, and Suzanne Wendy Allan
- Subjects
Oncology ,Cervical cancer ,medicine.medical_specialty ,Bevacizumab ,biology ,business.industry ,Histology ,Pembrolizumab ,medicine.disease ,Immune checkpoint ,Internal medicine ,PD-L1 ,Toxicity ,medicine ,biology.protein ,business ,Progressive disease ,medicine.drug - Abstract
Introduction/Background* The accelerated US Food and Drug Administration approval of pembrolizumab validated the efficacy of anti-PD-(L)1 therapy for patients with recurrent/metastatic cervical cancer; however, the objective response rate (ORR) with pembrolizumab was 14.3% in patients with PD-L1–expressing tumours. Human papillomavirus infection is implicated in >95% of cervical cancers and is linked to upregulation of TGF-β signalling. Bintrafusp alfa is a first-in-class bifunctional fusion protein composed of the extracellular domain of the TGF-βRII receptor (a TGF-β ‘trap’) fused to a human immunoglobulin G1 monoclonal antibody blocking PD-L1. We report pooled safety and efficacy in patients with pretreated, immune checkpoint inhibitor-naive, recurrent/metastatic cervical cancer treated with bintrafusp alfa in phase 1 (INTR@PID 001; NCT02517398) and phase 2 (study 012; NCT03427411) studies. Methodology Patients received bintrafusp alfa 0.3-30 mg/kg (phase 1 dose escalation) or 1200 mg every 2 weeks (phase 1 dose expansion and phase 2) until progressive disease, unacceptable toxicity, or withdrawal. Treatment past progression was allowed. Primary endpoints were safety (phase 1 dose escalation) and best overall response per RECIST 1.1 (phase 1 dose expansion and phase 2). Result(s)* As of May 15, 2020 (phase 1) and December 22, 2020 (phase 2), 39 patients had received bintrafusp alfa for a median duration of 2.8 months (range, 0.5-19.3). The median follow-up to data cutoff was 35.0 months and 24.1 months for the phase 1 and phase 2 studies, respectively. All patients had received prior anticancer therapy; 16 (41.0%) had received ≥3 regimens. Confirmed ORR was 28.2% (table 1); responses occurred irrespective of Moore criteria (phase 1), tumour histology, prior bevacizumab treatment, or radiation treatment. Median overall survival was 13.4 months. No new safety signals and no treatment-related deaths were observed; side effects were manageable. Conclusion* Bintrafusp alfa had a manageable safety profile and demonstrated clinical activity in patients with heavily pretreated, immune checkpoint inhibitor-naive recurrent/metastatic cervical cancer. © 2021 American Society of Clinical Oncology, Inc. Reused with permission. This abstract was accepted and previously presented at the 2021 ASCO Annual Meeting. All rights reserved.
- Published
- 2021
5. Results from Phase I Extension Study Assessing Pexidartinib Treatment in Six Cohorts with Solid Tumors including TGCT, and Abnormal CSF1 Transcripts in TGCT
- Author
-
Paul S. Lin, Charles Peterfy, Andrew J. Wagner, John H. Healey, Mike Sterba, Gregory M. Cote, Paul Severson, Heather L. Gelhorn, Eric J. Sherman, Chao Zhang, Henry H. Hsu, Geoffrey I. Shapiro, Arun S. Singh, Sandra Tong-Starksen, Stephen P. Anthony, Bartosz Chmielowski, Fadi Braiteh, Allen Lee Cohn, Igor Puzanov, William D. Tap, Brian L. West, Vicki L. Keedy, Xin Ye, and Zev A. Wainberg
- Subjects
Oncology ,Urologic Diseases ,Adult ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Oncology and Carcinogenesis ,Pexidartinib ,Aminopyridines ,Giant Cell Tumor of Tendon Sheath ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,Text mining ,Rare Diseases ,Pharmacokinetics ,Clinical Research ,Internal medicine ,Genetics ,Medicine ,Humans ,Pyrroles ,Oncology & Carcinogenesis ,Receptor ,Adverse effect ,Protein Kinase Inhibitors ,Cancer ,biology ,business.industry ,Toxicity ,biology.protein ,business - Abstract
Purpose: To assess the response to pexidartinib treatment in six cohorts of adult patients with advanced, incurable solid tumors associated with colony-stimulating factor 1 receptor (CSF1R) and/or KIT proto-oncogene receptor tyrosine kinase activity. Patients and Methods: From this two-part phase I, multicenter study, pexidartinib, a small-molecule tyrosine kinase inhibitor that targets CSF1R, KIT, and FMS-like tyrosine kinase 3 (FLT3), was evaluated in six adult patient cohorts (part 2, extension) with advanced solid tumors associated with dysregulated CSF1R. Adverse events, pharmacokinetics, and tumor responses were assessed for all patients; patients with tenosynovial giant cell tumor (TGCT) were also evaluated for tumor volume score (TVS) and patient-reported outcomes (PRO). CSF1 transcripts and gene expression were explored in TGCT biopsies. Results: Ninety-one patients were treated: TGCT patients (n = 39) had a median treatment duration of 511 days, while other solid tumor patients (n = 52) had a median treatment duration of 56 days. TGCT patients had response rates of 62% (RECIST 1.1) and 56% (TVS) for the full analysis set. PRO assessments for pain showed improvement in patient symptoms, and 76% (19/25) of TGCT tissue biopsy specimens showed evidence of abnormal CSF1 transcripts. Pexidartinib treatment of TGCT resulted in tumor regression and symptomatic benefit in most patients. Pexidartinib toxicity was manageable over the entire study. Conclusions: These results offer insight into outcome patterns in cancers whose biology suggests use of a CSF1R inhibitor. Pexidartinib results in tumor regression in TGCT patients, providing prolonged control with an acceptable safety profile.
- Published
- 2021
6. First-in-human, phase I study of PF-06647263, an anti-EFNA4 calicheamicin antibody-drug conjugate, in patients with advanced solid tumors
- Author
-
Erika Hamilton, Theresa L. Werner, Ignacio Garrido-Laguna, Christopher J. Zopf, Amy Weise, Ian E. Krop, Maysa M. Abu-Khalaf, Mani Lakshminarayanan, Steven Pirie-Shepherd, David S. Hong, Jaymes S. Holland, Raffaele Baffa, Fadi Braiteh, and Howard A. Burris
- Subjects
Cancer Research ,Antibody-drug conjugate ,medicine.medical_specialty ,business.industry ,Nausea ,medicine.disease ,Gastroenterology ,03 medical and health sciences ,chemistry.chemical_compound ,Regimen ,0302 clinical medicine ,Breast cancer ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Internal medicine ,Calicheamicin ,Toxicity ,Vomiting ,Medicine ,medicine.symptom ,business ,Ovarian cancer - Abstract
PF-06647263, a novel antibody-drug conjugate consisting of an anti-EFNA4 antibody linked to a calicheamicin payload, has shown potent antitumor activity in human xenograft tumor models, including triple-negative breast cancer (TNBC). In the dose-escalation part 1 of this multicenter, open-label, phase I study (NCT02078752), successive cohorts of patients (n, 48) with advanced solid tumors and no available standard therapy received PF-06647263 every 3 weeks (Q3W) or every week (QW), following a modified toxicity probability interval (mTPI) method (initial dosing: 0.015 mg/kg Q3W). Primary objective in part 1 was to estimate the maximum tolerated dose (MTD) and select the recommended phase 2 dose (RP2D). In part 2 (dose-expansion cohort), 12 patients with pretreated, metastatic TNBC received PF-06647263 at the RP2D to further evaluate tumor response and overall safety. PF-06647263 QW administration (n, 23) was better tolerated than the Q3W regimen (n, 25) with only 1 DLT reported (thrombocytopenia). The most common AEs with the QW regimen (fatigue, nausea, vomiting, mucosal inflammation, thrombocytopenia, and diarrhea) were mostly mild to moderate in severity. The MTD was not estimated. PF-06647263 exposures increased in a dose-related manner across the doses evaluated. The RP2D was determined to be 0.015 mg/kg QW. Six (10%) patients achieved a confirmed partial response and 22 (36.7%) patients had stable disease. No correlations were observed between tumor responses and EFNA4 expression levels. Study findings showed manageable safety and favorable PK for PF-06647263 administered QW at the RP2D, with preliminary evidence of limited antitumor activity in patients with TNBC and ovarian cancer.
- Published
- 2019
7. P47.02 EV-202: Phase 2 Study of Enfortumab Vedotin for Previously Treated Advanced Solid Tumors Including Non-Small Cell Lung Cancer
- Author
-
Xuan Li, Chunzhang Wu, T. Feinstein, Fadi Braiteh, K. Muro, Joaquina Baranda, J. Yang Bruce, and S. Gorla
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,business.industry ,Enfortumab vedotin ,Cancer research ,Medicine ,Phases of clinical research ,Non small cell ,business ,Previously treated ,Lung cancer ,medicine.disease - Published
- 2021
8. A Phase 1b Dose Escalation Trial of NC-6300 (Nanoparticle Epirubicin) in Patients with Advanced Solid Tumors or Advanced, Metastatic, or Unresectable Soft-tissue Sarcoma
- Author
-
Atsushi Osada, Juneko E. Grilley Olson, Richard F. Riedel, Fadi Braiteh, Sanjay Goel, Iulian Bobe, Warren Chow, Sant P. Chawla, and Arun S. Singh
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Neutropenia ,Side effect ,Maximum Tolerated Dose ,Population ,02 engineering and technology ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Neoplasm Metastasis ,education ,Aged ,Epirubicin ,education.field_of_study ,Endometrial stromal sarcoma ,Dose-Response Relationship, Drug ,business.industry ,Soft tissue sarcoma ,Proteins ,Sarcoma ,Middle Aged ,021001 nanoscience & nanotechnology ,medicine.disease ,Oncology ,030220 oncology & carcinogenesis ,Female ,0210 nano-technology ,business ,Febrile neutropenia ,medicine.drug - Abstract
Purpose: NC-6300 is a novel nanoparticle formulation of epirubicin that has a pH-sensitive linker conjugated to epirubicin. It exhibits selective tumor accumulation owing to enhanced permeability and retention effect. We conducted a phase 1b trial to determine MTD and recommended phase II dose (RP2D) of NC-6300 monotherapy in advanced, metastatic, or unresectable solid tumors, including soft-tissue sarcomas. Patients and Methods: This phase 1b dose-escalation trial of NC-6300 monotherapy employed a Bayesian continuous reassessment method design. NC-6300 was administered on day 1 of every 21-day cycle, with epirubicin-equivalent dose increments from 125 to 215 mg/m2. Safety, efficacy, quality of life, and pharmacokinetic profile of NC-6300 monotherapy were evaluated. Results: Twenty-nine subjects (16 male) were enrolled: 17 with soft-tissue sarcoma, one with osteosarcoma, and 11 with other solid tumors. Observed dose-limiting toxicities included thrombocytopenia, stomatitis, lung infection, and febrile neutropenia. The most common grade 3/4 adverse events were neutropenia (59%), anemia (24%), thrombocytopenia (24%), and febrile neutropenia (21%). MTD and RP2D were determined to be 185 mg/m2 and 150 mg/m2, respectively. The objective response rate in the evaluable population was 11%. Partial response was observed in angiosarcoma and endometrial stromal sarcoma. A dose-dependent increase was observed in both total and released epirubicin concentrations. Conclusions: NC-6300 was well tolerated with a manageable side effect profile, despite the MTD and RP2D being higher than conventional epirubicin doses. A signal of preliminary activity was observed in angiosarcoma. NC-6300 warrants further investigation in patients with advanced solid tumors, including sarcoma.
- Published
- 2020
9. Multi-Institutional, Prospective Clinical Utility Study Evaluating the Impact of the 92-Gene Assay (CancerTYPE ID) on Final Diagnosis and Treatment Planning in Patients With Metastatic Cancer With an Unknown or Unclear Diagnosis
- Author
-
Theresa N. Operana, Anthony R. Victorio, Sachdev P. Thomas, Brock Schroeder, Lauren E. Jacobson, Catherine A. Schnabel, and Fadi Braiteh
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,business.industry ,Improved survival ,Cancer ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Clinical history ,030220 oncology & carcinogenesis ,medicine ,Unknown primary ,In patient ,Medical diagnosis ,Intensive care medicine ,Radiation treatment planning ,business ,Routine care - Abstract
Purpose Metastatic cancers of unknown primary or with unclear diagnoses pose diagnostic and management challenges, often leading to poor outcomes. Studies of the 92-gene assay have demonstrated improved diagnostic accuracy compared with standard pathology techniques and improved survival in patients treated on the basis of assay results. The current study assessed the clinical impact of the 92-gene assay on diagnostic and treatment decisions for patients with unknown or uncertain diagnoses. Methods Patients in this prospective, multi-institutional, decision-impact study included those for whom the 92-gene assay was ordered as part of routine care. Participating physicians completed electronic case report forms that contained standardized, specialty-specific questionnaires. Data collection included patient and tumor characteristics and clinical history. The key study objective of clinical impact was calculated on the basis of changes in final diagnosis and treatment after testing. Results Data collection included 444 patients, 107 physicians (73 oncologists and 34 pathologists), and 28 sites. Molecular diagnoses from 22 different tumor types and subtypes across all cases were provided in 95.5% of patients with a reportable result (n = 397). Physicians reported that the 92-gene assay was used broadly for diagnostic dilemmas that ranged from single suspected tumor type (29%) to a differential diagnosis of two or more suspected tumor types (30%) or cancers of unknown primary (41%). Integration of 92-gene assay results led to a change in the recommended treatment in 47% of patients. Conclusion Findings from this clinical utility study demonstrate that the 92-gene assay led to a change in treatment decisions in every other patient case. These data additionally define the role of this assay in clinical practice and strongly support the consideration of molecular tumor typing in the diagnosis and treatment planning of patients with metastatic cancer with unknown or uncertain diagnosis.
- Published
- 2018
10. Signature program: a platform of basket trials
- Author
-
Todd M. Bauer, Steven Stein, Barinder P. Kang, Eric Daniel Slosberg, Julio Peguero, A. Salvado, Donald A. Berry, Fadi Braiteh, Funda Meric-Bernstam, Alexander I. Spira, Matthew H. Taylor, and Sarina Anne Piha-Paul
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Buparlisib ,Sonidegib ,tissue agnostic ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,PTEN ,biology ,Ceritinib ,business.industry ,Clinical study design ,Binimetinib ,mutations ,medicine.disease ,clinical trial design ,030104 developmental biology ,chemistry ,basket trial ,030220 oncology & carcinogenesis ,biology.protein ,Adenocarcinoma ,business ,signature ,Research Paper ,medicine.drug - Abstract
Investigating targeted therapies can be challenging due to diverse tumor mutations and slow patient accrual for clinical studies. The Signature Program is a series of 8 phase 2, agent-specific basket protocols using a rapid study start-up approach involving no predetermined study sites. Each protocol evaluated 1 agent (buparlisib, dovitinib, binimetinib, encorafenib, sonidegib, BGJ398, ceritinib, or ribociclib) in patients with solid or hematologic malignancies and an actionable mutation. The primary endpoint of each study was the clinical benefit rate (ie, complete or partial response, or stable disease) at 16 weeks. A total of 192 individual sites were opened in the United States, with a median start-up time of 3.6 weeks. The most common tumor types among the 595 treated patients were colorectal (9.2%), non-small cell lung adenocarcinoma (9.1%), and ovarian (8.4%). Frequent genetic alterations were in PIK3CA, RAS, p16, and PTEN. Overall, 30 partial or complete responses were observed with 6 compounds in 16 tumor types. The Signature Program presents a unique and successful approach for rapid signal finding across multiple tumors and allowed various agents to be evaluated in patients with rare alterations. Incorporating these program features in conventional studies could lead to improved trial efficiencies and patient outcomes.
- Published
- 2018
11. Hybrid Capture–Based Genomic Profiling of Circulating Tumor DNA from Patients with Advanced Cancers of the Gastrointestinal Tract or Anus
- Author
-
Lauren Young, Samuel J. Klempner, Jeffrey S. Ross, Bryan Leyland-Jones, Fadi Braiteh, Razelle Kurzrock, Siraj M. Ali, Brady Forcier, Alexa B. Schrock, Vincent A. Miller, Jason K. Sicklick, Allison Welsh, Craig Devoe, Rodolfo Bordoni, Dean Pavlick, Richard D. Carvajal, Jon Chung, Joseph Chao, and Philip J. Stephens
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.disease_cause ,Article ,Circulating Tumor DNA ,03 medical and health sciences ,0302 clinical medicine ,Biomarkers, Tumor ,medicine ,Humans ,Gene ,Aged ,Gastrointestinal Neoplasms ,Neoplasm Staging ,Aged, 80 and over ,Mutation ,Gastrointestinal tract ,business.industry ,Hybrid capture ,Nucleic Acid Hybridization ,Cancer ,Genomics ,Middle Aged ,Anus Neoplasms ,Anus ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Organ Specificity ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Cancer research ,Female ,KRAS ,Neoplasm Grading ,business ,Follow-Up Studies - Abstract
Purpose: Genomic profiling of tumor biopsies from advanced gastrointestinal and anal cancers is increasingly used to inform treatment. In some cases, tissue biopsy can be prohibitive, and we sought to investigate whether analysis of blood-derived circulating tumor DNA (ctDNA) may provide a minimally invasive alternative. Experimental Design: Hybrid capture–based genomic profiling of 62 genes was performed on blood-based ctDNA from 417 patients with gastrointestinal carcinomas to assess the presence of genomic alterations (GA) and compare with matched tissue samples. Results: Evidence of ctDNA was detected in 344 of 417 samples (82%), and of these, ≥1 reportable GA was detected in 89% (306/344) of samples. Frequently altered genes were TP53 (72%), KRAS (35%), PIK3CA (14%), BRAF (8%), and EGFR (7%). In temporally matched ctDNA and tissue samples available from 25 patients, 86% of alterations detected in tissue were also detected in ctDNA, including 95% of short variants, but only 50% of amplifications. Conversely, 63% of alterations detected in ctDNA were also detected in matched tissue. Examples demonstrating clinical utility are presented. Conclusions: Genomic profiling of ctDNA detected potentially clinically relevant GAs in a significant subset of patients with gastrointestinal carcinomas. In these tumor types, most alterations detected in matched tissue were also detected in ctDNA, and with the exception of amplifications, ctDNA sequencing routinely detected additional alterations not found in matched tissue, consistent with tumor heterogeneity. These results suggest feasibility and utility of ctDNA testing in advanced gastrointestinal cancers as a complementary approach to tissue testing, and further investigation is warranted. Clin Cancer Res; 24(8); 1881–90. ©2018 AACR.
- Published
- 2018
12. 555TiP A first-in-human trial of the integrin beta-6-targeted antibody–drug conjugate, SGN-B6A, in patients with advanced solid tumors
- Author
-
Amita Patnaik, Afshin Dowlati, P. Zhou, Sarina Anne Piha-Paul, Antoine Hollebecque, Juanita Lopez, K. Sehgal, Rachel E. Sanborn, Fadi Braiteh, N. Nazarenko, B. Bockorny, Vladimir Galvao, Emiliano Calvo, and S. Peters
- Subjects
Antibody-drug conjugate ,Oncology ,business.industry ,Cancer research ,Medicine ,In patient ,Integrin, beta 6 ,Hematology ,First in human ,business - Published
- 2021
13. P-95 EV-202: An open-label, multicenter, phase 2 study of enfortumab vedotin in patients with previously treated locally advanced or metastatic solid tumors, including upper gastrointestinal cancers
- Author
-
Joaquina Baranda, Chunzhang Wu, K. Muro, Fadi Braiteh, Justine Yang Bruce, and S. Gorla
- Subjects
medicine.medical_specialty ,business.industry ,Locally advanced ,Enfortumab vedotin ,Phases of clinical research ,Hematology ,Gastroenterology ,Oncology ,Internal medicine ,medicine ,Upper gastrointestinal ,In patient ,Open label ,business ,Previously treated - Published
- 2021
14. Detection of an ALK Fusion in Colorectal Carcinoma by Hybrid Capture-Based Assay of Circulating Tumor DNA
- Author
-
Andrea Z. Lai, Vincent A. Miller, Siraj M. Ali, Jeffrey S. Ross, Evgeny Yakirevich, Fadi Braiteh, Alexa B. Schrock, and Rachel L. Erlich
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,business.industry ,Colorectal cancer ,Microsatellite instability ,medicine.disease_cause ,medicine.disease ,Molecular biology ,digestive system diseases ,Staining ,Fusion gene ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Circulating tumor DNA ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Medicine ,Immunohistochemistry ,KRAS ,business ,neoplasms - Abstract
ALK rearrangements have been observed in 0.05%-2.5% of patients with colorectal cancers (CRCs) and are predicted to be oncogenic drivers largely mutually exclusive of KRAS, NRAS, or BRAF alterations. Here we present the case of a patient with metastatic CRC who was treatment naive at the time of molecular testing. Initial ALK immunohistochemistry (IHC) staining was negative, but parallel genomic profiling of both circulating tumor DNA (ctDNA) and tissue using similar hybrid capture-based assays each identified an identical STRN-ALK fusion. Subsequent ALK IHC staining of the same specimens was positive, suggesting that the initial result was a false negative. This report is the first instance of an ALK fusion in CRC detected using a ctDNA assay. Key points Current guidelines for colorectal cancer (CRC) only recommend genomic assessment of KRAS, NRAS, BRAF, and microsatellite instability (MSI) status.ALK rearrangements are rare in CRC, but patients with activating ALK fusions have responded to targeted therapiesALK rearrangements can be detected by genomic profiling of ctDNA from blood or tissue, and this methodology may be informative in cases where immunohistochemistry (IHC) or other standard testing is negative.
- Published
- 2017
15. Updated survival and biomarker analyses of a randomized phase II study of atezolizumab vs. docetaxel in 2L/3L NSCLC (POPLAR)
- Author
-
Keunchil Park, Conrad R. Lewanski, Fadi Braiteh, A. Artal-Cortes, Alan Sandler, Achim Rittmeyer, Julien Mazieres, Jing Yi, Pei He, Johan Vansteenkiste, Wei Zou, Louis Fehrenbacher, Daniel S. Chen, Alexander I. Spira, Marcus Ballinger, Daniel Waterkamp, and David J. Smith
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Phases of clinical research ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Docetaxel ,Atezolizumab ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Biomarker (medicine) ,business ,medicine.drug - Published
- 2017
16. Abstract P6-12-15: Efficacy results of a phase 1/2 study of glucocorticoid receptor (GR) antagonist mifepristone (MIFE) in combination with eribulin in GR-positive triple-negative breast cancer (TNBC)
- Author
-
Fadi Braiteh, Sharon Wilks, A Marcantonio, Rita Nanda, Donald A. Richards, Joyce A. O'Shaughnessy, Gabrielle M. Baker, Timothy J. Pluard, Carlos Becerra, Alexander I. Spira, HS Han, RS Fishman, Elisavet Paplomata, Modiano, and Suzanne D. Conzen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Cancer ,Pharmacology ,Neutropenia ,medicine.disease ,Metastatic breast cancer ,Regimen ,chemistry.chemical_compound ,Breast cancer ,chemistry ,Internal medicine ,medicine ,business ,Triple-negative breast cancer ,Eribulin - Abstract
Background: GR is variably expressed in TNBC and high expression is associated with poor prognosis in estrogen receptor-negative (ER-) early stage breast cancer. Treatment with mifepristone (MIFE) potentiates the effectiveness of chemotherapy in GR+ TNBC xenografts. Enrollment is complete in this study of patients with GR+ TNBC treated at the recommended Phase 2 dose (RP2D) of MIFE in combination with eribulin. Objectives: To determine the safety, tolerability, pharmacokinetics (PK) and clinical activity of the MIFE plus eribulin combination in pts with GR+ TNBC at the RP2D. Methods: Eligibility: In Part 1 (dose finding), pts with solid tumors; in Part 2 (expansion phase), pts with TNBC (GR result required at time of screening in Part 1, but could be pending at time of screening in Part 2). Up to 5 prior chemotherapy regimens for advanced disease; ECOG PS 0-1; adequate end-organ function. Design: 3 + 3 dose escalation scheme. After a 7-day lead-in of oral daily MIFE alone, MIFE was continued daily and eribulin was given on days 1 and 8 of a 21-day cycle. GR+ was defined as >10% of tumor cells with any intensity of GR staining. Results: 16 pts with metastatic breast cancer were treated in Part 1, and 21 pts with TNBC were treated in Part 2. Median age was 54 (range 30-81). MTD/RP2D was MIFE 300 mg/day + eribulin 1.1 mg/m2. Safety: DLT in Part 1 was neutropenia. Neutropenia occurred in 23/36 total patients (2 Grade [G] 1, 10 G3, 11 G4); 2 instances included neutropenic fever. Recovery of WBC was brisk with growth factor support. Neuropathy was observed in 8 pts (5 G1, 1 G2, 2 G3). Other most common AEs (fatigue, hypokalemia, nausea, alopecia) were mainly G1 or G2; among these, G3/G4 events were limited to fatigue (4 G3), hypokalemia (3 G3 and 1 G4) and nausea (1 G3). There were 2 instances of G1 vaginal bleeding. There was no impact of MIFE on eribulin PK. Efficacy: There were 23 evaluable pts with TNBC across Parts 1 and 2 treated at the RP2D: 21 GR+, 2 GR status unknown; median of 3 prior chemotherapy regimens; 1 patient had received prior eribulin. Responses were: 3 PR, 8 SD, 11 PD and one too early to assess. Median PFS was 9 weeks. Conclusions: MIFE plus eribulin was well tolerated and appears to be an active treatment regimen. Five TNBC patients had a PFS longer than the upper 95% CI for PFS (i.e., >15 wks) reported by Aogi et al. for TNBC treated with eribulin (Annals of Oncology 2012?23:144148). Clinical trial information: NCT02014337. Citation Format: Han HS, Wilks S, Paplomata E, Modiano MR, Becerra C, Braiteh FS, Spira AI, Pluard TJ, Richards DA, Conzen SD, Baker G, Fishman RS, Marcantonio A, O'Shaughnessy J, Nanda R. Efficacy results of a phase 1/2 study of glucocorticoid receptor (GR) antagonist mifepristone (MIFE) in combination with eribulin in GR-positive triple-negative breast cancer (TNBC) [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P6-12-15.
- Published
- 2017
17. Efficacy and Safety of Onartuzumab in Combination With First-Line Bevacizumab- or Pemetrexed-Based Chemotherapy Regimens in Advanced Non-Squamous Non–Small-Cell Lung Cancer
- Author
-
Thomas Lowe, Filippo de Braud, Silvia Novello, C. Daniel Kingsley, Thomas Cosgriff, Tarek Mekhail, Hervé Lena, Wolfgang Schütte, See Phan, Jessie Hao-Ru Hsu, Fadi Braiteh, Heather A. Wakelee, Diego Kaen, William E. Lawler, Zanete Zvirbule, and Michelle Boyer
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Lung Neoplasms ,Phases of clinical research ,Carboplatin ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,Aged, 80 and over ,education.field_of_study ,Antibodies, Monoclonal ,Middle Aged ,Prognosis ,Bevacizumab ,Survival Rate ,Pemetrexed ,Onartuzumab ,030220 oncology & carcinogenesis ,Cohort ,Female ,Safety ,medicine.drug ,Adult ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Paclitaxel ,Population ,Adenocarcinoma ,03 medical and health sciences ,Double-Blind Method ,Internal medicine ,medicine ,Humans ,Progression-free survival ,education ,Lung cancer ,Aged ,Neoplasm Staging ,business.industry ,medicine.disease ,Surgery ,030104 developmental biology ,Cisplatin ,business ,Follow-Up Studies - Abstract
Background Onartuzumab is a monovalent monoclonal antibody that binds with the extracellular domain of the MET receptor. Given the role of MET in non–small-cell lung cancer (NSCLC), we investigated whether onartuzumab added to first-line chemotherapy efficacy in non-squamous NSCLC. Methods Patients with untreated stage IIIB/IV non-squamous NSCLC, stratified by MET diagnostic status, were randomized to receive onartuzumab (15 mg/kg intravenously every 3 weeks) or placebo in combination with either paclitaxel/platinum/bevacizumab (bevacizumab cohort), or in combination with platinum/pemetrexed (pemetrexed cohort) with maintenance bevacizumab or pemetrexed and onartuzumab/placebo as appropriate. Co-primary endpoints of this phase II study were progression-free survival (PFS) in all patients and in MET+ patients (2+/3+), defined by the Ventana immunohistochemistry assay; secondary endpoints included overall survival (OS), objective response rate (ORR), safety, and pharmacokinetics. Results Efficacy data were available for 139 and 120 patients in the bevacizumab and pemetrexed cohorts, respectively. No benefit was seen in the PFS endpoint in the intent-to treat population of either cohort, but was numerically worse in the onartuzumab arm of the MET+ subgroup of the bevacizumab cohort. The onartuzumab and placebo arms had similar ORR and OS results in both cohorts. A higher incidence of some adverse events was observed with onartuzumab versus placebo, including peripheral edema (30% vs. 3%, bevacizumab cohort; 48% vs. 14%, pemetrexed cohort) and venous thromboembolic events (bevacizumab cohort only, 15% vs. 6%). Conclusion Onartuzumab does not appear to provide any additional clinical benefit when given in combination with current first-line standard-of-care chemotherapy for non-squamous NSCLC.
- Published
- 2017
18. Safety, clinical activity and biomarker assessments of atezolizumab from a Phase I study in advanced/recurrent ovarian and uterine cancers
- Author
-
Ana Oaknin, Leisha A. Emens, Luciana Molinero, Erika Hamilton, Indrani Sarkar, Marcella Fassò, Yvonne G. Lin, Joyce F. Liu, Joseph Paul Eder, Fadi Braiteh, Yulei Wang, Jennifer Taylor Veneris, Carol O'Hear, Michael S. Gordon, and Ani Sarkis Balmanoukian
- Subjects
0301 basic medicine ,Oncology ,Adult ,medicine.medical_specialty ,Programmed Cell Death 1 Receptor ,Antineoplastic Agents ,Carcinoma, Ovarian Epithelial ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Uterine cancer ,Atezolizumab ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Adverse effect ,Aged ,Ovarian Neoplasms ,Dose-Response Relationship, Drug ,business.industry ,Obstetrics and Gynecology ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Clinical trial ,030104 developmental biology ,Tolerability ,030220 oncology & carcinogenesis ,Cohort ,Uterine Neoplasms ,Biomarker (medicine) ,Female ,Neoplasm Recurrence, Local ,Ovarian cancer ,business - Abstract
Objective Patients with advanced/recurrent epithelial ovarian and uterine cancers have limited treatment options beyond platinum chemotherapy. Both tumor types can express programmed death-ligand 1 (PD-L1), providing a potential therapeutic target for these patients. Here we present data from the ovarian and uterine cancer cohorts of the Phase I atezolizumab monotherapy study (PCD4989g). Methods This Phase I, multi-center, first-in-human, open-label, dose-escalation/expansion clinical trial investigated single-agent atezolizumab in cohorts of patients with recurrent epithelial ovarian or uterine cancer. The primary objective was to evaluate the safety and tolerability of single-agent atezolizumab. Anti-tumor activity and preliminary assessment of potential biomarkers were evaluated as secondary and exploratory objectives, respectively. Results The ovarian and uterine cancer cohorts enrolled 12 and 15 patients, respectively (10 [83%] and 5 [33%], respectively, had PD-L1 ≥ 5% on tumor-infiltrating immune cells). Atezolizumab was generally well tolerated with no new safety signals identified. The safety profiles in both cohorts were consistent with the known profile of atezolizumab monotherapy. Treatment-related adverse events (AEs) were mostly Grade ≤ 2, with no treatment-related Grade ≥ 4 AEs reported. Preliminary anti-tumor activity, with long durations of response, was observed in 2 patients from each cohort (ovarian cancer, 8.1 and 30.6+ months; uterine cancer, 7.3 and 16.6+ months). High microsatellite instability and tumor mutational burden were noted in the responders from the uterine cancer cohort. Conclusions Atezolizumab monotherapy was well tolerated in patients with epithelial ovarian or uterine cancer and may have clinical activity warranting further investigation. Trial registration ClinicalTrials.gov identifier: NCT01375842
- Published
- 2019
19. Derazantinib (ARQ 087) in advanced or inoperable FGFR2 gene fusion-positive intrahepatic cholangiocarcinoma
- Author
-
Nevena Damjanov, Sherrie Bhoori, Kyriakos P. Papadopoulos, Terence Hall, Michele Droz dit Busset, Fadi Braiteh, Christian Cotsoglou, Nicola Personeni, William P. Harris, Walid L. Shaib, Yunxia Wang, Brian Schwartz, Vincenzo Mazzaferro, Gianluca Masi, Vittorina Zagonel, Lorenza Rimassa, Julia Kazakin, Bassel F. El-Rayes, and Filippo de Braud
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Oncogene Proteins, Fusion ,medicine.medical_treatment ,Gastroenterology ,Article ,Cholangiocarcinoma ,Fusion gene ,03 medical and health sciences ,Hyperphosphatemia ,Alkaloids ,Targeted therapies ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,In patient ,Receptor, Fibroblast Growth Factor, Type 2 ,Intrahepatic Cholangiocarcinomas ,Adverse effect ,Protein Kinase Inhibitors ,Intrahepatic Cholangiocarcinoma ,Aged ,Aged, 80 and over ,Clinical Trials as Topic ,Chemotherapy ,Aniline Compounds ,business.industry ,Bile duct cancer ,Middle Aged ,medicine.disease ,Bile Ducts, Intrahepatic ,Bile Duct Neoplasms ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Toxicity ,Quinazolines ,Female ,Gene Fusion ,business - Abstract
Background Next-generation sequencing has identified actionable genetic aberrations in intrahepatic cholangiocarcinomas (iCCA), including the fibroblast growth factor receptor 2 (FGFR2) fusions. Derazantinib (ARQ 087), an orally bioavailable, multi-kinase inhibitor with potent pan-FGFR activity, has shown preliminary therapeutic activity against FGFR2 fusion-positive iCCA. Methods This multicentre, phase 1/2, open-label study enrolled adult patients with unresectable iCCA with FGFR2 fusion, who progressed, were intolerant or not eligible to first-line chemotherapy (NCT01752920). Subjects received derazantinib in continuous daily doses. Tumour response was assessed according to RECIST 1.1 every 8 weeks. Results Twenty-nine patients (18 women/11 men; median age, 58.7 years), 2 treatment-naive and 27 who progressed after at least one prior systemic therapy, were enrolled. Overall response rate was 20.7%, disease control rate was 82.8%. Estimated median progression-free survival was 5.7 months (95% CI: 4.04–9.2 months). Treatment-related adverse events (AE) were observed in 27 patients (93.1%, all grades), including asthenia/fatigue (69.0%), eye toxicity (41.4%), and hyperphosphatemia (75.9%). Grade ≥ 3 AEs occurred in 8 patients (27.6%). Conclusion Derazantinib demonstrated encouraging anti-tumour activity and a manageable safety profile in patients with advanced, unresectable iCCA with FGFR2 fusion who progressed after chemotherapy. A pivotal trial of derazantinib in iCCA is ongoing (NCT03230318).
- Published
- 2019
20. A phase 1 study of SGN-B6A, an antibody-drug conjugate targeting integrin beta-6, in patients with advanced solid tumors (SGNB6A-001, Trial in Progress)
- Author
-
Antoine Hollebecque, Emiliano Calvo, Rachel E. Sanborn, Vladimir Galvao, Afshin Dowlati, Amita Patnaik, Fadi Braiteh, N. Nazarenko, Sarina Anne Piha-Paul, Peigen Zhou, and Juanita Lopez
- Subjects
Cancer Research ,Antibody-drug conjugate ,biology ,business.industry ,Cell ,Integrin ,Therapeutic targeting ,Extracellular matrix ,Pathogenesis ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Medicine ,Integrin, beta 6 ,In patient ,business - Abstract
TPS3144 Background: The extracellular matrix (ECM) plays an important role in solid tumor pathogenesis and is a major focus of research and therapeutic targeting. Integrin beta-6 is a cell surface receptor that interacts with the ECM to mediate cellular adhesion. Integrin beta-6 is overexpressed in numerous solid tumors and its expression is a negative prognostic marker in cancers including colorectal, non-small cell lung, gastric, and cervical cancers. SGN-B6A is an investigational vedotin, an antibody-drug conjugate directed against integrin beta-6 to selectively deliver the cytotoxic agent monomethyl auristatin E, which binds tubulin and induces apoptosis. In multiple xenograft models, treatment with SGN-B6A resulted in tumor growth delay and regression in tumor volume when compared to non-binding control. Methods: SGNB6A-001 (NCT04389632) is a phase 1, first-in-human, open-label, multicenter study designed to evaluate the safety, tolerability, pharmacokinetics (PK), and antitumor activity of SGN-B6A in adults with select advanced solid tumors. Primary objectives are to evaluate the safety and tolerability of SGN-B6A in patients with advanced solid tumors, identify the maximum tolerated dose, and identify a recommended dose and schedule. The study has 2 parts: dose escalation (Part A) and dose expansion with multiple disease-specific cohorts and a biology cohort (Part B). SGN-B6A will initially be given by intravenous infusion on Days 1, 8, and 15 of 21-day cycles. The dose escalation (Part A) will be conducted using the modified toxicity probability interval method to determine a dose that demonstrates a dose-limiting toxicity rate of 25% with a 5% margin. The dose and schedule for Part B will be determined based on evaluation of safety, PK, and pharmacodynamic biomarkers. Response evaluations will be based on RECIST v1.1. Patients must be ≥18 years old and have histologically or cytologically confirmed metastatic or unresectable solid malignancy within one of the following tumor types: non-small cell lung cancer, head and neck squamous cell cancer, breast cancer, esophageal cancer, ovarian cancer, cutaneous squamous cell cancer, exocrine pancreatic adenocarcinoma, bladder cancer, cervical cancer, or gastric cancer. After an appropriate dose and schedule are determined in Part A, safety and preliminary antitumor efficacy of SGN-B6A will be evaluated in indication-specific cohorts (Part B). This study is ongoing in sites across North America and Europe. Clinical trial information: NCT04389632.
- Published
- 2021
21. Circulating tumor DNA-based genomic profiling of small bowel adenocarcinoma
- Author
-
Christopher R. Cogle, Pat Gulhati, Leylah Drusbosky, Sujith Kalmadi, Jason S. Starr, Karan Pandya, Hiba I. Dada, and Fadi Braiteh
- Subjects
Cancer Research ,Genomic profiling ,business.industry ,Small bowel adenocarcinoma ,Malignancy ,medicine.disease ,Lower incidence ,Oncology ,Circulating tumor DNA ,medicine ,Overall survival ,Cancer research ,business ,Intestinal Cancer ,neoplasms ,Stage at diagnosis - Abstract
3523 Background: Small bowel adenocarcinoma (SBA) is a rare malignancy, with lower incidence, later stage at diagnosis, and worse overall survival compared to other intestinal cancers, such as colorectal cancer (CRC). Since the majority of small bowel tumors are not accessible to endoscopic biopsy, comprehensive genomic profiling using circulating tumor DNA (ctDNA) may enable non-invasive detection of targetable genomic alterations (GA) in SBA patients. In this study, we characterize the ctDNA GA landscape in SBA. Methods: Analysis of 299 ctDNA samples prospectively collected from 265 SBA patients between 2017 to 2020 was performed using a 73 gene next generation sequencing panel (Guardant360). A subset of patients underwent longitudinal analysis of changes in GA associated with systemic therapy. Results: Of the 265 patients, 160 (60.3%) were male; the median age was 66 (range: 21-93 years). The most common GA identified in SBA patients included TP53 [58%], KRAS [44%], and APC [40%]. MSI was detected in 3.4% of SBA patients. When stratified by primary tumor location, APC, KRAS, TP53, PIK3CA, and ARID1A were the most common GA identified in both duodenal and jejunal adenocarcinomas. ERBB2, BRCA2 and CDK6 alterations were enriched in duodenal adenocarcinoma, while NOTCH and BRAF alterations were enriched in jejunal adenocarcinoma. The most common currently-targetable GA identified were ATM [18%], PIK3CA [17%], EGFR [15%], CDK4/6 [11%], BRAF [10%], and ERBB2 [10%]. Unique differences in GA between SBA and CRC were identified: i) the majority of ERBB2 alterations are mutations (89%) in the extracellular domain and kinase domain, not amplifications (11%); ii) the majority of BRAF alterations are non V600E mutations (69%) and amplifications (28%); iii) there is a significantly lower rate of APC mutations (40%). Alterations in DNA damage response pathway proteins, including ATM and BRCA 1/2, were identified in 30% of SBA patients. ATM alterations were more common in patients ³65 years old. The most common mutations predicted to be related to clonal hematopoiesis of indeterminate potential were TP53, KRAS and GNAS. Longitudinal ctDNA analysis in 4 SBA patients revealed loss of mutations associated with therapeutic response (TP53 R342*, MAPK3 R189Q) and acquired mutations associated with therapeutic resistance (NF1 R1968*, MET S170N, RAF1 L613V). Conclusions: This study represents the first large-scale blood-based ctDNA genomic profiling of SBA. SBA represents a unique molecular entity with differences in frequency and types of GA compared to CRC. Variations in GA were noted based on anatomic origin within the small intestine. Longitudinal ctDNA monitoring revealed novel GA associated with therapeutic resistance. Identification of multiple targetable GA may facilitate clinical decision making and improve patient outcomes in SBA, especially when a tissue biopsy is not feasible or sufficient for comprehensive genomic profiling.
- Published
- 2021
22. Evaluation of bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in cervical cancer: Data from phase 1 and phase 2 studies
- Author
-
Emiliano Calvo, William Jeffery Edenfield, Marika Rasschaert, Alexander I. Spira, G. Jehl, Luis Paz-Ares, Tjoung-Won Park-Simon, Andrés Cervantes, Isabelle Dussault, Julius Strauss, Fadi Braiteh, James L. Gulley, Maria de Miguel, Tianhong Li, Laureen S. Ojalvo, Federico Longo, and Suzanne Wendy Allan
- Subjects
Cervical cancer ,Cancer Research ,biology ,business.industry ,Fda approval ,Pembrolizumab ,medicine.disease ,Fusion protein ,Oncology ,PD-L1 ,medicine ,Cancer research ,biology.protein ,business ,Objective response ,Metastatic cervical cancer ,Transforming growth factor - Abstract
5509 Background: The accelerated FDA approval of pembrolizumab validated the efficacy of anti–PD-(L)1 therapy for pts with recurrent/metastatic cervical cancer; however, the objective response rate (ORR) with pembrolizumab was 14.3% in pts with PD-L1 expressing tumors. HPV infection is implicated in > 95% of cervical cancers and is linked to upregulation of TGF-β signaling. Bintrafusp alfa is a first-in-class bifunctional fusion protein composed of the extracellular domain of the TGF-βRII receptor (a TGF-β “trap”) fused to a human IgG1 mAb blocking PD-L1. We report pooled safety and efficacy in pts with immune checkpoint inhibitor–naive, recurrent/metastatic cervical cancer treated with bintrafusp alfa in phase 1 (INTR@PID 001; NCT02517398) and phase 2 (study 012; NCT03427411) studies. Methods: Pts with pretreated, immune checkpoint inhibitor–naive, recurrent/metastatic cervical cancer received bintrafusp alfa 0.3-30 mg/kg (phase 1 dose escalation) or 1200 mg Q2W (phase 1 expansion/phase 2) until progressive disease, unacceptable toxicity, or withdrawal. Treatment past progression was allowed. Primary endpoints were safety for the dose-escalation part of the phase 1 study and best overall response per RECIST 1.1 for the expansion part of phase 1 and phase 2 studies. Secondary endpoints for the expansion part of the phase 1 and 2 studies included safety. Results: As of May 15, 2020 (phase 1) and December 22, 2020 (phase 2), 39 pts had received bintrafusp alfa for a median duration of 2.8 months (range, 0.5-19.3). The median follow-up to data cutoff was 35.0 months and 24.1 months for the phase 1 and phase 2 studies, respectively. All pts had received prior anticancer therapy; 16 pts (41.0%) had received ≥3 prior anticancer regimens. There were 2 complete responses and 9 partial responses (PRs; ORR per RECIST 1.1, 28.2%). Median duration of response was 11.7 months (range, 1.4-41.2), and 5 pts (45.5%) had ongoing responses (duration 1.5-41.2 months). An additional delayed PR was observed (duration 23.7 months). Reponses occurred irrespective of tumor histology or prior bevacizumab or radiation treatment. Median overall survival (mOS) was 13.4 months (95% CI, 5.5 to not reached); 24-month OS rate was 33.2%. Any-grade treatment-related adverse events (TRAEs) occurred in 33 pts (84.6%). Grade 3 TRAEs occurred in 8 pts (20.5%; anemia, colitis, gastroparesis, upper gastrointestinal hemorrhage, keratoacanthoma, cystitis noninfective, hematuria, pneumonitis, rash macular [n = 1 each]); 1 patient (2.6%) had a grade 4 TRAE (asymptomatic hypokalemia related to the above grade 3 gastroparesis). No treatment-related deaths occurred. Conclusions: Bintrafusp alfa had a manageable safety profile and demonstrated clinical activity in pts with heavily pretreated, immune checkpoint inhibitor–naive recurrent/metastatic cervical cancer. Clinical trial information: NCT02517398 , NCT03427411.
- Published
- 2021
23. Phase 1 dose-escalation study of STRO-002, an antifolate receptor alpha (FRα) antibody drug conjugate (ADC), in patients with advanced, progressive platinum-resistant/refractory epithelial ovarian cancer (EOC)
- Author
-
Clifford DiLea, Lin Lu, Craig Jerome Berman, Erika Hamilton, Denise Uyar, Michael Palumbo, Arturo Molina, Sami Diab, David M. O'Malley, Venita I. De Almeida, John Paul Diaz, R. Wendel Naumann, Lainie P. Martin, Richard T. Penson, John W. Moroney, Shannon Matheny, Russell J. Schilder, and Fadi Braiteh
- Subjects
Cancer Research ,Antibody-drug conjugate ,business.industry ,Alpha (ethology) ,chemistry.chemical_compound ,Oncology ,Refractory ,chemistry ,Antifolate ,Cancer research ,Medicine ,Epithelial ovarian cancer ,In patient ,business ,Receptor ,Platinum resistant - Abstract
5550 Background: STRO-002-GM1 is a Phase 1, open-label study in patients (pts) with advanced, platinum resistant or refractory EOC. STRO-002 is a novel FRα-targeting ADC with a precise DAR of 4 using site-specific conjugation technology to circumvent limitations of current ADCs. STRO-002 induces immunogenic cell death and contains the tubulin-targeting 3-aminophenyl hemiasterlin warhead SC209, a potent cytotoxin that is a weak substrate for P-gp. Methods: STRO-002 is given IV on Day 1 of each 21-day cycle until disease progression. Ocular exams are performed at baseline and every other cycle. Prophylactic corticosteroid eyedrops are not administered. FRα expression was not required for eligibility and retrospective analysis of FRα expression in archival tumor tissue is ongoing. Results: Enrollment has been completed with 39 pts treated at 9 dose levels (0.5 to 6.4 mg/kg). Data cut-off is Jan 30, 2021. Median age was 61 years (range 48-79). Median number of prior systemic therapies was 6 (range 2-11). 86% of treatment emergent adverse events (AEs) were Grade 1-2. The most common treatment related Grade 3 and 4 AEs were reversible neutrophil count decreased (36%) and neutropenia (33%), Grade 3 arthralgia (12.8%) and neuropathy (7.7%). Two pts developed neutropenic fever that resolved with antibiotic therapy. 34 pts were treated at clinically active doses (≥ 2.9 mg/kg) and 31/34 are evaluable for RECIST 1.1 response. Objective responses were seen in 10/31 pts - 1 CR, 4 confirmed PR, and 5 unconfirmed PR (imaging studies under review in 1 pt with uPR). Disease control rate (CR+PR+SD) is 74% at ≥ 12 weeks and 61% at ≥ 16 weeks. 5 pts remain on treatment with 3 ongoing at > 74 weeks. FRα-expression results are available in 14 pts treated at ≥ 2.9 mg/kg with 50% low, 21% medium and 29% high FRα-expressing tumors per PS2+ scoring algorithm. 12/13 pts with H-scores of ≥ 105 achieved disease control with PR or SD. Maximum plasma concentrations of STRO-002 were achieved at the end of the 1 hour infusion and exposure increased in an apparent dose proportionate/linear manner. Conclusions: STRO-002 is a novel FRα-targeting ADC with an encouraging emerging safety and efficacy profile in heavily pretreated relapsed/refractory EOC. No ocular toxicity signals have been observed. Durable responses and anti-tumor activity have been demonstrated across a broad range of FRα expression levels in evaluable pts treated at ≥ 2.9 mg/kg. 48% (15/31) of pts were on treatment without disease progression for ≥ 24 weeks and 13% (4/31) remain on treatment for over a year, suggesting that STRO-002 is well tolerated in long-term responding patients. A randomized expansion cohort comparing STRO-002 at 4.3 mg/kg vs 5.2 mg/kg dose levels in less heavily pretreated EOC pts is ongoing. Clinical trial information: NCT03748186.
- Published
- 2021
24. Lack of pharmacokinetic drug–drug interaction between ramucirumab and irinotecan in patients with advanced solid tumors
- Author
-
Fadi Braiteh, Ling Gao, Christopher John Asakiewicz, James J. Lee, Yong Lin, F. Nasroulah, Ding Wang, Archana Chaudhary, Dale R. Shepard, Crystal S. Denlinger, and Patricia LoRusso
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Leucovorin ,Phases of clinical research ,Antineoplastic Agents ,SN-38 ,Neutropenia ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Irinotecan ,Toxicology ,Gastroenterology ,Ramucirumab ,03 medical and health sciences ,Folinic acid ,chemistry.chemical_compound ,0302 clinical medicine ,Asian People ,Pharmacokinetics ,Neoplasms ,Internal medicine ,medicine ,Humans ,Drug Interactions ,Pharmacology (medical) ,Aged ,Antibiotics, Antineoplastic ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Antineoplastic Agents, Phytogenic ,030104 developmental biology ,Oncology ,chemistry ,Area Under Curve ,030220 oncology & carcinogenesis ,FOLFIRI ,Camptothecin ,Female ,Fluorouracil ,business ,medicine.drug - Abstract
The objective of this phase II study was to evaluate the potential of pharmacokinetic (PK) drug–drug interactions between ramucirumab and irinotecan or its metabolite, SN-38, when administered with folinic acid and 5-fluorouracil (FOLFIRI). Patients received intravenous infusions of FOLFIRI and ramucirumab 8 mg/kg on Day 1 of a 2-week cycle. FOLFIRI was administered alone in Cycle 1; ramucirumab followed by FOLFIRI was administered in all subsequent cycles. Blood was collected at regular intervals after infusions in Cycles 1 and 2 to determine irinotecan, SN-38, and ramucirumab concentrations. PK parameters were derived by noncompartmental analysis. Twenty-nine patients received treatment. The dose-normalized area under the concentration versus time curve from zero to infinity [AUC(0–∞)] and the maximum observed concentration (C max) of irinotecan and SN-38 were comparable between Cycle 1 (FOLFIRI alone) and Cycle 2 (ramucirumab + FOLFIRI). The ratios of geometric least squares (LS) means for irinotecan were 0.93 (90 % CI 0.83–1.05) for AUC(0–∞) and 1.04 (90 % CI 0.97–1.12) for C max. The ratios of geometric LS means for SN-38 were 0.95 (90 % CI 0.88–1.04) for AUC(0–∞) and 0.97 (90 % CI 0.85–1.12) for C max. The most common treatment-emergent adverse events, regardless of grade, were fatigue (19 patients, 65.5 %), diarrhea, (16 patients, 55.2 %), and neutropenia (15 patients, 51.7 %). Grade ≥3 neutropenia was reported in 7 (24.1 %) patients. There was no PK drug–drug interaction between ramucirumab and irinotecan or its metabolite, SN-38. Ramucirumab with FOLFIRI was well tolerated in this study, with no new safety concerns.
- Published
- 2016
25. Abstract LB-387: Efficacy and safety of AB928 plus modified FOLFOX-6 (mFOLFOX-6) in participants with metastatic colorectal cancer (mCRC): Initial results at the recommended dose for expansion (ARC-3)
- Author
-
Daniel DiRenzo, Houston Gilbert, Melissa Paoloni, Fadi Braiteh, Matt J. Walters, Manuel Modiano, Michael Cecchini, Olivia Gardner, Lisa Seitz, Fang-Fang Yin, Rachel Woloski, and Ki Y. Chung
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,Colorectal cancer ,business.industry ,medicine.medical_treatment ,Cancer ,Immunotherapy ,Neutropenia ,medicine.disease ,FOLFOX ,Internal medicine ,Concomitant ,medicine ,Adverse effect ,business ,medicine.drug - Abstract
Background: The release of ATP from dying cancer cells in response to platinum-based chemotherapy increases extracellular immunosuppressive adenosine (A), which binds to and activates the A2a and A2b receptors on immune cells resulting in an ineffective anti-tumor immune response. Concomitant adenosine receptor blockade may therefore enhance the therapeutic efficacy of some chemotherapeutic agents. AB928, the first clinical-stage small molecule dual adenosine receptor antagonist, is highly potent, pharmacodynamically active, and has been well tolerated in dose escalation studies as a single agent or in combination with chemo/immunotherapy. Methods: ARC-3 (NCT03720678) is a Phase 1/1b, open-label study in participants (pts) with advanced CRC. Phase 1 escalation identified AB928 150 mg orally once daily as the recommended dose in combination with standard mFOLFOX-6. Phase 1b expansion is ongoing and includes at least 15 and up to 40 pts. Eligible pts must have unresectable or mCRC, ECOG performance status 0-1, and at least one RECIST measurable lesion. Phase 1 eligibility included up to 5 lines of prior therapy; Phase 1b is similarly scoped. Exploratory biomarker analyses include immunohistochemistry of the adenosine axis, tumoral next gene sequencing, and tumor/blood immune correlates. Results: As of 27Dec19, 21 pts received AB928 150 mg + mFOLFOX-6: 7 in Phase 1 and 14 in Phase 1b. All previously treated pts (n=12) were FOLFOX- and/or FOLFIRI-experienced. Prior metastatic therapies range from 3 to 5 in Phase 1 escalation and 0 to 3 in Phase 1b expansion. Adverse events (AEs) reported in >30% of pts included fatigue, diarrhea, and thrombocytopenia. AEs related to AB928 occurred in 13 pts and were mostly mild to moderate. AB928-related Grade 3 AEs reported by 3 pts were diarrhea, AST increase, and neutropenia; there were no Grade 4-5 AB928-related AEs. Out of 15 evaluable pts, by investigator assessment, the disease control rate was 100% with 2 partial responses (13%; 1 confirmed, 1 pending confirmation) and 13 stable disease (87%). Of pts with stable disease, 6/13 (46%) had tumor shrinkage >15%. Median time on treatment was 15.4 (range: 1.7 - 40.6+) and 11.9 (range: 2.7 - 15.7+) weeks for Phase 1 and Phase 1b, respectively, with initiation of Phase 1b dosing on 09Sep19. Enrollment up to 40 pts is proceeding based on early efficacy gates; 15 pts are currently receiving study treatment. Conclusions: AB928 with mFOLFOX-6 has been well tolerated without significant evidence of additive toxicity in pts with mCRC. Combination treatment was associated with disease control in all evaluable pts, including those with microsatellite stable and RAS/BRAF mutated mCRC. Additional updates on the safety, clinical activity, and correlative biomarker results for all escalation/expansion pts will be presented. Citation Format: Michael Cecchini, Manuel Modiano, Fadi Braiteh, Olivia S. Gardner, Houston N. Gilbert, Daniel DiRenzo, Lisa Seitz, Matt J. Walters, Fangfang Yin, Rachel Woloski, Melissa C. Paoloni, Ki Y. Chung. Efficacy and safety of AB928 plus modified FOLFOX-6 (mFOLFOX-6) in participants with metastatic colorectal cancer (mCRC): Initial results at the recommended dose for expansion (ARC-3) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr LB-387.
- Published
- 2020
26. Abstract CT165: An open label, multi-cohort, phase Ib study of xentuzumab and abemaciclib: Preliminary results from the advanced non-small cell lung cancer (NSCLC) cohort
- Author
-
Molly C. Hardebeck, Marie Paule Sablin, Francesco Ricci, Marta Capelán, Marta Puig, Aleix Prat, Mingchi Hsu, Javier Garcia-Corbacho, Anthony Gonçalves, Fadi Braiteh, Douglas Yee, Dennis Chin-Lun Huang, Hiroji Iwata, and Patricia LoRusso
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,non-small cell lung cancer (NSCLC) ,Cancer ,Neutropenia ,medicine.disease ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Cohort ,Clinical endpoint ,Medicine ,business ,Abemaciclib ,Progressive disease - Abstract
Introduction: Insulin-like growth factor (IGF) signaling and the cyclin D-cyclin-dependent kinase (CDK) 4 & 6-retinoblastoma pathway are implicated in cancer progression and treatment resistance. Activation of the IGF receptor results in upregulation of cyclin D1 and progression through the cell cycle. Dual inhibition of IGF and CDK 4 & 6 should lead to decreased cell proliferation through disruption of cell-cycle progression. This phase Ib trial assessed safety and preliminary efficacy of xentuzumab, an IGF-ligand neutralizing antibody, in combination with abemaciclib, a CDK 4 & 6 inhibitor. In a dose-finding cohort in patients (pts) with solid tumors, the recommended phase II dose was determined as xentuzumab 1000 mg weekly intravenously plus abemaciclib 150 mg every 12 hours (Q12h) orally. Here, we present data from an expansion cohort in pts with NSCLC. Methods: Eligible pts had stage IV NSCLC with measurable disease which had progressed after platinum-based chemotherapy and immunotherapy, and had either received 1-2 prior chemotherapies for advanced/metastatic disease or were judged ineligible for further standard second-line chemotherapy. Prior CDK 4 & 6 inhibitor therapy was not permitted. Pts received xentuzumab 1000 mg weekly plus abemaciclib 150 mg Q12h until disease progression or intolerability of study medication. The primary endpoint was objective response defined as best overall response of complete response or partial response (PR) per RECIST 1.1. Secondary endpoints included progression-free survival (PFS). Results: 25 pts with NSCLC were treated (17 male [68%], median age 64 years [range 53-76]). 4 pts remained on treatment at time of analysis. Median treatment exposure was 1.6 months (range 0.5-19.7). Of 19 pts evaluable for response, 1 had a best overall response of PR (65% target lesion shrinkage), 9 had stable disease (duration ≥24 weeks in 3 pts) and 9 had progressive disease. Median PFS was 2.1 months (95% CI 1.2-5.3). All pts experienced adverse events (AEs) and 23 experienced treatment-related AEs (TRAEs), most commonly diarrhea (n=15; 60%), nausea (n=10; 40%) and platelet count decreased (n=8; 32%). The most common grade ≥3 TRAEs were thrombocytopenia (n=4; 16%) and neutropenia (n=3; 12%). No AEs of hyperglycemia or blood glucose increased were reported. Serious AEs were experienced by 13 pts (52%). 2 pts had fatal serious AEs due to disease progression and respiratory failure. Conclusion: The combination of xentuzumab and abemaciclib showed preliminary efficacy in pts with NSCLC who had progressed on chemotherapy and an immune checkpoint inhibitor. One PR was reported. The combination had a manageable safety profile. Citation Format: Patricia LoRusso, Javier García-Corbacho, Fadi S. Braiteh, Anthony Gonçalves, Francesco Ricci, Hiroji Iwata, Marta Capelán, Marie Paule Sablin, Aleix Prat, Molly Catherine Hardebeck, Marta Puig, Dennis Chin-Lun Huang, Mingchi Hsu, Douglas Yee. An open label, multi-cohort, phase Ib study of xentuzumab and abemaciclib: Preliminary results from the advanced non-small cell lung cancer (NSCLC) cohort [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT165.
- Published
- 2020
27. Abstract CT169: A phase Ia study to evaluate RO7198457, an individualized Neoantigen Specific immunoTherapy (iNeST), in patients with locally advanced or metastatic solid tumors
- Author
-
Rachel Sabado, Mahesh Yadav, Patrick Twomey, Patrick McDonald, Michael S. Gordon, Johanna C. Bendell, D.R. Camidge, Jack Huang, Felicitas Müller, Lillian L. Siu, Ani Sarkis Balmanoukian, Jingbin Zhang, Sam Klempner, Ugur Sahin, Lars Mueller, Fadi Braiteh, Özlem Türeci, Matthew D. Hellmann, Evelyna Derhovanessian, Leila Delamarre, and Patricia LoRusso
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Melanoma ,medicine.medical_treatment ,Cancer ,Pembrolizumab ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cancer immunotherapy ,Atezolizumab ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Prior Immunotherapy ,Stage (cooking) ,Ovarian cancer ,business - Abstract
Background: Neoantigens arising from somatic mutations are attractive targets for cancer immunotherapy as they may be recognized as foreign by the immune system. RO7198457 is a systemically administered RNA-Lipoplex iNeST designed to stimulate T cell responses against neoantigens. A first-in-human Phase Ia study of RO7198457 was conducted in patients with locally advanced or metastatic solid tumors. Methods: RO7198457 is manufactured on a per-patient basis and contains up to 20 patient- specific neoantigens. Nine doses of RO7198457 were administered i.v. at weekly and bi-weekly intervals during the 12-week induction stage and every 24 weeks during the maintenance stage. Results: In total, 29 patients enrolled in cohorts with doses ranging from 25-100 μg. The most common tumor types were HR+/HER2+ breast, prostate, and ovarian cancer. The median number of prior therapies was 5 (range 1-17). 34% of patients received prior immunotherapy. Most patients had low PD-L1 expression (97% patients with Conclusion: RO7198457 can be manufactured for individual patients with clinically relevant turn-around times. RO7198457 has a manageable safety profile consistent with its mechanism of action, and induced strong neoantigen-specific immune responses in patients with low and intermediate mutational load tumors types. A Ph1b study in combination with atezolizumab and a randomized Ph2 study of RO7198457 in 1L melanoma patients with pembrolizumab have been initiated, and two randomized clinical trials are planned in resected lung and CRC. Citation Format: Fadi Braiteh, Patricia LoRusso, Ani Balmanoukian, Sam Klempner, D R. Camidge, Matthew Hellmann, Michael Gordon, Johanna Bendell, Lars Mueller, Rachel Sabado, Patrick Twomey, Leila Delamarre, Jack Huang, Mahesh Yadav, Jingbin Zhang, Patrick McDonald, Felicitas Müller, Evelyna Derhovanessian, Özlem Türeci, Ugur Sahin, Lillian Siu. A phase Ia study to evaluate RO7198457, an individualized Neoantigen Specific immunoTherapy (iNeST), in patients with locally advanced or metastatic solid tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT169.
- Published
- 2020
28. Abstract CT301: A phase Ib study to evaluate RO7198457, an individualized Neoantigen Specific immunoTherapy (iNeST), in combination with atezolizumab in patients with locally advanced or metastatic solid tumors
- Author
-
Charles G. Drake, Jingbin Zhang, Brian S. Henick, Guy Jerusalem, Rachel Sabado, Christian H. Ottensmeier, Scott A. Laurie, Jack Huang, Lawrence Fong, Lars Mueller, Fadi Braiteh, Thomas Powles, George A. Fisher, Ignacio Melero, Patricia LoRusso, Patrick Twomey, Ani Sarkis Balmanoukian, Raid Aljumaily, Jeffery Yachnin, Achim Rittmeyer, Michael S. Gordon, Eelke Gort, Rom Leidner, Aglaia Schiza, R. Camidge, Ryan J. Sullivan, Marcus Schmidt, Manesh Yadav, Sylvie Rottey, Marco A. J. Iafolla, Martin Schuler, Matthew D. Hellmann, Juanita Lopez, Johanna C. Bendell, Felicitas Mueller, Özlem Türeci, Luc Dirix, Laura Molenaar-Kuijsten, Ugur Sahin, Kit Wong, Patrick A. Ott, and Evelyna Derhovanessian
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Melanoma ,Medizin ,Locally advanced ,Specific immunotherapy ,Cancer ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cancer immunotherapy ,Atezolizumab ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Prior Immunotherapy ,In patient ,business - Abstract
Background: Neoantigens arising from somatic mutations are attractive targets for cancer immunotherapy as they may be recognized as foreign by the immune system. RO7198457, a systemically administered RNA-Lipoplex iNeST was designed to stimulate T cell responses against neoantigens. A first-in-human Phase Ib study of RO7198457, in combination with the aPD-L1 antibody atezolizumab is being conducted in patients with locally advanced or metastatic solid tumors. Methods: RO7198457 is manufactured on a per-patient basis and contains up to 20 tumor-specific neoepitopes. Nine doses of RO7198457 were administered i.v. in weekly and bi-weekly intervals during the 12-week induction stage and every 24 weeks during the maintenance stage. Atezolizumab 1200 mg was administered on Day 1 of each 21-day cycle. Results: In total, 132 patients enrolled in cohorts with doses ranging from 15-50 μg RO7198457 in combination with atezolizumab. Most common tumor types were NSCLC, TNBC, melanoma and CRC. The median number of prior therapies was 3 (range 1-11). 39% of patients received prior immunotherapy. Most patients had low levels of PD-L1 expression (93% patients with Citation Format: Juanita S. Lopez, Ross Camidge, Marco Iafolla, Sylvie Rottey, Martin Schuler, Matthew Hellmann, Ani Balmanoukian, Luc Dirix, Michael Gordon, Ryan Sullivan, Brian S. Henick, Charles Drake, Kit Wong, Patricia LoRusso, Patrick Ott, Lawrence Fong, Aglaia Schiza, Jeffery Yachnin, Christian Ottensmeier, Fadi Braiteh, Johanna Bendell, Rom Leidner, George Fisher, Guy Jerusalem, Laura Molenaar-Kuijsten, Marcus Schmidt, Scott A. Laurie, Raid Aljumaily, Achim Rittmeyer, Eelke Gort, Ignacio Melero, Lars Mueller, Rachel Sabado, Patrick Twomey, Jack Huang, Manesh Yadav, Jingbin Zhang, Felicitas Mueller, Evelyna Derhovanessian, Ugur Sahin, Özlem Türeci, Thomas Powles. A phase Ib study to evaluate RO7198457, an individualized Neoantigen Specific immunoTherapy (iNeST), in combination with atezolizumab in patients with locally advanced or metastatic solid tumors [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT301.
- Published
- 2020
29. Phase I results from a phase 1/2 multi-center study of nab-sirolimus combined with mFOLFOX6+bevacizumab (FB) as first-line (1L) therapy in patients (pts) with metastatic colorectal cancer (mCRC) with or without PTEN loss
- Author
-
Neil Desai, Shihe Hou, Fadi Braiteh, Anita N. Schmid, Sunil Sharma, Angela Tatiana Alistar, Carlos Becerra, Berta Grigorian, Marc R. Matrana, and E. Gabriela Chiorean
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Everolimus ,biology ,Bevacizumab ,Colorectal cancer ,business.industry ,First line ,medicine.disease ,Sirolimus ,Internal medicine ,Multi center study ,medicine ,biology.protein ,PTEN ,business ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
e16050 Background: The PI3K/mTOR pathway overactivation and loss of PTEN occur in 20-40% of mCRC pts. A recent phase 1/2 study in mCRC pts showed evidence that everolimus added to 1L SOC treatment improved outcomes, in particular in pts with PTEN loss (Gilcrease, 2019). This prospective, single-arm phase 1/2 study aims to identify the optimal dose and schedule of nab-sirolimus (ABI-009), an mTOR inhibitor, plus FB and evaluate the safety and preliminary efficacy of this regimen. Methods: Eligible pts have no prior therapy, ≥18 years old, ECOG performance status of 0-2. PTEN loss (by IHC) is evaluated for all pts. nab-Sirolimus IV starting dose and schedule was 30mg/m2 weekly for 3 weeks followed by a week of rest (on D1, 8, and 15 in a 28-day cycle, qw3/4) plus standard doses of FB on D1 and D15. Dose escalation to 45 and 60mg/m2 or de-escalation to 20 and 10mg/m2 followed the 3+3 design. Dose limiting toxicity (DLT) was assessed in cycle 1. Results: As of Jan 31, 2020, 24 pts were treated in the phase 1 study and 18 were evaluable for response. No DLTs were observed in the first 3 pts treated with nab-sirolimus at 30mg/m2 qw3/4; however, all 3 pts missed D8 doses (due to G2 thrombocytopenia and G3 neutropenia) and the cohort expanded to additional 3 pts for further observation. 5/6 pts missed D8 doses and a new dose cohort enrolled pts at 20mg/m2 qw3/4. No DLTs were observed and the cohort was expanded to 10 pts. A safety review of the first 18 pts observed that 9/18 pts missed doses on D8 and a new cohort was added to enroll pts at 20 mg/m2 q2w (D1 and D15, as for FB). This cohort enrolled 8 pts with no DLTs and represents the recommended-phase-2-dose (RP2D) and continues enrollment in phase 2. For all pts, 15/24 (63%) pts had G3-4 treatment-related adverse events (TRAEs); most common were neutropenia (6/24, 25%) and thrombocytopenia (4/24, 17%). At the RP2D 3/8 (38%) pts had G3-4 TRAEs (thrombocytopenia, weight loss, and hypertension, 1 pt each). Among 18 evaluable pts, best response was: 7/18 (39%) partial response, 10/18 (56%) stable disease, and 16/18 (89%) had tumor shrinkage. PTEN was assessed in 14 pts: 4/14 (29%) had PTEN loss, 2/4 (50%) PTEN loss pts responded, while 3/10 (30%) pts that were PTEN+ (WT) had a response. Conclusions: The RP2D of nab-sirolimus is 20mg/m2 q2w in combination with standard mFOLFOX+bevacizumab. The phase 2 portion of the study is ongoing. Clinical trial information: NCT03439462 .
- Published
- 2020
30. EV-202: A phase II study of enfortumab vedotin in patients with select previously treated locally advanced or metastatic solid tumors
- Author
-
Lajos Pusztai, Seema Rao Gorla, Fadi Braiteh, Chunzhang Wu, Justine Yang Bruce, and Joaquina Baranda
- Subjects
Cancer Research ,business.industry ,Locally advanced ,Enfortumab vedotin ,Phases of clinical research ,medicine.disease ,Transmembrane protein ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,In patient ,Non small cell ,business ,Previously treated ,030215 immunology - Abstract
TPS3647 Background: Nectin-4, a transmembrane cell adhesion protein, is highly expressed in urothelial carcinoma (UC), breast cancer (BC), non-small cell lung cancer (NSCLC), and gastroesophageal cancers (GEC); targeting Nectin-4 on these tumors may provide a novel treatment approach. Enfortumab vedotin (EV), an investigational human monoclonal antibody-drug conjugate, binds to Nectin-4 and upon internalization releases MMAE resulting in cell cycle arrest and cell death. Recently, EV received accelerated approval by the FDA for the treatment of adults with locally advanced/metastatic UC who previously received a PD-1 or PD-L1 inhibitor, and a platinum-containing chemotherapy in the neoadjuvant/adjuvant, locally advanced or metastatic setting. Use of EV in this study is investigational. Methods: This open-label phase 2 study (NCT04225117) will assess the efficacy and safety/tolerability of EV in patients (pts) with previously treated locally advanced/metastatic malignant solid tumors. Adult pts (~240) with histologically or cytologically confirmed disease and an ECOG ≤1 will be enrolled into 1 of 6 tumor-specific cohorts (Table), with ~40 pts each. While Nectin-4 expression is not required for enrollment, it is being tested retrospectively. Patients with active CNS metastases, grade ≥2 preexisting sensory or motor neuropathy, grade ≥3 immunotherapy-related hypothyroidism or panhypopituitarism, ongoing grade >3 immunotherapy-related AEs requiring high-dose steroids, or a history of uncontrolled diabetes mellitus within 3 months of the study will be excluded. All pts will receive EV 1.25 mg/kg IV on Days 1, 8, and 15 of each 28-day cycle until treatment discontinuation criteria are met; dose reductions/interruptions will be permitted. For all cohorts, the primary endpoint is investigator-assessed confirmed objective response rate (RECIST v1.1); secondary endpoints include duration of response, disease control rate, progression-free and overall survival, and safety/tolerability of EV. This study is recruiting as of February 2020. Clinical trial information: NCT04225117 . [Table: see text]
- Published
- 2020
31. Long-term Clinical Outcomes and Biomarker Analyses of Atezolizumab Therapy for Patients With Metastatic Triple-Negative Breast Cancer: A Phase 1 Study
- Author
-
Indrani Sarkar, Cathie Chung, Peter Schmid, Fadi Braiteh, Ehab Elgabry, Rita Nanda, William Grossman, Jean Pierre Delord, Carol O'Hear, Sara M. Tolaney, Marcella Fassò, Cristina Cruz, Leisha A. Emens, Ching-Wei Chang, Irene Kuter, Michael S. Gordon, Luciana Molinero, Philippe A. Cassier, and Joseph Paul Eder
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Time Factors ,Triple Negative Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Antineoplastic Agents, Immunological ,Atezolizumab ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Progression-free survival ,Neoplasm Metastasis ,Adverse effect ,Triple-negative breast cancer ,Aged ,Original Investigation ,Aged, 80 and over ,business.industry ,Middle Aged ,medicine.disease ,Progression-Free Survival ,United States ,Clinical trial ,Europe ,Oncology ,Tolerability ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,business - Abstract
Importance Atezolizumab (anti–programmed cell death ligand 1 [PD-L1]) is well tolerated and clinically active in multiple cancer types. Its safety and clinical activity in metastatic triple-negative breast cancer (mTNBC) has not been reported. Objective To evaluate the safety, clinical activity, and biomarkers associated with the use of single-agent atezolizumab in patients with mTNBC. Design, Setting, and Participants Women with mTNBC (defined by investigator assessment) were enrolled between January 2013 and February 2016 in a multicohort open-label, phase 1 study at US and European academic medical centers. Median follow-up was 25.3 months (range, 0.4-45.6 months). Eligible patients regardless of line of therapy had measurable disease by Response Evaluation Criteria in Solid Tumors, version 1.1; Eastern Cooperative Oncology Group performance status of 0 to 1; and a representative tumor sample for assessment of immune cell (IC) PD-L1 expression. Interventions Atezolizumab was given intravenously every 3 weeks until unacceptable toxic effects or loss of clinical benefit. Main Outcomes and Measures Primary outcome was safety and tolerability. Activity and exploratory outcomes included objective response rate (ORR), duration of response, progression-free survival (PFS), and overall survival (OS). Outcomes were assessed in all patients and in key patient subgroups. Results Among 116 evaluable patients (median age, 53 years [range, 29-82 years]), treatment-related adverse events occurred in 73 (63%); 58 (79%) were grade 1 to 2. Most adverse events occurred within the first treatment year. The ORRs were numerically higher in first-line (5 of 21 [24%]) than in second-line or greater patients (6 of 94 [6%]). Median duration of response was 21 months (range, 3 to ≥38 months). Median PFS was 1.4 (95% CI, 1.3-1.6) months by RECIST and 1.9 (95% CI, 1.4-2.5) months by irRC. In first-line patients, median OS was 17.6 months (95% CI, 10.2 months to not estimable). Patients with PD-L1 expression of at least 1% tumor-infiltrating ICs had higher ORRs and longer OS (12% [11 of 91]; 10.1 [95% CI, 7.0-13.8] months, respectively) than those with less than 1% ICs (0 of 21; 6.0 [95% CI, 2.6-12.6] months, respectively). High levels of ICs (>10%) were independently associated with higher ORRs and longer OS. Conclusions and Relevance Single-agent atezolizumab was well tolerated and provided durable clinical benefit in patients with mTNBC with stable or responding disease and in earlier lines of treatment. Trial Registration ClinicalTrials.gov identifier:NCT01375842
- Published
- 2018
32. Ramucirumab combined with FOLFOX as front-line therapy for advanced esophageal, gastroesophageal junction, or gastric adenocarcinoma: a randomized, double-blind, multicenter Phase II trial
- Author
-
Harry H. Yoon, Johanna C. Bendell, Philip A. Philip, D. M. Anderson, Fadi Braiteh, David Ferry, Ling Gao, Zev A. Wainberg, Edward Arrowsmith, Jonathan D. Schwartz, Yanzhi Hsu, Irfan Firdaus, Steven R. Alberts, Nancy L. Lewis, Allen Lee Cohn, and Yihuan Xu
- Subjects
0301 basic medicine ,Oncology ,Male ,Esophageal Neoplasms ,Organoplatinum Compounds ,Leucovorin ,Phases of clinical research ,Kaplan-Meier Estimate ,Gastroenterology ,0302 clinical medicine ,FOLFOX ,Antineoplastic Combined Chemotherapy Protocols ,education.field_of_study ,Antibodies, Monoclonal ,Hematology ,Esophageal cancer ,Middle Aged ,Corrigenda ,medicine.anatomical_structure ,Treatment Outcome ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Female ,Esophagogastric Junction ,Fluorouracil ,medicine.drug ,Adult ,medicine.medical_specialty ,Population ,Antibodies, Monoclonal, Humanized ,Gastroesophageal Junction ,Disease-Free Survival ,Ramucirumab ,Double blind ,03 medical and health sciences ,Gastric adenocarcinoma ,Double-Blind Method ,Stomach Neoplasms ,Internal medicine ,medicine ,Humans ,Esophagus ,education ,Aged ,business.industry ,Front line ,medicine.disease ,030104 developmental biology ,business ,Progressive disease - Abstract
This phase II trial examined the addition of ramucirumab, a vascular endothelial growth factor receptor-2 monoclonal antibody, to mFOLFOX6 as front-line therapy for patients with advanced gastric/GEJ or esophageal adenocarcinoma. A survival benefit was not observed in the ITT population, but an exploratory analysis suggested a potential benefit for ramucirumab in the gastric/GEJ cancer subgroup. Background We report the first randomized, Phase II trial of ramucirumab, an anti-vascular endothelial growth factor receptor-2 monoclonal antibody, as front-line therapy in patients with advanced adenocarcinoma of the esophagus or gastric/gastroesophageal junction (GEJ). Patients and methods Patients from the USA with advanced esophageal, gastric, or GEJ adenocarcinoma randomly received (1:1) mFOLFOX6 plus ramucirumab (8 mg/kg) or mFOLFOX6 plus placebo every 2 weeks. The primary end point was progression-free survival (PFS) with 80% power to detect a hazard ratio (HR) of 0.71 (one-sided α = 0.15). Secondary end points included evaluation of response and overall survival (OS); an exploratory ramucirumab exposure–response analysis was undertaken. Results Of 168 randomized patients, 52% of tumors were located in the stomach/GEJ and 48% in the esophagus. The trial did not meet the primary end point of PFS [6.4 versus 6.7 months, HR 0.98 (95% confidence interval 0.69–1.37)] or the secondary end point of OS (11.7 versus 11.5 months) in the intent-to-treat (ITT) population. Objective response rates (45.2% versus 46.4%) were similar between arms. Most Grade ≥3 toxicities did not differ significantly between arms, yet premature discontinuation of FOLFOX and ramucirumab (for reasons other than progressive disease) was more common among ramucirumab- versus placebo-treated patients. In an exploratory analysis that censored for premature discontinuation, the HR for PFS favored the ramucirumab arm (HR 0.76), particularly in patients with gastric/GEJ cancer. An exploratory exposure–response analysis indicated that patients with higher ramucirumab exposure had longer OS. Conclusion The addition of ramucirumab to front-line mFOLFOX6 did not improve PFS in the ITT population. Clinicaltrials.gov identifier NCT01246960.
- Published
- 2019
33. Abstract C095: Antitumor activity of STRO-002, a novel anti-folate receptor-α (FRα) antibody drug conjugate (ADC), in patient-derived xenograft (PDX) models and preliminary phase I dose escalation safety outcomes in patients with ovarian carcinoma (OC)
- Author
-
Erika Hamilton, Arturo Molina, Jennifer A. Smith, Michael Palumbo, John W. Moroney, Russell J. Schilder, Fadi Braiteh, Sami Diab, Cristina Abrahams, Shannon Matheny, Venita I. DeAlmeida, R. Wendel Naumann, Richard T. Penson, and Denise Uyar
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Antibody-drug conjugate ,Nausea ,business.industry ,Cancer ,Neutropenia ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Toxicity ,medicine ,Carcinoma ,Vomiting ,medicine.symptom ,Adverse effect ,business - Abstract
Introduction: FRα is overexpressed in 80% of OC and endometrial carcinoma (EC), with minimal expression in normal tissues. STRO-002 is a novel FRα-targeting ADC designed to circumvent limitations of current ADCs. STRO-002 was generated with Sutro’s cell-free antibody production system (XpressCF™) and site-specific conjugation (XpressCF+™) platform, which uses the non-natural amino acid pAMF. STRO-002 has a drug-antibody ratio (DAR) of 4 and contains the proprietary tubulin-targeting 3-aminophenyl hemiasterlin warhead SC209, a potent cytotoxin that is rapidly cleared and a weak substrate for efflux pumps. STRO-002 demonstrated potent cytotoxic activity in multiple FRα-positive tumor cell lines and significant therapeutic efficacy in OC xenograft models. Toxicology studies demonstrate dose-dependent and reversible neutropenia when STRO-002 is administered to cynomolgus monkeys. No ocular toxicity was observed. Herein we report additional preclinical efficacy in PDX models and preliminary safety experience of STRO-002 in patients (pts) with OC. PDX model methods: FRα expression was evaluated in EC PDX models by IHC. Single agent efficacy of 10 mg/kg STRO-002 was assessed in models with negative, low (+), medium (++) and high (+++) FRα expression. Phase 1 trial methods: STRO-002-GM1 is a first-in-human Phase I, open-label, multicenter, dose escalation (Part 1) study with dose expansion (Part 2) to identify the maximum tolerated dose (MTD), and recommended Phase II dose (RP2D) in pts with advanced, relapsed or refractory OC and EC. STRO-002 is given IV over an hour on Day 1 of each 21-day cycle until disease progression. Results: Statistically significant efficacy was observed in 60% of the FRα-positive PDX models tested, with STRO-002 response appearing to positively correlate with FRα expression levels. Though high FRα PDX models showed the highest response rates, some models with low and medium FRα also exhibited good responses. 5 pts with OC have completed at least one cycle (21 days) of STRO-002 at 3 dose levels: 0.5, 1.0, and 1.8 mg/kg and are evaluable for safety and toxicity. Median age is 63 (r, 52-64). Median ECOG performance status is 0 (r, 0-1). Median # of prior therapies is 6 (r, 2-8). Four pts have received PARP inhibitors. Median number of STRO-002 doses administered is 2 (r, 1-5). 3 additional pts have been treated at the 2.9 mg/kg dose level and continue in the first cycle. No grade ≥ 3 treatment-related adverse events (TRAEs) have been observed. 93% of AEs are grade 1 or 2. The most common TRAEs in ≥20 % of pts includes nausea, vomiting, abdominal pain, fatigue and insomnia. Four grade 3 events occurred in 2 pts after completion of the first cycle and were related to disease progression, including small intestine obstruction, gross hematuria, dehydration and vomiting. 2 pts have discontinued due to disease progression. 6 pts remain on treatment and dose escalation is ongoing. Conclusions: STRO-002 is the first ADC generated with cell-free protein synthesis technology and site-specific conjugation of a novel hemiasterlin linker-warhead to be tested in pts with solid tumors. The potent anti-tumor activity of STRO-002 in PDX models further supports the clinical development of this agent. The preliminary safety profile in 5 OC pts treated with STRO-002 is encouraging. MTD has not been reached, no DLTs have been observed and dose escalation is continuing. Updated results will be presented. This study is registered with clinicaltrials.gov identifier NCT03748186. Citation Format: Denise Uyar, Russell J Schilder, R Wendel Naumann, Fadi S Braiteh, Erika Hamilton, Sami Diab, John Moroney, Richard T Penson, Jennifer Smith, Cristina Abrahams, Michael Palumbo, Venita DeAlmeida, Shannon Matheny, Arturo Molina. Antitumor activity of STRO-002, a novel anti-folate receptor-α (FRα) antibody drug conjugate (ADC), in patient-derived xenograft (PDX) models and preliminary phase I dose escalation safety outcomes in patients with ovarian carcinoma (OC) [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr C095. doi:10.1158/1535-7163.TARG-19-C095
- Published
- 2019
34. Atezolizumab (MPDL3280A) Monotherapy for Patients With Metastatic Urothelial Cancer: Long-term Outcomes From a Phase 1 Study
- Author
-
Yohann Loriot, Nicholas J. Vogelzang, Constanze Kaiser, Rafael Morales-Barrera, Thomas Powles, Joseph Kim, Daniel P. Petrylak, Howard A. Burris, Marcella Fassò, Beiying Ding, Carol O'Hear, Joaquim Bellmunt, and Fadi Braiteh
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Metastatic Urothelial Carcinoma ,Time Factors ,Drug-Related Side Effects and Adverse Reactions ,Population ,Antibodies, Monoclonal, Humanized ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,Carcinoma ,medicine ,Humans ,Neoplasm Metastasis ,education ,Adverse effect ,Survival analysis ,Original Investigation ,Aged ,Aged, 80 and over ,education.field_of_study ,Carcinoma, Transitional Cell ,Dose-Response Relationship, Drug ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Survival Analysis ,030104 developmental biology ,Oncology ,Urinary Bladder Neoplasms ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Female ,Urothelium ,business ,Cohort study ,Follow-Up Studies - Abstract
Importance Atezolizumab (anti–programmed death ligand 1) has demonstrated safety and activity in advanced and metastatic urothelial carcinoma, but its long-term clinical profile remains unknown. Objective To report long-term clinical outcomes with atezolizumab therapy for patients with metastatic urothelial carcinoma. Design, Setting, and Participants Patients were enrolled in an expansion cohort of an ongoing, open-label, phase 1 study. Median follow-up was 37.8 months (range, >0.7 to 44.4 months). Enrollment occurred between March 2013 and August 2015 at US and European academic medical centers. Eligible patients had measurable disease per Response Evaluation Criteria in Solid Tumors version 1.1, Eastern Cooperative Oncology Group performance status 0 to 1, and a representative tumor sample. Programmed death ligand 1 expression on immune cells was assessed (VENTANA SP142 assay). Interventions Atezolizumab was given intravenously every 3 weeks until unacceptable toxic effects, protocol nonadherence, or loss of clinical benefit. Main Outcomes and Measures Primary outcome was safety. Secondary outcomes included objective response rate, duration of response, and progression-free survival. Response and overall survival were assessed in key baseline subgroups. Results Ninety-five patients were evaluable (72 [76%] male; median age, 66 years [range, 36-89 years]). Forty-five (47%) received atezolizumab as third-line therapy or greater. Nine patients (9%) had a grade 3 to 4 treatment-related adverse event, mostly within the first treatment year; no serious related adverse events were observed thereafter. One patient (1%) discontinued treatment due to a related event. No treatment-related deaths occurred. Responses occurred in 26% (95% CI, 18%-36%) of patients. Median duration of response was 22.1 months (range, 2.8 to >41.0 months), and median progression-free survival was 2.7 months (95% CI, 1.4-4.3 months). Median overall survival was 10.1 months (95% CI, 7.3-17.0 months); 3-year OS rate was 27% (95% CI, 17%-36%). Response occurred in 40% (95% CI, 26%-55%; n = 40) and 11% (95% CI, 4%-25%; n = 44) of patients with programmed death ligand 1 expression of at least 5% tumor-infiltrating immune cells (IC2/3) or less than 5% (IC0/1), respectively. Median overall survival in patients with IC2/3 and IC0/1 was 14.6 months (95% CI, 9.0 months to not estimable) and 7.6 months (95% CI, 4.7 to 13.9 months), respectively. Conclusions and Relevance Atezolizumab remained well tolerated and provided durable clinical benefit to a heavily pretreated metastatic urothelial carcinoma population in this long-term study. Trial Registration clinicaltrials.gov Identifier:NCT01375842
- Published
- 2018
35. Long-term effects of crizotinib in ALK-positive tumors (excluding NSCLC): A phase 1b open-label study
- Author
-
Yuankai Shi, Matthew H. Taylor, Sergey Orlov, Li Zhang, Keizo Horibe, Fadi Braiteh, Paulina Selaru, William Jeffrey Edenfield, Tae Min Kim, Jin Seok Ahn, Shang Ju Wu, Brian A. Van Tine, Kenji Tamura, Hui-Qiang Huang, Carlo Gambacorti-Passerini, Jolanda Paolini, Taito Esaki, Joseph Thaddeus Beck, Gambacorti Passerini, C, Orlov, S, Zhang, L, Braiteh, F, Huang, H, Esaki, T, Horibe, K, Ahn, J, Beck, J, Edenfield, W, Shi, Y, Taylor, M, Tamura, K, Van Tine, B, Wu, S, Paolini, J, Selaru, P, and Kim, T
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Adolescent ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Crizotinib ,Internal medicine ,hemic and lymphatic diseases ,Carcinoma, Non-Small-Cell Lung ,Neoplasms ,medicine ,ROS1 ,Anaplastic lymphoma kinase ,Humans ,Anaplastic Lymphoma Kinase ,Lung cancer ,Research Articles ,Aged ,business.industry ,Medullary thyroid cancer ,Cancer ,Hematology ,Middle Aged ,medicine.disease ,Lymphoma ,030104 developmental biology ,Treatment Outcome ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Mutation ,Female ,business ,Hematology, anaplastic lymphoma kinase creatine, kinasecrizotinib ,medicine.drug ,Research Article - Abstract
Crizotinib, an inhibitor of anaplastic lymphoma kinase (ALK), MET, and ROS1, is approved for treatment of patients with ALK‐positive or ROS1‐positive advanced non‐small‐cell lung cancer (NSCLC). However, ALK rearrangements are also implicated in other malignancies, including anaplastic large‐cell lymphoma and inflammatory myofibroblastic tumors (IMTs). In this ongoing, multicenter, single‐arm, open‐label phase 1b study (PROFILE 1013; NCT01121588), patients with ALK‐positive advanced malignancies other than NSCLC were to receive a starting dose of crizotinib 250 mg twice daily. Primary endpoints were safety and objective responses based on Response Evaluation Criteria in Solid Tumors version 1.1 or National Cancer Institute International Response Criteria. Forty‐four patients were enrolled (lymphoma, n = 18; IMT, n = 9; other tumors, n = 17). The objective response rate was 53% (95% confidence interval [CI], 28–77) for lymphoma, with 8 complete responses (CRs) and 1 partial response (PR); 67% (95% CI, 30–93) for IMTs, with 1 CR and 5 PRs; and 12% (95% CI, 2–36) for other tumors, with 2 PRs in patients affected by colon carcinoma and medullary thyroid cancer, respectively. The median duration of treatment was almost 3 years for patients with lymphoma and IMTs, with 2‐year progression‐free survival of 63% and 67%, respectively. The most common treatment‐related adverse events were diarrhea (45.5%) and vision disorders (45.5%), mostly grade 1. These findings indicate strong and durable activity of crizotinib in ALK‐positive lymphomas and IMTs. The safety profile was consistent with the known safety profile of crizotinib even with long‐term treatment.
- Published
- 2018
36. EP1.12-06 New Treatment Option for ES-SCLC: Patient Characteristics and Use of an Atezolizumab Regimen in the Real-World Setting
- Author
-
S. Lam, Alexander I. Spira, Maen A. Hussein, Davey B. Daniel, Steven L. McCune, T. Ton, Ivor John Percent, S. Morris, Aaron S. Mansfield, A. Johnson, J. Goldschmidt, Leora Horn, Tarek Mekhail, S. Whipple, R. Induru, Fadi Braiteh, G. Macvicar, and Ticiana A. Leal
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Regimen ,Oncology ,business.industry ,Atezolizumab ,medicine ,Patient characteristics ,Treatment options ,Intensive care medicine ,business - Published
- 2019
37. Efficacy of derazantinib (DZB) in patients (pts) with intrahepatic cholangiocarcinoma (iCCA) expressing FGFR2-fusion or FGFR2 mutations/amplifications
- Author
-
Lorenza Rimassa, N. Damjanov, Stephan Braun, Fadi Braiteh, Bassel F. El-Rayes, S. Lonardi, Sherrie Bhoori, M. Droz Dit Busset, Brian Schwartz, William P. Harris, Mikael Saulay, Monica Niger, Kyri Papadopoulos, Marc Engelhardt, Vincenzo Mazzaferro, Gianluca Masi, Walid L. Shaib, and Nicola Personeni
- Subjects
0301 basic medicine ,business.industry ,Disease progression ,Stock options ,Hematology ,Disease control ,Management ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Clinical investigation ,Honorarium ,%22">Fish ,Medicine ,In patient ,Until Disease Progression ,business - Abstract
Background FGFR2 gene aberrations (GA) include fusions and mutations/amplifications, with an estimated prevalence of approximately 15% and 5% in iCCA, respectively. While FGFR2 fusions are acknowledged oncogenic drivers, the oncogenic potential of FGFR2 mutations and amplifications is less well defined, due to ambiguous available preclinical and clinical data. In a non-comparative Phase 2a study (NCT01752920), the potent pan-FGFR inhibitor DZB was administered to iCCA pts expressing either FGFR2-fusion (N=29), FGFR2 mutations/amplifications (N=6) or no FGFR gene aberration (N=9). Here, we present a post-hoc analysis of outcomes of iCCA pts expressing FGFR mutations/amplifications (N=6) or no FGFR GA (N=9) as compared to previously reported data of iCCA pts (N=29) expressing FGFR2 fusions (Mazzaferro et al. 2018 BJC). Methods Pts received 300mg DZB QD PO. Eligibility criteria included a locally confirmed (FISH or NGS) testing of FGFR2 GA (fusions vs mutations/amplifications or no aberrations). Objective responses were determined per RECIST 1.1. Disease control rate (DCR) was defined by the summation of CR, PR and SD, and PFS, calculated from treatment initiation until disease progression or death. Results The table shows the efficacy outcomes in the 3 patient groups. Types of drug-related adverse events were similar across groups. No new safety signals were identified.Table721PTableiCCA patient groupFGFR2 fusion N=29FGFR2 mutation/ amplification N=6No FGFR2 genomic aberration N=9Objective response rate (RECIST 1.1)6 (21%)0 (0%)0 (0%)DCR (PR or SD)24 (83%)4 (67%)2 (22%)Any target lesion diameter reduction18 (62%)4 (67%)0 (0%)Median (95% CI) Progression free survival (PFS), months5.76.71.5(4.0-9.2)(1.0-14.7)(0.7 – NA)% PFS at 3 months83%67%0%% PFS at 6 months47%50%0% Conclusions Anti-tumor efficacy of DZB as measured by DCR and PFS in iCCA patients harboring an FGFR2 GA seems to be similar for patients with FGFR2 fusions and FGFR2 mutations/amplifications, while patients without any detectable FGFR GA appear to derive no benefit from DZB treatment. While the influence of prognostic variables still has to be confirmed, the anti-tumor efficacy of DZB seen in pts with FGFR2 GA other than fusions warrant further clinical investigation. Legal entity responsible for the study Basilea Pharmaceutica International Ltd. Funding Basilea Pharmaceutica International Ltd. Disclosure S. Braun: Full / Part-time employment: Basilea Pharmaceutica Int. Ltd.; Shareholder / Stockholder / Stock options: Ipsen. B. El-Rayes: Advisory / Consultancy: Merrimack; Advisory / Consultancy: BTG; Honoraria (self), Advisory / Consultancy: Bayer; Advisory / Consultancy: Loxo; Honoraria (self), Advisory / Consultancy: RTI Health Solutions; Honoraria (self), Speaker Bureau / Expert testimony: Lexicon; Speaker Bureau / Expert testimony, Research grant / Funding (institution): Bristol-Myers Squibb; Research grant / Funding (institution): Taiho Pharmaceutical; Research grant / Funding (institution): Boston Biomedical; Research grant / Funding (institution): Cleave Biosciences; Research grant / Funding (institution): Genentech; Research grant / Funding (institution): AVEO; Research grant / Funding (institution): Pfizer; Research grant / Funding (institution): Novartis; Research grant / Funding (institution): Hoosier Cancer Research Network; Research grant / Funding (institution): Five Prime Therapeutics; Research grant / Funding (institution): PPD; Research grant / Funding (institution): Merck; Research grant / Funding (institution): ICON Clinical Research. W.P. Harris: Advisory / Consultancy: Neo Therma; Advisory / Consultancy, Research grant / Funding (institution): Bayer; Advisory / Consultancy, Research grant / Funding (institution), Travel / Accommodation / Expenses: Eisai; Advisory / Consultancy, Research grant / Funding (institution): Exelixis; Advisory / Consultancy, Research grant / Funding (institution): Bristol-Myers Squibb; Research grant / Funding (institution): ArQule; Research grant / Funding (institution): Halozyme; Research grant / Funding (institution): Nordion; Research grant / Funding (institution): MedImmune; Research grant / Funding (institution): Agios. N. Damjanov: Advisory / Consultancy: Bayer; Advisory / Consultancy: Amgen; Advisory / Consultancy: Celgene; Honoraria (self): Sirtex Medical. L. Rimassa: Advisory / Consultancy: Bayer; Advisory / Consultancy: Sirtex Medical; Advisory / Consultancy: Italfarmaco; Advisory / Consultancy: Sanofi; Advisory / Consultancy, Travel / Accommodation / Expenses: ArQule; Advisory / Consultancy: Baxter; Advisory / Consultancy, Travel / Accommodation / Expenses: Ipsen; Advisory / Consultancy: Exelixis; Advisory / Consultancy: Amgen; Advisory / Consultancy: Incyte; Advisory / Consultancy: Celgene; Advisory / Consultancy: Eisai; Advisory / Consultancy: Lilly; Honoraria (self): AstraZeneca; Honoraria (self): AbbVie; Honoraria (self): Gilead Sciences; Honoraria (self): Roche. F. Braiteh: Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Amgen; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses, Shareholder / Stockholder / Stock options: Bristol-Myers Squibb; Advisory / Consultancy, Travel / Accommodation / Expenses, Shareholder / Stockholder / Stock options: Clovis Oncology; Shareholder / Stockholder / Stock options: Agios; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Ipsen; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses, Shareholder / Stockholder / Stock options: Insys Therapeutics; Travel / Accommodation / Expenses, Shareholder / Stockholder / Stock options: Tesaro; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Genentech/Roche; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony: Lilly; Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Incyte; Honoraria (self), Advisory / Consultancy, Travel / Accommodation / Expenses: Bayer; Advisory / Consultancy, Travel / Accommodation / Expenses: Sanofi; Advisory / Consultancy, Travel / Accommodation / Expenses: Regeneron; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: AstraZeneca; Advisory / Consultancy, Speaker Bureau / Expert testimony: Merck; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Celgene; Honoraria (self), Advisory / Consultancy, Travel / Accommodation / Expenses: Lexicon; Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Pfizer; Travel / Accommodation / Expenses: Novartis; Honoraria (self), Advisory / Consultancy, Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Boehringer Ingelheim; Honoraria (self), Speaker Bureau / Expert testimony, Travel / Accommodation / Expenses: Taiho Pharmaceutical; Travel / Accommodation / Expenses: Heron. S. Lonardi: Advisory / Consultancy, Research grant / Funding (self): Amgen; Advisory / Consultancy, Speaker Bureau / Expert testimony, Research grant / Funding (self): Merck Serono; Advisory / Consultancy, Speaker Bureau / Expert testimony: Lilly; Speaker Bureau / Expert testimony: Roche; Speaker Bureau / Expert testimony: Bristol-Myers Squibb; Speaker Bureau / Expert testimony: Servier. M. Engelhardt: Full / Part-time employment: Basilea Pharmaceutica Int. Ltd. M. Saulay: Full / Part-time employment: Basilea Pharmaceutica Int. Ltd. B. Schwartz: Leadership role, Shareholder / Stockholder / Stock options, Full / Part-time employment: ArQule; Advisory / Consultancy: CIRM. W.L. Shaib: Research grant / Funding (institution): ArQule; Research grant / Funding (institution): Lilly. V. Mazzaferro: Advisory / Consultancy: Terumo Europe NV; Speaker Bureau / Expert testimony: Ipsen AB. K.P. Papadopoulos: Advisory / Consultancy: Bayer; Advisory / Consultancy, Research grant / Funding (institution): ArQule; Research grant / Funding (institution): AbbVie; Research grant / Funding (institution): MedImmune; Research grant / Funding (institution): Daiichi Sankyo; Research grant / Funding (institution): Regeneron; Research grant / Funding (institution): Sanofi; Research grant / Funding (institution): ARMO BioSciences; Research grant / Funding (institution): Amgen; Research grant / Funding (institution): Calithera Biosciences; Research grant / Funding (institution): Curegenix; Research grant / Funding (institution): Incyte; Research grant / Funding (institution): Merck; Research grant / Funding (institution): Peloton Therapeutics; Research grant / Funding (institution): ADC Therapeutics; Research grant / Funding (institution): 3D Medicines; Research grant / Funding (institution): Formation Biologics; Research grant / Funding (institution): EMD Serono; Research grant / Funding (institution): Syros Pharmaceuticals; Research grant / Funding (institution): Mersana; Research grant / Funding (institution): OncoMed. All other authors have declared no conflicts of interest.
- Published
- 2019
38. Phase 3 (COSMIC-312) study of cabozantinib in combination with atezolizumab vs sorafenib in patients with advanced hepatocellular carcinoma (aHCC) who have not received previous systemic anticancer therapy
- Author
-
R. Sinha, Anne Borgman, Lorenza Rimassa, Fawzi Benzaghou, Fadi Braiteh, S. Hazra, Robin Katie Kelley, A-L Cheng, Andrew X. Zhu, and Z. Kayali
- Subjects
0301 basic medicine ,Sorafenib ,Oncology ,medicine.medical_specialty ,Cabozantinib ,business.industry ,Immune checkpoint inhibitors ,Hematology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Atezolizumab ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,Tumor growth ,In patient ,Merck Sharp & Dohme ,Multiple tumors ,business ,medicine.drug - Abstract
Background C inhibits tyrosine kinases involved in tumor growth, angiogenesis, and immune regulation, including MET, VEGFR, and TAM kinases (Tyro3, AXL, MER). C is approved in the United States and Europe for treatment of aHCC after prior S based on improved overall survival (OS) vs placebo in the phase 3 CELESTIAL trial (Abou-Alfa NEJM 2018). C may promote an immune-permissive tumor environment, which could enhance response to immune checkpoint inhibitors. C is being evaluated in combination with the anti-PD-L1 antibody A in multiple tumor types including HCC in a phase 1 study; recommended dose, preliminary clinical activity, and safety of the combination have been established in aRCC (Agarwal Ann Oncol 2018). A in combination with bevacizumab, an anti-VEGF antibody, has shown preliminary clinical activity in first-line aHCC (Pishvaian Ann Oncol 2018). Here, we present the study design of a phase 3 trial of C + A vs S in pts with aHCC who have not received prior systemic therapy. Trial design This global, randomized, open-label phase 3 trial (NCT03755791) is evaluating the efficacy and safety of C + A vs S as first-line treatment for aHCC. C vs S will also be evaluated as a secondary endpoint. Eligibility criteria include age ≥18 years, BCLC stage B or C, Child-Pugh A, ECOG PS ≤ 1, and measurable disease per RECIST 1.1. Pts are randomized 2:1:1 to an experimental arm of C (40 mg qd) + A (1200 mg infusion q3w), a control arm of S (400 mg bid), and a C monotherapy arm (60 mg qd). 740 pts are planned to be enrolled at ∼250 sites globally. Randomization is stratified by disease etiology (HBV [with or without HCV], HCV [without HBV], or other), region (Asia, other), and the presence of extrahepatic disease and/or macrovascular invasion (yes, no). OS and progression-free survival (PFS) for C+A vs S are primary endpoints, and PFS for C vs S is a secondary endpoint. Additional endpoints include safety, pharmacokinetics, and correlation of biomarker analyses with clinical outcomes. The first patient was enrolled in December 2018, and enrollment is ongoing. Clinical trial identification NCT03755791 (Other Study ID Numbers: XL184-312). Legal entity responsible for the study Exelixis. Funding Exelixis. Disclosure T. Yau: Advisory / Consultancy: Exelixis; Honoraria (self): Ipsen, Exelixis. L. Rimassa: Advisory / Consultancy: Lilly, Bayer, Baxter, Sirtex Medical, Italfarmaco, Sanofi, ArQule, Baxter, Ipsen, Exelixis, Amgen, Celgene, Eisai, Hengrui, MSD; Honoraria (self): AstraZeneca, AbbVie, Gilead, Roche; Travel / Accommodation / Expenses: ArQule, Ipsen. A-L. Cheng: Advisory / Consultancy, Advisory Board: Bayer Schering Pharma, Bristol-Myers Squibb, Eisai, Merck Serono, Novartis, Ono Pharmaceutical, Exelixis, Nucleix Ltd., Roche/Genentech, and IQVIA; Advisory / Consultancy, Consultancy: Bayer Schering Pharma, Bristol-Myers Squibb, Eisai, Merck Serono, Novartis, Ono Pharmaceutical, Exelixis, Nucleix Ltd., Roche/Genentech, and IQVIA; Honoraria (self): Bayer, Eisai, and Merck Sharp Dohme, Merck Serono, Novartis, Ono Pharmaceutical, Roche/Genentech, and IQVIA. J-W. Park: Advisory / Consultancy, Advisory board: Astra-Zeneca, Ono, BMS, Bayer, Midatech; Research grant / Funding (self): Ono-BMS, AstraZeneca, Blueprint, Roche, Eisai, Exelixis, Kowa; Advisory / Consultancy, Consultancy: Ono, Genetech, Roche, BMS, Bayer, Ipsen; Honoraria (self): Bayer, Ono, Eisai. F. Braiteh: Shareholder / Stockholder / Stock options: Agios; Bristol-Myers Squibb; Clovis Oncology; Insys Therapeutics; Tesaro; Honoraria (self): Abbott Nutrition; Amgen; ARIAD; AstraZeneca; Bayer; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene; Genentech/Roche; HERON; Incyte; Insys Therapeutics; Insys Therapeutics; Ipsen; Lexicon; Lilly; Taiho Pharmaceutical; Advisory / Consultancy: Ambry Genetics; Amgen; AstraZeneca; Bayer; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene; Clovis Oncology; Genentech/Roche; Incyte; Insys Therapeutics; Ipsen; Ipsen; Lexicon; Lilly; Merck; Merrimack; Pfizer; Regeneron; Sanofi; Speaker Bureau / Expert testimony: Amgen; AstraZeneca; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene; Genentech/Roche; Incyte; Insys Therapeutics; Ipsen; Lilly; Merck; Merck; Merrimack; Pfizer; Taiho Pharmaceutical; Travel / Accommodation / Expenses: - Amgen; AstraZeneca/MedImmune; Bayer; Bayer/Onyx; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene; Clovis Oncology; Exelixis; HERON; Incyte; Insys Therapeutics; Ipsen; Lexicon; Merrimack; Novartis; Pfizer; Regeneron; Roche/Genentech; Sanofi; Taiho Ph. F. Benzaghou: Full / Part-time employment: IPSEN; Shareholder / Stockholder / Stock options: IPSEN. P. Thuluvath: Honoraria (self): AbbVie; Gilead Sciences; Advisory / Consultancy: AbbVie; Eisai; Gilead Sciences; Speaker Bureau / Expert testimony: AbbVie; Gilead Sciences; Research grant / Funding (institution): AbbVie (Inst); Allergan (Inst); Conatus (Inst); Cymabay Therapeutics (Inst); Eisai (Inst); Sillajen (Inst); Target Pharmasolutions (Inst); Tobira Therapeutics (Inst); Zydus Pharmaceuticals (Inst). S. Hazra: Full / Part-time employment: Exelixis; Shareholder / Stockholder / Stock options: Exelixis. S. Milwee: Full / Part-time employment: Exelixis; Shareholder / Stockholder / Stock options: Exelixis. B. Tan: Research grant / Funding (institution): Exelixis. A.X. Zhu: Advisory / Consultancy: AstraZeneca; Bayer; Bristol-Myers Squibb; eisai; Exelixis; Lilly; Merck; Novartis; Sanofi; Research grant / Funding (institution): Bayer (Inst); Bristol-Myers Squibb (Inst); Lilly (Inst); Merck (Inst); Novartis (Inst). R.K. Kelley: Advisory / Consultancy, funding to institution: Agios, AstraZeneca, Bayer, BMS; Advisory / Consultancy, funding to self: IDMC: Genentech/Roche; Research grant / Funding (institution): Agios, AstraZeneca, Bayer, BMS, Eli Lilly, Exelixis, EMD Serono, Medimmune, Merck, Novartis, QED, Taiho. All other authors have declared no conflicts of interest.
- Published
- 2019
39. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine Versus Nab-Paclitaxel/Gemcitabine in Patients With Untreated, Metastatic Pancreatic Ductal Adenocarcinoma
- Author
-
Fadi Braiteh, Dimitrios Chondros, Mark M. Zalupski, Ping Jiang, Andrea Wang-Gillam, Jie Pu, Paul E. Oberstein, W. Wu, Paul S. Ritch, Tara Elisabeth Seery, Sihem Khelifa, Darren Sigal, William P. Harris, Nathan Bahary, Carrie Aldrich, Sunil R. Hingorani, Andrew Eugene Hendifar, Lei Zheng, and Andrea J. Bullock
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Time Factors ,Paclitaxel ,Phases of clinical research ,Hyaluronoglucosaminidase ,Deoxycytidine ,03 medical and health sciences ,0302 clinical medicine ,Fibrinolytic Agents ,Internal medicine ,Albumins ,Thromboembolism ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Carcinoma ,Biomarkers, Tumor ,Humans ,Progression-free survival ,Enoxaparin ,Hyaluronic Acid ,Aged ,Aged, 80 and over ,Tumor microenvironment ,business.industry ,Middle Aged ,medicine.disease ,Gemcitabine ,Progression-Free Survival ,Clinical trial ,Pancreatic Neoplasms ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,business ,Perfusion ,medicine.drug ,Carcinoma, Pancreatic Ductal - Abstract
Purpose Metastatic pancreatic ductal adenocarcinoma is characterized by excessive hyaluronan (HA) accumulation in the tumor microenvironment, elevating interstitial pressure and impairing perfusion. Preclinical studies demonstrated pegvorhyaluronidase alfa (PEGPH20) degrades HA, thereby increasing drug delivery. Patients and Methods Patients with previously untreated metastatic pancreatic ductal adenocarcinoma were randomly assigned to treatment with PEGPH20 plus nab-paclitaxel/gemcitabine (PAG) or nab-paclitaxel/gemcitabine (AG). Tumor HA levels were measured retrospectively using a novel affinity histochemistry assay. Primary end points were progression-free survival (PFS; overall) and thromboembolic (TE) event rate. Secondary end points included overall survival, PFS by HA level, and objective response rate. An early imbalance in TE events in the PAG arm led to a clinical hold; thereafter, patients with TE events were excluded and enoxaparin prophylaxis was initiated. Results A total of 279 patients were randomly assigned; 246 had HA data; 231 were evaluable for efficacy; 84 (34%) had HA-high tumors (ie, extracellular matrix HA staining ≥ 50% of tumor surface at any intensity). PFS was significantly improved with PAG treatment overall (hazard ratio [HR], 0.73; 95% CI, 0.53 to 1.00; P = .049) and for patients with HA-high tumors (HR, 0.51; 95% CI, 0.26 to 1.00; P = .048). In patients with HA-high tumors (PAG v AG), the objective response rate was 45% versus 31%, and median overall survival was 11.5 versus 8.5 months (HR, 0.96; 95% CI, 0.57 to 1.61). The most common treatment-related grade 3/4 adverse events with significant differences between arms (PAG v AG) included muscle spasms (13% v 1%), neutropenia (29% v 18%), and myalgia (5% v 0%). TE events were comparable after enoxaparin initiation (14% PAG v 10% AG). Conclusion This study met its primary end points of PFS and TE event rate. The largest improvement in PFS was observed in patients with HA-high tumors who received PAG. A similar TE event rate was observed between the treatment groups in stage 2 of the trial.
- Published
- 2017
40. Comparative effectiveness and resource utilization of nab-paclitaxel plus gemcitabine vs FOLFIRINOX or gemcitabine for the first-line treatment of metastatic pancreatic adenocarcinoma in a US community setting
- Author
-
Monika Parisi, Fadi Braiteh, Siyeon Park, Claudio Faria, Quanhong Ni, and Manish Patel
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,FOLFIRINOX ,business.industry ,Cancer ,Metastatic Pancreatic Adenocarcinoma ,medicine.disease ,Gemcitabine ,Discontinuation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tolerability ,Cancer Management and Research ,030220 oncology & carcinogenesis ,Internal medicine ,Cohort ,medicine ,business ,Adverse effect ,medicine.drug - Abstract
Fadi Braiteh,1 Manish B Patel,2 Monika Parisi,2 Quanhong Ni,2 Siyeon Park,2,3 Claudio Faria2 1Comprehensive Cancer Centers of Nevada, University of Nevada School of Medicine, Las Vegas, NV, 2Celgene Corporation, Summit, NJ, 3The Ohio State University, Columbus, OH, USA Introduction: Despite a clinically relevant, statistically significant survival benefit with nab-paclitaxel plus gemcitabine and FOLFIRINOX vs single-agent gemcitabine for metastatic pancreatic cancer (mPC), little is known regarding their real-world effectiveness. We analyzed patients with mPC using a nationally representative electronic medical records database to address this unmet need.Methods: This retrospective analysis of the Navigating Cancer database compared outcomes among patients who received first-line nab-paclitaxel plus gemcitabine, FOLFIRINOX, or gemcitabine for mPC. Effectiveness, safety, and supportive care use were examined. nab-Paclitaxel plus gemcitabine was the reference for statistical comparisons.Results: Baseline characteristics were similar except age (oldest patients were in the gemcitabine cohort followed by nab-paclitaxel plus gemcitabine, then FOLFIRINOX). Patients receiving nab-paclitaxel plus gemcitabine (n=122) demonstrated similar time to treatment discontinuation (TTD; median, 3.4 vs 3.8 months; P=0.947) and database persistence (DP; median, 8.6 vs 8.6 months; P=0.534) vs FOLFIRINOX (n=80); however, TTD (median, 3.4 vs 2.2 months; P
- Published
- 2017
41. Randomized Phase II Trial of Parsatuzumab (Anti-EGFL7) or Placebo in Combination with FOLFOX and Bevacizumab for First-Line Metastatic Colorectal Cancer
- Author
-
Mark Stroh, Maria Anderson, Jacek Jassem, Ira Gore, Eric Van Cutsem, Daniel S. Chen, Ilsung Chang, Herbert Hurwitz, Guillem Argiles, Roel Funke, Bruce McCall, Fernando Rivera, Priti S. Hegde, Zev A. Wainberg, Rocio Garcia-Carbonero, Niall C. Tebbutt, Eric Wakshull, Ina Rhee, Fadi Braiteh, and Weilan Ye
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Organoplatinum Compounds ,Colorectal cancer ,Leucovorin ,Endothelial Growth Factors ,law.invention ,0302 clinical medicine ,Randomized controlled trial ,FOLFOX ,law ,Antineoplastic Combined Chemotherapy Protocols ,Monoclonal ,80 and over ,Neoplasm Metastasis ,Humanized ,6.2 Cellular and gene therapies ,Cancer ,Aged, 80 and over ,Hazard ratio ,Antibodies, Monoclonal ,Middle Aged ,Antibodies, Anti-Idiotypic ,Colo-Rectal Cancer ,Gene Expression Regulation, Neoplastic ,Bevacizumab ,Anti-Idiotypic ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Female ,Fluorouracil ,Erratum ,Colorectal Neoplasms ,medicine.drug ,Biotechnology ,Adult ,medicine.medical_specialty ,EGF Family of Proteins ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antibodies, Monoclonal, Humanized ,Placebo ,Antibodies ,Disease-Free Survival ,03 medical and health sciences ,Folinic acid ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Aged ,Neoplastic ,business.industry ,Clinical Trial Results ,Prevention ,Calcium-Binding Proteins ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Oxaliplatin ,030104 developmental biology ,Gene Expression Regulation ,business ,Digestive Diseases - Abstract
Lessons LearnedThese negative phase II results for parsatuzumab highlight the challenges of developing an agent intended to enhance the efficacy of vascular endothelial growth factor inhibition without the benefit of validated pharmacodynamic biomarkers or strong predictive biomarker hypotheses. Any further clinical development of anti-EGFL7 is likely to require new mechanistic insights and biomarker development for antiangiogenic agents.BackgroundEGFL7 (epidermal growth factor-like domain 7) is a tumor-enriched vascular extracellular matrix protein that supports endothelial cell survival. This phase II trial evaluated the efficacy of parsatuzumab (also known as MEGF0444A), a humanized anti-EGFL7 IgG1 monoclonal antibody, in combination with modified FOLFOX6 (mFOLFOX6) (folinic acid, 5-fluorouracil, and oxaliplatin) bevacizumab in patients with previously untreated metastatic colorectal cancer (mCRC).MethodsOne-hundred twenty-seven patients were randomly assigned to parsatuzumab, 400 mg, or placebo, in combination with mFOLFOX6 plus bevacizumab, 5 mg/kg. Treatment cycles were repeated every 2 weeks until disease progression or unacceptable toxicity for a maximum of 24 months, with the exception of oxaliplatin, which was administered for up to 8 cycles.ResultsThe progression-free survival (PFS) hazard ratio was 1.17 (95% confidence interval [CI], 0.71–1.93; p = .548). The median PFS was 12 months for the experimental arm versus 11.9 months for the control arm. The hazard ratio for overall survival was 0.97 (95% CI, 0.46–2.1; p = .943). The overall response rate was 59% in the parsatuzumab arm and 64% in the placebo arm. The adverse event profile was similar in both arms.ConclusionsThere was no evidence of efficacy for the addition of parsatuzumab to the combination of bevacizumab and chemotherapy for first-line mCRC.
- Published
- 2017
42. Cabozantinib in hepatocellular carcinoma: results of a phase 2 placebo-controlled randomized discontinuation study
- Author
-
Yihua Lee, Robin Kate Kelley, H. A. Burris, Allen Lee Cohn, E. Van Cutsem, Wu Chou Su, P. Foster, Chris Verslype, Alexander I. Spira, T.-S. Yang, Nicholas J. Vogelzang, and Fadi Braiteh
- Subjects
Male ,Pyridines ,Kaplan-Meier Estimate ,Gastroenterology ,chemistry.chemical_compound ,0302 clinical medicine ,Clinical endpoint ,Medicine ,Anilides ,Cancer ,Liver Disease ,Liver Neoplasms ,Hematology ,hepatocellular carcinoma ,Middle Aged ,Sorafenib ,Oncology ,Response Evaluation Criteria in Solid Tumors ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,Female ,medicine.drug ,Liver Cancer ,Adult ,Niacinamide ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Cabozantinib ,Drug-Related Side Effects and Adverse Reactions ,overall survival ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Placebo ,Disease-Free Survival ,03 medical and health sciences ,Rare Diseases ,Double-Blind Method ,Clinical Research ,cabozantinib ,Internal medicine ,Humans ,Progression-free survival ,Oncology & Carcinogenesis ,Protein Kinase Inhibitors ,Aged ,tumor response ,business.industry ,Phenylurea Compounds ,Carcinoma ,Evaluation of treatments and therapeutic interventions ,Hepatocellular ,Discontinuation ,chemistry ,Liver function ,Digestive Diseases ,business ,progression-free survival ,vascular endothelial growth factor receptor - Abstract
BACKGROUND: Cabozantinib, an orally bioavailable inhibitor of tyrosine kinases including MET, AXL, and VEGF receptors, was assessed in patients with hepatocellular carcinoma (HCC) as part of a phase 2 randomized discontinuation trial with nine tumor-type cohorts. PATIENTS AND METHODS: Eligible patients had Child-Pugh A liver function and ≤1 prior systemic anticancer regimen, completed ≥4 weeks before study entry. The cabozantinib starting dose was 100 mg daily. After an initial 12-week cabozantinib treatment period, patients with stable disease (SD) per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.0 were randomized to cabozantinib or placebo. The primary endpoint of the lead-in stage was objective response rate (ORR) at week 12, and the primary endpoint of the randomized stage was progression-free survival (PFS). RESULTS: Among the 41 HCC patients enrolled, the week 12 ORR was 5%, with 2 patients achieving a confirmed partial response (PR). The week 12 disease control rate (PR or SD) was 66% (Asian subgroup: 73%). Of patients with ≥1 post-baseline scan, 78% had tumor regression, with no apparent relationship to prior sorafenib therapy. Alpha-fetoprotein (AFP) response (>50% reduction from baseline) occurred in 9 of the 26 (35%) patients with elevated baseline AFP and ≥1 post-baseline measurement. Twenty-two patients with SD at week 12 were randomized. Median PFS after randomization was 2.5 months with cabozantinib and 1.4 months with placebo, although this difference was not statistically significant. Median PFS and overall survival from Day 1 in all patients were 5.2 and 11.5 months, respectively. The most common grade 3/4 adverse events, regardless of attribution, were diarrhea (20%), hand-foot syndrome (15%), and thrombocytopenia (15%). Dose reductions were utilized in 59% of patients. CONCLUSIONS: Cabozantinib has clinical activity in HCC patients, including objective tumor responses, disease stabilization, and reductions in AFP. Adverse events were managed with dose reductions. TRIAL REGISTRATION NUMBER: NCT00940225. ispartof: Annals of Oncology vol:28 issue:3 pages:528-534 ispartof: location:England status: published
- Published
- 2017
43. P2.13-39 A Phase Ib Trial of the HSP90 Inhibitor AUY922 in Combination with Pemetrexed in Metastatic Non-Squamous, Non-Small Cell Lung Cancer Patients
- Author
-
Brad Adams, W. Lawler, D. Melancon, Jonathan W. Goldman, B. Telivala, Fadi Braiteh, Dorothy S. Martinez, Edward B. Garon, Benjamin P Jones, B DiCarlo, Xiaoyan Wang, Zorawar S. Noor, and K. Kennedy
- Subjects
Pulmonary and Respiratory Medicine ,Pemetrexed ,Oncology ,business.industry ,Non squamous ,Cancer research ,Medicine ,Non small cell ,business ,Lung cancer ,medicine.disease ,Hsp90 Inhibitor AUY922 ,medicine.drug - Published
- 2018
44. Abstract CT046: Predictive biomarkers and pharmacodynamic changes in tumor RNA expression in a phase I study of anti PD-1 monoclonal antibody PF-06801591
- Author
-
Vinicius Bonato, Fadi Braiteh, Siwen Hu-Lieskovan, Xiao Wang, Melissa Lynne Johnson, Juneko E. Grilley-Olson, Shobha Potluri, Alison Forgie, and Jeffrey Chou
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,biology ,medicine.drug_class ,business.industry ,Cancer ,Cell cycle ,Monoclonal antibody ,medicine.disease ,Internal medicine ,Gene expression ,Biopsy ,medicine ,biology.protein ,Immunohistochemistry ,Antibody ,Receptor ,business - Abstract
Introduction: PF-06801591 is a humanized IgG4 monoclonal antibody that binds to programmed cell death (PD-1) receptor, blocking its interaction with PD-1 ligands. In a phase I study (NCT02573259; dose-escalation) of 40 patients (pts) with multiple solid tumor types treated with PF-06891591 intravenously (IV) or subcutaneously (SC), 7 pts showed partial response. PF-06801591 was well tolerated at all doses. Biopsy samples were evaluated for predictive biomarkers and pharmacodynamic effects. Methods: Biopsy samples at baseline and on-treatment (IV:day 29; SC:day 36) were collected from 4 dose cohorts (IV:1, 3 and 10 mg/kg; SC:300 mg). Whole-exome sequencing (WES) and RNA-seq of biopsy tissue were used to estimate tumor mutation burden (TMB) and gene expression. Regression analysis was used to identify TMB/genes/pathways potentially associated with response in baseline tissue. Differential analysis of baseline and on-treatment biopsies identified genes/pathways potentially upregulated by PF-06801591. PD-L1 (clone SP-263; Ventana) expression was evaluated in tumor biopsies by immunohistochemistry (IHC) and pathologist scoring. Results: In a combined WES analysis of samples from baseline biopsies (all doses, n=24 pts), higher TMB was significantly associated with improved objective response (R^2=0.215, P=0.013). From RNA-seq analysis, genes positively associated with response were involved in interferon-gamma (IFNG) and PD-1 signaling, and cell cycle; CTLA4 was among the genes most significantly associated with response (P Conclusion: Based on a mixed set of tumor types with possible sensitivity to anti-PD-1, high TMB, IFNG signaling, and PD-L1 were associated with response to PF-06801591, as reported for other anti-PD-1 antibodies. Albeit based on a small sample size and diverse indication profile, baseline expression of CTLA4 appears to be highly associated with response and PD-L1 RNA levels may be better associated with response than protein expression by IHC. Increased expression of genes/pathways associated with adaptive immune activation in on-treatment biopsies indicates an active, immunomodulatory mechanism with anti-PD-1 therapy. This ongoing study will evaluate predictive biomarkers in dose-expansion cohorts of PF-06801591 in non-small cell lung cancer and urothelial cancer. Acknowledgments Study sponsor: Pfizer. Medical writing support: Chu Kong Liew (Engage Scientific Solutions), funded by Pfizer. Citation Format: Siwen Hu-Lieskovan, Fadi Braiteh, Juneko E. Grilley-Olson, Shobha Potluri, Xiao Wang, Alison Forgie, Vinicius Bonato, Jeffrey Chou, Melissa L. Johnson. Predictive biomarkers and pharmacodynamic changes in tumor RNA expression in a phase I study of anti PD-1 monoclonal antibody PF-06801591 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT046.
- Published
- 2019
45. Assessment of Subcutaneous vs Intravenous Administration of Anti–PD-1 Antibody PF-06801591 in Patients With Advanced Solid Tumors
- Author
-
Fadi Braiteh, Melissa Lynne Johnson, Ira Jacobs, Juneko E. Grilley-Olson, Siwen Hu-Lieskovan, Farhad Kazazi, Jasmine Davda, Jeffrey Chou, Ruifeng Li, and Alison Forgie
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Injections, Subcutaneous ,Programmed Cell Death 1 Receptor ,Gastroenterology ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Internal medicine ,Dose escalation ,Humans ,Medicine ,030212 general & internal medicine ,Dosing ,Adverse effect ,Aged ,biology ,business.industry ,Immunogenicity ,Antibodies, Monoclonal ,Middle Aged ,Antibodies, Neutralizing ,Clinical trial ,Treatment Outcome ,Oncology ,Immunoglobulin G ,030220 oncology & carcinogenesis ,Cohort ,biology.protein ,Administration, Intravenous ,Female ,Antibody ,business - Abstract
Importance We assessed feasibility of monthly subcutaneous administration of PF-06801591, a humanized immunoglobulin G4 monoclonal antibody that binds to the programmed cell death (PD-1) receptor and blocks its interaction with PD-1 ligands. Objective To evaluate the safety, efficacy, and pharmacokinetics of PF-06801591 administered intravenously vs subcutaneously. Design, Setting, and Participants Ongoing phase 1, open-label, multicenter, dose-escalation study of 40 patients, 18 years or older, with locally advanced or metastatic solid tumors, enrolled between March 8, 2016, and March 5, 2018, from 4 US medical centers. Interventions An intravenous dose of 0.5, 1, 3, or 10 mg/kg of PF-06801591 was administered every 3 weeks or a subcutaneous dose of 300 mg was administered every 4 weeks. Dose escalation occurred after 2 to 4 patients were enrolled per dose level, with additional patients enrolled in each cohort for further assessment. Main Outcomes and Measures The primary end points were dose-limiting toxic effects and safety. Secondary end points included pharmacokinetics, immunogenicity, PD-1 receptor occupancy, and efficacy. Results Of 40 enrolled patients (12 men and 28 women; mean [SD] age, 61 [13] years) in this phase 1 dose-escalation trial, 25 received PF-06801591 intravenously at escalating dose levels (0.5, 1, 3, or 10 mg/kg) and 15 patients received the monoclonal antibody subcutaneously at a single dose level. No dose-limiting toxic effects were observed. Grade 3 or higher treatment-related adverse events occurred in 4 (16%) patients treated intravenously and 1 (6.7%) patient treated subcutaneously. Immune-related adverse events occurred in 10 (40%) patients treated intravenously and 3 (20%) treated subcutaneously. No dose–adverse event associations were observed during intravenous dose escalation, and no serious skin toxic effects occurred with subcutaneous delivery. Responses were seen in 5 patients receiving PF-06801591 intravenously and in 2 patients treated subcutaneously for an overall objective response rate of 18.4%. Median overall survival was not reached with intravenous dosing vs 10.7 months with subcutaneous administration. Exposure to PF-06801591 increased in a dose-proportional manner over the range of intravenous doses. Median time to maximum observed serum concentration was 8 days after subcutaneous administration. Full PD-1 receptor occupancy was seen in all dose cohorts. Conclusions and Relevance Anti–PD-1 antibody PF-06801591 was tolerable and showed antitumor activity in a variety of tumor types across all dose levels of intravenous and subcutaneous administration. Monthly subcutaneous administration of PF-06801591 offers a convenient, effective alternative to currently available intravenously administered checkpoint inhibitors. Trial Registration ClinicalTrials.gov identifier:NCT02573259
- Published
- 2019
46. Phase 3 (COSMIC-312) study of cabozantinib (C) in combination with atezolizumab (A) versus sorafenib (S) in patients (pts) with advanced hepatocellular carcinoma (aHCC) who have not received previous systemic anticancer therapy
- Author
-
Joong-Won Park, Steven Milwee, Ann-Lii Cheng, Anthony B. El-Khoueiry, Lorenza Rimassa, Fawzi Benzaghou, Anne Borgman, Andrew X. Zhu, Fadi Braiteh, Robin Kate Kelley, and Zeid K Kayali
- Subjects
Sorafenib ,Cancer Research ,Cabozantinib ,Kinase ,Angiogenesis ,business.industry ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Oncology ,chemistry ,Atezolizumab ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,medicine ,Cancer research ,business ,Tyrosine kinase ,030215 immunology ,medicine.drug ,TYRO3 - Abstract
TPS4157 Background: C inhibits tyrosine kinases involved in tumor growth, angiogenesis, and immune regulation, including MET, VEGFR, and TAM kinases (Tyro3, AXL, MER). C is approved for treatment of aHCC after prior S based on improved overall survival (OS) vs placebo in the phase 3 CELESTIAL trial (Abou-Alfa NEJM 2018). Standard of care for first-line treatment of aHCC is tyrosine kinase inhibition with S or lenvatinib, and phase 3 trials of immune checkpoint inhibitors (ICIs) in first- and second- line aHCC are ongoing. C may promote an immune-permissive tumor environment, which could enhance response to ICIs. C is being evaluated in combination with the anti-PD-L1 antibody A in multiple tumor types including HCC in a phase 1 study; and dose, preliminary clinical activity, and safety have been established in aRCC (Agarwal Ann Oncol 2018). A in combination with bevacizumab, an anti-VEGF antibody, has shown preliminary clinical activity in first-line aHCC (Pishvaian Ann Oncol 2018). Here, we present the study design of a phase 3 trial of C+A vs S in pts with aHCC who have not received prior systemic therapy. Methods: This international, randomized, open-label phase 3 trial (NCT03755791) is evaluating the efficacy and safety of C+A vs S as first-line treatment for aHCC. Eligibility criteria include age ≥18 years, BCLC stage B or C, Child-Pugh A, ECOG PS 0 or 1, and measurable disease per RECIST 1.1. Patients are randomized 6:3:1 to an experimental arm of C (40 mg qd) + A (1200 mg infusion q3w), a control arm of S (400 mg bid), and an exploratory arm of C monotherapy (60 mg qd). 640 pts are planned at ~200 sites globally. Randomization is stratified by disease etiology (HBV [with or without HCV], HCV [without HBV], or other), region (Asia, other), and the presence of extrahepatic disease and/or macrovascular invasion (yes, no). OS and progression-free survival are coprimary endpoints and objective response rate is a secondary endpoint. Additional endpoints include safety, pharmacokinetics, and correlation of biomarker analyses with clinical outcomes. Enrollment in COSMIC-312 is ongoing. Clinical trial information: NCT03755791.
- Published
- 2019
47. Derazantinib (DZB) provides antitumor efficacy regardless of line of therapy in patients (pts) with FGFR2-fusion positive advanced intrahepatic cholangiocarcinoma (iCCA)
- Author
-
Brian Schwartz, Vincenzo Mazzaferro, Gianluca Masi, Nevena Damjanov, William P. Harris, Nicola Personeni, Lorenza Rimassa, Kyriakos P. Papadopoulos, Monica Niger, Mikael Saulay, Walid L. Shaib, Stephan Braun, Fadi Braiteh, Marc Engelhardt, Julia Kazakin, Sara Lonardi, Bassel F. El-Rayes, Sherrie Bhoori, and Michele Droz dit Busset
- Subjects
Cancer Research ,business.industry ,Line of therapy ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,In patient ,A kinase ,business ,Intrahepatic Cholangiocarcinoma ,030215 immunology - Abstract
e15607 Background: FGFR2 fusions are prevalent in 13-22% of iCCA and known oncogenic drivers. DZB is a kinase inhibitor with potent pan-FGFR activity. In a non-comparative Phase 2a study, DZB was administered to 29 pts with FGFR2-fusion positive advanced, inoperable iCCA, either as first-line (1L) (n = 2), 2L (n = 13), 3L (n = 10), 4L (n = 2) or 5L therapy (n = 2). The objective response rate (ORR) with DZB was 21%, disease-control rate (DCR) 83% and median PFS 5.7 months (Mazzaferro et al. 2018 BJC). Data from biliary tract cancer studies suggest decreasing treatment effects of chemotherapy with increasing lines of treatment. Here, we present a post-hoc analysis of outcomes of pts treated with DZB in 1L/2L (n = 15) compared to pts treated post-2L (n = 14). Methods: Pts received 300 mg DZB QD PO. Eligibility criteria included locally confirmed, positive testing of FGFR2 fusion expression (FISH or NGS), ECOG PS 0-1. Objective responses were determined using RECIST 1.1. Disease control rate was defined as CR, PR or SD. Results: The mean age of pts treated in 1L/2L was 66y and 55y in post-2L; 73% were females in 1L/2L and 50% in post-2L treatment; other demographic variables were balanced between groups (87% vs 86% of liver target lesions, median baseline lesion size of 97.5 mm vs 109.5 mm, ECOG PS0 was 60% vs 71%). Of 15 1L/2L group pts, 12 (80%) had prior platinum-based chemotherapy as compared to all 14 pts in the post-2L group. In the 1L/2L and post-2L groups, ORR was 20% and 21%, DCR was 80% and 86%, and a reduction in sum of the largest diameter of target lesions was observed in 60% and 64% of pts, respectively. Median PFS was 5.5 mo (95% CI, 1.9-11.9) and 6.2 mo (3.6-9.2) for the 1L/2L and post-2L groups, respectively. Types of drug-related adverse events were similar in 1L/2L and post-2L. Conclusions: Anti-tumor efficacy of DZB in iCCA patients measured either by ORR, DCR, tumor shrinkage or PFS was numerically similar irrespective of treatment line. These data suggest that DZB is an effective treatment option that can be applied early in the treatment continuum of iCCA patients or at later stages to offer anti-tumor efficacy and disease control. Clinical trial information: 01752920.
- Published
- 2019
48. Tumor mutational burden (TMB) may be a promising predictive biomarker of response to PD-1/PD-L1 targeting in MSI-H colorectal cancer
- Author
-
Leah Chase, May Thet Cho, Ethan Sokol, Fadi Braiteh, Jeffrey S. Ross, Marwan Fakih, Ching Ouyang, Joseph Chao, Steven Brad Maron, Vincent A. Miller, Samuel J. Klempner, Siraj M. Ali, Daniel V.T. Catenacci, Dean Lim, Alexa B. Schrock, and Jaideep Sandhu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,Colorectal cancer ,business.industry ,Pembrolizumab ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,PD-L1 ,Internal medicine ,medicine ,biology.protein ,In patient ,Nivolumab ,business ,030215 immunology ,Predictive biomarker - Abstract
43 Background: PD-1 targeting with pembrolizumab or nivolumab leads to durable clinical benefits in patients (pts) with microsatellite instability-high (MSI-H) tumors. However, 30-35% of mCRC pts with MSI-H tumors will experience progressive disease (PD) as a best response when treated with anti-PD1 agents, highlighting the need of additional predictive biomarkers. Methods: We performed a retrospective multi-center clinical investigation to evaluate the impact of TMB, age, gender, stage at initial presentation, pattern of metastatic disease, tumor grade, and RAS/RAF status on response to anti-PD1/PD-L1 in MSI-H mCRC. TMB and MSI status were determined by hybrid capture-based next-generation sequencing (Foundation Medicine [FM]). The TMB distribution in MSI-H CRC was estimated from a large data set from FM. Results: 22 eligible MSI-H mCRC pts were identified across 5 cancer centers: 19 pts received pembrolizumab, 1 pt received nivolumab, 1 pt received nivolumab + ipilimumab, and 1 pt received durvalumab + tremelimumab. Among tested variables, TMB (as a continuous variable) showed the strongest association with an objective response (OR; p < 0.001). Also, both univariate and multivariate analyses supported that TMB serves as an independent prognostic variable in predicting progression-free survival (PFS; p < 0.001 and p < 0.01, respectively). Using log-rank statistics, the optimal predictive cut-point for TMB was estimated between 37-41 mutations/Mb to dichotomize pts into TMBhigh and TMBlow groups. All 13 pts (100%) with TMBhigh had an OR, while only 2/9 (22%) pts with TMBlow had an OR and 6/9 had PD. The median PFS for TMBhigh pts has not been reached (no progressors, median follow-up > 18 mos), while the median PFS for TMBlow pts was 2 mos. Amongst 821 MSI-H CRC cases tested at FM, the 25th, 35th, 50th and 75th percentile TMB cutoffs were 33.1, 37.4, 46.1, and 61.8 mutations/Mb, respectively. Our optimal TMB cut-point range suggests that MSI-H mCRC with the lowest 35th percentile of TMB have a low likelihood of benefit from anti-PD1. Conclusions: These TMB findings require validation in prospective trials and may guide the sequencing of PD-1 inhibitor monotherapy in MSI-H mCRC.
- Published
- 2019
49. Safety, Tolerability, and Preliminary Activity of LB-100, an Inhibitor of Protein Phosphatase 2A, in Patients with Relapsed Solid Tumors: An Open-Label, Dose Escalation, First-in-Human, Phase I Trial
- Author
-
Vincent Chung, Henry J. Durivage, Fadi Braiteh, Donald A. Richards, Francis Johnson, John S. Kovach, Richard S. Ungerleider, and Aaron S. Mansfield
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Anemia ,Renal function ,Gastroenterology ,Drug Administration Schedule ,Piperazines ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Neoplasms ,medicine ,Humans ,Protein Phosphatase 2 ,Adverse effect ,Aged ,Neoplasm Staging ,Creatinine ,Dose-Response Relationship, Drug ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,Surgery ,030104 developmental biology ,Oncology ,Tolerability ,chemistry ,030220 oncology & carcinogenesis ,Toxicity ,Female ,business ,Hyponatremia - Abstract
Purpose: To determine the MTD and to assess the safety, tolerability, and potential activity of LB-100, a first-in-class small-molecule inhibitor of protein phosphatase 2A (PP2A) in adult patients with progressive solid tumors. Experimental Design: LB-100 was administered intravenously daily for 3 days in 21-day cycles in a 3 + 3 dose escalation design. Results: There were 29 patient entries over 7 dose escalations. One patient stopped treatment after one dose because of an acute infection and was reenrolled after recovery; each course was analyzed as a separate patient entry. Two patients had dose-limiting toxicity (reversible increases in serum creatinine or calculated serum creatinine clearance) at the 3.1 mg/m2 level. Probable or possible study drug-related grade 3 adverse events occurred in 6 (20.7%) patients [anemia (n = 2), decreased creatinine clearance, dyspnea, hyponatremia, and lymphopenia]. Ten (50%) of 20 response-evaluable patients had stable disease for four or more cycles. One patient with pancreatic adenocarcinoma had a partial response noted after 10 cycles, which was maintained for five additional cycles. The other patients achieving stable disease had one of the following: fibrosarcoma, chondrosarcoma, thymoma, atypical carcinoid of lung, or ovarian, testicular, breast (n = 2), and prostate cancer. The recommended phase II dose of LB-100 is 2.33 mg/m2 daily for 3 days every 3 weeks. Conclusions: The safety, tolerability, preliminary evidence of antitumor activity, and novel mechanism of action of LB-100 support its continued development alone and in combination with other therapies. Clin Cancer Res; 23(13); 3277–84. ©2016 AACR.
- Published
- 2016
50. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial
- Author
-
Louis, Fehrenbacher, Alexander, Spira, Marcus, Ballinger, Marcin, Kowanetz, Johan, Vansteenkiste, Julien, Mazieres, Keunchil, Park, David, Smith, Angel, Artal-Cortes, Conrad, Lewanski, Fadi, Braiteh, Daniel, Waterkamp, Pei, He, Wei, Zou, Daniel S, Chen, Jing, Yi, Alan, Sandler, Achim, Rittmeyer, and Charles, Weissman
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Population ,Antineoplastic Agents ,Docetaxel ,Antibodies ,B7-H1 Antigen ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Humans ,education ,Lung cancer ,Survival rate ,Aged ,Aged, 80 and over ,education.field_of_study ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Surgery ,Europe ,Survival Rate ,030104 developmental biology ,Treatment Outcome ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,North America ,Female ,Taxoids ,Nivolumab ,PD-L1 inhibitor ,business ,medicine.drug - Abstract
Outcomes are poor for patients with previously treated, advanced or metastatic non-small-cell lung cancer (NSCLC). The anti-programmed death ligand 1 (PD-L1) antibody atezolizumab is clinically active against cancer, including NSCLC, especially cancers expressing PD-L1 on tumour cells, tumour-infiltrating immune cells, or both. We assessed efficacy and safety of atezolizumab versus docetaxel in previously treated NSCLC, analysed by PD-L1 expression levels on tumour cells and tumour-infiltrating immune cells and in the intention-to-treat population.In this open-label, phase 2 randomised controlled trial, patients with NSCLC who progressed on post-platinum chemotherapy were recruited in 61 academic medical centres and community oncology practices across 13 countries in Europe and North America. Key inclusion criteria were Eastern Cooperative Oncology Group performance status 0 or 1, measurable disease by Response Evaluation Criteria In Solid Tumors version 1.1 (RECIST v1.1), and adequate haematological and end-organ function. Patients were stratified by PD-L1 tumour-infiltrating immune cell status, histology, and previous lines of therapy, and randomly assigned (1:1) by permuted block randomisation (with a block size of four) using an interactive voice or web system to receive intravenous atezolizumab 1200 mg or docetaxel 75 mg/m(2) once every 3 weeks. Baseline PD-L1 expression was scored by immunohistochemistry in tumour cells (as percentage of PD-L1-expressing tumour cells TC3≥50%, TC2≥5% and50%, TC1≥1% and5%, and TC01%) and tumour-infiltrating immune cells (as percentage of tumour area: IC3≥10%, IC2≥5% and10%, IC1≥1% and5%, and IC01%). The primary endpoint was overall survival in the intention-to-treat population and PD-L1 subgroups at 173 deaths. Biomarkers were assessed in an exploratory analysis. We assessed safety in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov, number NCT01903993.Patients were enrolled between Aug 5, 2013, and March 31, 2014. 144 patients were randomly allocated to the atezolizumab group, and 143 to the docetaxel group. 142 patients received at least one dose of atezolizumab and 135 received docetaxel. Overall survival in the intention-to-treat population was 12·6 months (95% CI 9·7-16·4) for atezolizumab versus 9·7 months (8·6-12·0) for docetaxel (hazard ratio [HR] 0·73 [95% CI 0·53-0·99]; p=0·04). Increasing improvement in overall survival was associated with increasing PD-L1 expression (TC3 or IC3 HR 0·49 [0·22-1·07; p=0·068], TC2/3 or IC2/3 HR 0·54 [0·33-0·89; p=0·014], TC1/2/3 or IC1/2/3 HR 0·59 [0·40-0·85; p=0·005], TC0 and IC0 HR 1·04 [0·62-1·75; p=0·871]). In our exploratory analysis, patients with pre-existing immunity, defined by high T-effector-interferon-γ-associated gene expression, had improved overall survival with atezolizumab. 11 (8%) patients in the atezolizumab group discontinued because of adverse events versus 30 (22%) patients in the docetaxel group. 16 (11%) patients in the atezolizumab group versus 52 (39%) patients in the docetaxel group had treatment-related grade 3-4 adverse events, and one (1%) patient in the atezolizumab group versus three (2%) patients in the docetaxel group died from a treatment-related adverse event.Atezolizumab significantly improved survival compared with docetaxel in patients with previously treated NSCLC. Improvement correlated with PD-L1 immunohistochemistry expression on tumour cells and tumour-infiltrating immune cells, suggesting that PD-L1 expression is predictive for atezolizumab benefit. Atezolizumab was well tolerated, with a safety profile distinct from chemotherapy.F Hoffmann-La Roche/Genentech Inc.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.