503 results on '"Ghassan K. Abou-Alfa"'
Search Results
52. Supplementary Data from Serum Alpha-fetoprotein Levels and Clinical Outcomes in the Phase III CELESTIAL Study of Cabozantinib versus Placebo in Patients with Advanced Hepatocellular Carcinoma
- Author
-
Ghassan K. Abou-Alfa, Anthony B. El-Khoueiry, Ann-Lii Cheng, Rajesh Kaldate, David W. Markby, Vincenzo Dadduzio, Albert Tran, Vincent C. Tam, Jean-Frederic Blanc, Stephen L. Chan, Thomas Yau, Joong-Won Park, Philippe Merle, Lorenza Rimassa, Tim Meyer, and Robin Kate Kelley
- Abstract
Supplementary Data_Clean File
- Published
- 2023
53. Supp Figure S3 from Serum Alpha-fetoprotein Levels and Clinical Outcomes in the Phase III CELESTIAL Study of Cabozantinib versus Placebo in Patients with Advanced Hepatocellular Carcinoma
- Author
-
Ghassan K. Abou-Alfa, Anthony B. El-Khoueiry, Ann-Lii Cheng, Rajesh Kaldate, David W. Markby, Vincenzo Dadduzio, Albert Tran, Vincent C. Tam, Jean-Frederic Blanc, Stephen L. Chan, Thomas Yau, Joong-Won Park, Philippe Merle, Lorenza Rimassa, Tim Meyer, and Robin Kate Kelley
- Abstract
Figure S3. AFP response cutoff determination for overall survival by maximally selected rank statistics. Cutpoint determination plots showing (A) histogram distribution and (B) rank statistic plot of percent AFP change from baseline at Week 8 for patients in the cabozantinib group with baseline AFP {greater than or equal to}20 ng/mL. *Calculated value of 0.48%, rounded to 0% for all subsequent analyses.
- Published
- 2023
54. Supplementary Figure 1 from Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention
- Author
-
Ghassan K. Abou-Alfa, Leonard B. Saltz, David S. Klimstra, David M. Hyman, Nikolaus Schultz, David B. Solit, Jaclyn F. Hechtman, Andrea Cercek, Michael I. D'Angelica, James J. Harding, William R. Jarnagin, Imane El-Dika, Eileen M. O'Reilly, Nancy E. Kemeny, Marinela Capanu, Ahmet Zehir, Michael F. Berger, Emmet Jordan, Ryan Ptashkin, and Maeve A. Lowery
- Abstract
Overall survival from diagnosis with advanced disease by KRAS, ERBB2 and CDKN2A/B status
- Published
- 2023
55. Supplemental Table 1 from Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies
- Author
-
Ghassan K. Abou-Alfa, Nikolaus Schultz, David B. Solit, Yelena Y. Janjigian, David S. Klimstra, William Jarnagin, David M. Hyman, Michael F. Berger, Michael I. D'Angelica, T. Peter Kingham, Yichao Sun, Richard K. Do, Imane El Dika, Ritika Kundra, Jaclyn F. Hechtman, Jinru Shia, Michele Ly, Melanie Albano, Danny N. Khalil, Joshua Armenia, Subhiksha Nandakumar, and James J. Harding
- Abstract
MSK IMPACT Gene Panels
- Published
- 2023
56. Supplementary from Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection
- Author
-
Eileen M. O'Reilly, Nadeem Riaz, Christine A. Iacobuzio-Donahue, Jorge S. Reis-Filho, Simon N. Powell, Timothy A. Chan, Nikolaus Schultz, Michael F. Berger, Liying Zhang, Mark E. Robson, Ghassan K. Abou-Alfa, David P. Kelsen, Nicolas Lecomte, Xin Pei, Pier Selenica, Vladimir Makarov, Sree B. Chalasani, Zoe McKinnell, Nima Ghalehsari, James J. Harding, Danny N. Khalil, Imane El Dika, Caitlin A. McIntyre, Vinod Balachandran, Marinela Capanu, Winston Wong, Kenneth H. Yu, Anna M. Varghese, Joanne F. Chou, Jiapeng Chen, and Wungki Park
- Abstract
Supplementary Data 1-8
- Published
- 2023
57. Supplemental Figure 1 from Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies
- Author
-
Ghassan K. Abou-Alfa, Nikolaus Schultz, David B. Solit, Yelena Y. Janjigian, David S. Klimstra, William Jarnagin, David M. Hyman, Michael F. Berger, Michael I. D'Angelica, T. Peter Kingham, Yichao Sun, Richard K. Do, Imane El Dika, Ritika Kundra, Jaclyn F. Hechtman, Jinru Shia, Michele Ly, Melanie Albano, Danny N. Khalil, Joshua Armenia, Subhiksha Nandakumar, and James J. Harding
- Abstract
Supplemental Figure 1: Distribution of tumor mutation burden (TMB) across different tumor types, comparing TMB in this HCC cohort (N=127) to the most common other tumor types in the MSK-IMPACT clinical sequencing cohort. The mutation rate was plotted as in (Zehir et al. Nat Med 2017).47
- Published
- 2023
58. Supplementary Figure Legends from A Phase Ib/II Study of Ramucirumab in Combination with Emibetuzumab in Patients with Advanced Cancer
- Author
-
Johanna C. Bendell, Volker Wacheck, Arantxa Uruñuela, Brian A. Moser, Xuejing Aimee Wang, Sameera R. Wijayawardana, Ghassan K. Abou-Alfa, Charles S. Fuchs, Martin H. Voss, Alexander Drilon, Toni K. Choueiri, Todd M. Bauer, Andrew X. Zhu, and James J. Harding
- Abstract
Supplementary figures titles and legends are placed in this document
- Published
- 2023
59. Data from Phase II Single-arm Study of Durvalumab and Tremelimumab with Concurrent Radiotherapy in Patients with Mismatch Repair–proficient Metastatic Colorectal Cancer
- Author
-
Leonard B. Saltz, Christopher H. Crane, Nancy Kemeny, Yoshiya Yamada, Stephen Solomon, Joseph Erinjeri, Marinela Capanu, Joanne F. Chou, Krishna Juluru, Travis J. Hollmann, Efsevia Vakiani, Taha Merghoub, Phillip Wong, Pallavi Vedantam, Aliya Holland, Matthew J. Adamow, Martinique Ogle, Mark L. Solter, Pamela Vaiskauskas, Kathleen C. Mcauliffe, Ghassan K. Abou-Alfa, David Faleck, Louise C. Connell, Rona Yaeger, Karuna Ganesh, Anna M. Varghese, Zsofia K. Stadler, Paul B. Romesser, Martin R. Weiser, T. Jonathan Yang, John Cuaron, Diane Reidy-Lagunes, Danny N. Khalil, Andreas Rimner, Abraham J. Wu, Geoffrey Ku, Andrea Cercek, and Neil H. Segal
- Abstract
Purpose:Immune checkpoint inhibition (ICI) alone is not active in mismatch repair–proficient (MMR-P) metastatic colorectal cancer (mCRC), nor does radiotherapy alone result in objective systemic benefit. However, combined radiotherapy plus ICI can induce systemic antitumor immunity in preclinical and clinical models.Patients and Methods:In this single-center, phase II study, patients with chemotherapy-refractory MMR-P mCRC received durvalumab 1,500 mg plus tremelimumab 75 mg every 4 weeks plus radiotherapy. The primary endpoint was objective response rate (ORR) in nonirradiated lesions. Treatment and efficacy were correlated with peripheral immune cell profiles.Results:We enrolled 24 patients, and report outcomes after a median follow-up of 21.8 (range: 15.9–26.3) months. The ORR was 8.3% (2 patients) [95% confidence interval (CI), 1.0–27.0]. The median progression-free survival was 1.8 (95% CI, 1.7–1.9) months, median overall survival was 11.4 (95% CI, 10.1–17.4) months. Twenty five percent of patients (n = 6) had treatment-related grade 3–4 adverse events. We observed increased circulating CD8+ T lymphocyte activation, differentiation, and proliferation in patients with objective response.Conclusions:This combination of radiotherapy plus ICI study did not meet the prespecified endpoint criteria to be considered worthwhile for further study. However, rare instances of systemic immune augmentation and regression in nonirradiated lesions were observed (an abscopal response). Combination durvalumab and tremelimumab plus radiotherapy is feasible in MMR-P mCRC with a manageable safety profile. Further studies of novel immunotherapy combinations, and identification of biomarkers predictive of abscopal response are warranted.
- Published
- 2023
60. Figure S1 from Phase II Single-arm Study of Durvalumab and Tremelimumab with Concurrent Radiotherapy in Patients with Mismatch Repair–proficient Metastatic Colorectal Cancer
- Author
-
Leonard B. Saltz, Christopher H. Crane, Nancy Kemeny, Yoshiya Yamada, Stephen Solomon, Joseph Erinjeri, Marinela Capanu, Joanne F. Chou, Krishna Juluru, Travis J. Hollmann, Efsevia Vakiani, Taha Merghoub, Phillip Wong, Pallavi Vedantam, Aliya Holland, Matthew J. Adamow, Martinique Ogle, Mark L. Solter, Pamela Vaiskauskas, Kathleen C. Mcauliffe, Ghassan K. Abou-Alfa, David Faleck, Louise C. Connell, Rona Yaeger, Karuna Ganesh, Anna M. Varghese, Zsofia K. Stadler, Paul B. Romesser, Martin R. Weiser, T. Jonathan Yang, John Cuaron, Diane Reidy-Lagunes, Danny N. Khalil, Andreas Rimner, Abraham J. Wu, Geoffrey Ku, Andrea Cercek, and Neil H. Segal
- Abstract
Study design
- Published
- 2023
61. Data from Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection
- Author
-
Eileen M. O'Reilly, Nadeem Riaz, Christine A. Iacobuzio-Donahue, Jorge S. Reis-Filho, Simon N. Powell, Timothy A. Chan, Nikolaus Schultz, Michael F. Berger, Liying Zhang, Mark E. Robson, Ghassan K. Abou-Alfa, David P. Kelsen, Nicolas Lecomte, Xin Pei, Pier Selenica, Vladimir Makarov, Sree B. Chalasani, Zoe McKinnell, Nima Ghalehsari, James J. Harding, Danny N. Khalil, Imane El Dika, Caitlin A. McIntyre, Vinod Balachandran, Marinela Capanu, Winston Wong, Kenneth H. Yu, Anna M. Varghese, Joanne F. Chou, Jiapeng Chen, and Wungki Park
- Abstract
Purpose:Genomic methods can identify homologous recombination deficiency (HRD). Rigorous evaluation of their outcome association to DNA damage response–targeted therapies like platinum in pancreatic ductal adenocarcinoma (PDAC) is essential in maximizing therapeutic outcome.Experimental Design:We evaluated progression-free survival (PFS) and overall survival (OS) of patients with advanced-stage PDAC, who had both germline- and somatic-targeted gene sequencing. Homologous recombination gene mutations (HRm) were evaluated: BRCA1, BRCA2, PALB2, ATM, BAP1, BARD1, BLM, BRIP1, CHEK2, FAM175A, FANCA, FANCC, NBN, RAD50, RAD51, RAD51C, and RTEL1 HRm status was grouped as: (i) germline versus somatic; (ii) core (BRCAs and PALB2) versus non-core (other HRm); and (iii) monoallelic versus biallelic. Genomic instability was compared using large-scale state transition, signature 3, and tumor mutation burden.Results:Among 262 patients, 50 (19%) had HRD (15% germline and 4% somatic). Both groups were analyzed together due to lack of difference in their genomic instability and outcome. Median [95% confidence interval (CI)] follow-up was 21.9 (1.4–57.0) months. Median OS and PFS were 15.5 (14.6–19) and 7 (6.1–8.1) months, respectively. Patients with HRD had improved PFS compared with no HRD when treated with first-line (1L) platinum [HR, 0.44 (95% CI: 0.29–0.67); P < 0.01], but not with 1L-non-platinum. Multivariate analysis showed HRD patients had improved OS regardless of their first-line treatment, but most had platinum exposure during their course. Biallelic HRm (11%) and core HRm (12%) had higher genomic instability, which translated to improved PFS on first-line platinum (1L-platinum) versus 1L-non-platinum.Conclusions:Pathogenic HRm identifies HRD in patients with PDAC with the best outcome when treated with 1L-platinum. Biallelic HRm and core HRm further enriched benefit from 1L-platinum from HRD.
- Published
- 2023
62. Supplementary Tables 1-3 from A Phase Ib/II Study of Ramucirumab in Combination with Emibetuzumab in Patients with Advanced Cancer
- Author
-
Johanna C. Bendell, Volker Wacheck, Arantxa Uruñuela, Brian A. Moser, Xuejing Aimee Wang, Sameera R. Wijayawardana, Ghassan K. Abou-Alfa, Charles S. Fuchs, Martin H. Voss, Alexander Drilon, Toni K. Choueiri, Todd M. Bauer, Andrew X. Zhu, and James J. Harding
- Abstract
Supplementary Tables 1-3
- Published
- 2023
63. Supplementary Legend from Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection
- Author
-
Eileen M. O'Reilly, Nadeem Riaz, Christine A. Iacobuzio-Donahue, Jorge S. Reis-Filho, Simon N. Powell, Timothy A. Chan, Nikolaus Schultz, Michael F. Berger, Liying Zhang, Mark E. Robson, Ghassan K. Abou-Alfa, David P. Kelsen, Nicolas Lecomte, Xin Pei, Pier Selenica, Vladimir Makarov, Sree B. Chalasani, Zoe McKinnell, Nima Ghalehsari, James J. Harding, Danny N. Khalil, Imane El Dika, Caitlin A. McIntyre, Vinod Balachandran, Marinela Capanu, Winston Wong, Kenneth H. Yu, Anna M. Varghese, Joanne F. Chou, Jiapeng Chen, and Wungki Park
- Abstract
Supplementary Data Legends
- Published
- 2023
64. Supplemental Table 2 from Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies
- Author
-
Ghassan K. Abou-Alfa, Nikolaus Schultz, David B. Solit, Yelena Y. Janjigian, David S. Klimstra, William Jarnagin, David M. Hyman, Michael F. Berger, Michael I. D'Angelica, T. Peter Kingham, Yichao Sun, Richard K. Do, Imane El Dika, Ritika Kundra, Jaclyn F. Hechtman, Jinru Shia, Michele Ly, Melanie Albano, Danny N. Khalil, Joshua Armenia, Subhiksha Nandakumar, and James J. Harding
- Abstract
MSK cohort compared to TCGA
- Published
- 2023
65. Data from A Phase Ib/II Study of Ramucirumab in Combination with Emibetuzumab in Patients with Advanced Cancer
- Author
-
Johanna C. Bendell, Volker Wacheck, Arantxa Uruñuela, Brian A. Moser, Xuejing Aimee Wang, Sameera R. Wijayawardana, Ghassan K. Abou-Alfa, Charles S. Fuchs, Martin H. Voss, Alexander Drilon, Toni K. Choueiri, Todd M. Bauer, Andrew X. Zhu, and James J. Harding
- Abstract
Purpose:Inhibition of the VEGFR-2 blocks angiogenesis and attenuates tumor growth, but cancers may evade this effect through activation of the hepatocyte growth factor receptor MET. Here we report results of the phase Ib/II study of ramucirumab, a monoclonal anti-VEGFR-2 antibody, plus the anti-MET mAb emibetuzumab.Patients and Methods:A 3+3 dose escalation of emibetuzumab plus ramucirumab (phase Ib) was followed by tumor-specific expansion cohorts. Primary objectives were to determine the recommended phase II dose and to evaluate antitumor activity. Secondary objectives included safety, pharmacokinetics, and immunogenicity. Tumoral MET expression was explored by immunohistochemistry (IHC).Results:A total of 97 patients with solid tumor [6 phase Ib, 16 gastric or gastroesophageal junction adenocarcinoma, 45 hepatocellular carcinoma (HCC), 15 renal cell carcinoma, and 15 non–small lung cancer] received emibetuzumab at 750 or 2,000 mg flat dosing plus ramucirumab at 8 mg/kg every 2 weeks. No dose-limiting toxicities were observed. Common adverse events were primarily mild or moderate and included fatigue (36.1%), peripheral edema (28.9%), and nausea (14.4%). Emibetuzumab exposures were similar as in previous studies with no apparent drug–drug interactions. Five partial responses (5.2%) were observed across all tumor types. The greatest antitumor activity was noted in HCC with a 6.7% overall response rate, 60% disease control rate, and 5.42 months (95% confidence interval, 1.64–8.12) progression-free survival (PFS). HCC with high MET expression showed improved PFS with approximately 3-fold increase in PFS (8.1 vs. 2.8 months) relative to low MET expression.Conclusions:Ramucirumab plus emibetuzumab was safe and exhibited cytostatic antitumor activity. MET expression may help to select patients benefitting most from this combination treatment in select tumor types.
- Published
- 2023
66. Supplemental Table 3 from Prospective Genotyping of Hepatocellular Carcinoma: Clinical Implications of Next-Generation Sequencing for Matching Patients to Targeted and Immune Therapies
- Author
-
Ghassan K. Abou-Alfa, Nikolaus Schultz, David B. Solit, Yelena Y. Janjigian, David S. Klimstra, William Jarnagin, David M. Hyman, Michael F. Berger, Michael I. D'Angelica, T. Peter Kingham, Yichao Sun, Richard K. Do, Imane El Dika, Ritika Kundra, Jaclyn F. Hechtman, Jinru Shia, Michele Ly, Melanie Albano, Danny N. Khalil, Joshua Armenia, Subhiksha Nandakumar, and James J. Harding
- Abstract
Outcomes to treatment based on genomic mutation
- Published
- 2023
67. Supplementary Figure Legends from Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection
- Author
-
Eileen M. O'Reilly, Nadeem Riaz, Christine A. Iacobuzio-Donahue, Jorge S. Reis-Filho, Simon N. Powell, Timothy A. Chan, Nikolaus Schultz, Michael F. Berger, Liying Zhang, Mark E. Robson, Ghassan K. Abou-Alfa, David P. Kelsen, Nicolas Lecomte, Xin Pei, Pier Selenica, Vladimir Makarov, Sree B. Chalasani, Zoe McKinnell, Nima Ghalehsari, James J. Harding, Danny N. Khalil, Imane El Dika, Caitlin A. McIntyre, Vinod Balachandran, Marinela Capanu, Winston Wong, Kenneth H. Yu, Anna M. Varghese, Joanne F. Chou, Jiapeng Chen, and Wungki Park
- Abstract
Supplementary Figure Legends
- Published
- 2023
68. Table S1 from Phase II Single-arm Study of Durvalumab and Tremelimumab with Concurrent Radiotherapy in Patients with Mismatch Repair–proficient Metastatic Colorectal Cancer
- Author
-
Leonard B. Saltz, Christopher H. Crane, Nancy Kemeny, Yoshiya Yamada, Stephen Solomon, Joseph Erinjeri, Marinela Capanu, Joanne F. Chou, Krishna Juluru, Travis J. Hollmann, Efsevia Vakiani, Taha Merghoub, Phillip Wong, Pallavi Vedantam, Aliya Holland, Matthew J. Adamow, Martinique Ogle, Mark L. Solter, Pamela Vaiskauskas, Kathleen C. Mcauliffe, Ghassan K. Abou-Alfa, David Faleck, Louise C. Connell, Rona Yaeger, Karuna Ganesh, Anna M. Varghese, Zsofia K. Stadler, Paul B. Romesser, Martin R. Weiser, T. Jonathan Yang, John Cuaron, Diane Reidy-Lagunes, Danny N. Khalil, Andreas Rimner, Abraham J. Wu, Geoffrey Ku, Andrea Cercek, and Neil H. Segal
- Abstract
Radiation treatment summary
- Published
- 2023
69. Supplemental Tables 1 - 3 and Figures 1 - 2 from Pharmacogenomic Modeling of Circulating Tumor and Invasive Cells for Prediction of Chemotherapy Response and Resistance in Pancreatic Cancer
- Author
-
Eileen M. O'Reilly, Mariola Sadowska, Rena Lapidus, Brandon Cooper, Kristen Gary, Joseph F. Crotty, Leonard B. Saltz, Maeve A. Lowery, Ghassan K. Abou-Alfa, Manuel Hidalgo, Mark Ricigliano, and Kenneth H. Yu
- Abstract
Supplementary Table 1: List of genes used for PGx modeling Supplementary Table 2: Correlation of PGx model prediction in tumor tissue to CTICs, soft tissue sarcoma. Supplementary Table 3. Normalized Enrichment Scores for chemotherapeutic agents modeled in three pancreatic cancer derived xenografts, referenced in (Figure 2). Supplementary Figure 1. Correlation of PGx model prediction in tumor tissue to CTICs, soft tissue sarcoma. Supplementary Figure 2. Correlation of Treatment Score to (A) PFS and (B) OS.
- Published
- 2023
70. Data from Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention
- Author
-
Ghassan K. Abou-Alfa, Leonard B. Saltz, David S. Klimstra, David M. Hyman, Nikolaus Schultz, David B. Solit, Jaclyn F. Hechtman, Andrea Cercek, Michael I. D'Angelica, James J. Harding, William R. Jarnagin, Imane El-Dika, Eileen M. O'Reilly, Nancy E. Kemeny, Marinela Capanu, Ahmet Zehir, Michael F. Berger, Emmet Jordan, Ryan Ptashkin, and Maeve A. Lowery
- Abstract
Purpose: Various genetic driver aberrations have been identified among distinct anatomic and clinical subtypes of intrahepatic and extrahepatic cholangiocarcinoma, and these molecular alterations may be prognostic biomarkers and/or predictive of drug response.Experimental Design: Tumor samples from patients with cholangiocarcinoma who consented prospectively were analyzed using the MSK-IMPACT platform, a targeted next-generation sequencing assay that analyzes all exons and selected introns of 410 cancer-associated genes. Fisher exact tests were performed to identify associations between clinical characteristics and genetic alterations.Results: A total of 195 patients were studied: 78% intrahepatic and 22% extrahepatic cholangiocarcinoma. The most commonly altered genes in intrahepatic cholangiocarcinoma were IDH1 (30%), ARID1A (23%), BAP1 (20%), TP53 (20%), and FGFR2 gene fusions (14%). A tendency toward mutual exclusivity was seen between multiple genes in intrahepatic cholangiocarcinoma including TP53:IDH1, IDH1:KRAS, TP53:BAP1, and IDH1:FGFR2. Alterations in CDKN2A/B and ERBB2 were associated with reduced survival and time to progression on chemotherapy in patients with locally advanced or metastatic disease. Genetic alterations with potential therapeutic implications were identified in 47% of patients, leading to biomarker-directed therapy or clinical trial enrollment in 16% of patients.Conclusions: Cholangiocarcinoma is a genetically diverse cancer. Alterations in CDKN2A/B and ERBB2 are associated with negative prognostic implications in patients with advanced disease. Somatic alterations with therapeutic implications were identified in almost half of patients. These prospective data provide a contemporary benchmark for guiding the development of targeted therapies in molecularly profiled cholangiocarcinoma, and support to the use of molecular profiling to guide therapy selection in patients with advanced biliary cancers. Clin Cancer Res; 24(17); 4154–61. ©2018 AACR.
- Published
- 2023
71. Data from Pharmacogenomic Modeling of Circulating Tumor and Invasive Cells for Prediction of Chemotherapy Response and Resistance in Pancreatic Cancer
- Author
-
Eileen M. O'Reilly, Mariola Sadowska, Rena Lapidus, Brandon Cooper, Kristen Gary, Joseph F. Crotty, Leonard B. Saltz, Maeve A. Lowery, Ghassan K. Abou-Alfa, Manuel Hidalgo, Mark Ricigliano, and Kenneth H. Yu
- Abstract
Purpose: Despite a challenging prognosis, modern cytotoxic therapy can induce tumor responses and extend life in pancreatic adenocarcinoma (PDAC). Pharmacogenomic (PGx) modeling of tumor tissue can predict the efficacy of chemotherapeutic agents in preclinical cancer models. We hypothesized that PGx profiling of circulating tumor and invasive cells (CTIC) isolated from peripheral blood could predict tumor response, progression, and resistance.Experimental Design: A PGx model was created and validated in preclinical models. A prospective clinical trial was conducted. Fifty patients with advanced PDAC were enrolled. Before treatment, 10 mL of peripherally drawn blood was collected. CTICs isolated from this blood sample were expression profiled and the PGx model was used to predict effective and ineffective chemotherapeutic agents. The treating physicians were blinded to PGx prediction.Results: We found that CTICs could be reliably isolated, total RNA extracted and profiled from 10 mL of peripheral blood from patients with unresectable PDAC before chemotherapy treatment and at disease progression. Using previously created PGx models to predict chemotherapy sensitivity, we found that clinical benefit was seen for study participants treated with chemotherapy regimens predicted to be effective versus chemotherapy regimens predicted to be ineffective with regard to progression-free (10.4 mo vs. 3.6 mo; P < 0.0001; HR, 0.14) and overall survival (17.2 mo vs. 8.3 mo; P < 0.0249; HR, 0.29).Conclusions: These findings suggest that PGx profiling of CTICs can predict treatment response. Clin Cancer Res; 20(20); 5281–9. ©2014 AACR.
- Published
- 2023
72. Association between posttreatment α‐fetoprotein reduction and outcomes in real‐world US patients with advanced hepatocellular carcinoma
- Author
-
Ghassan K. Abou‐Alfa, Xiaoliang Wang, Christina M. Parrinello, Anala Gossai, Richard Kim, Kelly Magee, and Rebecca A. Miksad
- Subjects
Cancer Research ,Oncology - Published
- 2023
73. First-in-Human Study of INCB062079, a Fibroblast Growth Factor Receptor 4 Inhibitor, in Patients with Advanced Solid Tumors
- Author
-
James J. Harding, Christiane Jungels, Jean-Pascal Machiels, David C. Smith, Chris Walker, Tao Ji, Ping Jiang, Xin Li, Ekaterine Asatiani, Eric Van Cutsem, Ghassan K. Abou-Alfa, UCL - SSS/IREC/MIRO - Pôle d'imagerie moléculaire, radiothérapie et oncologie, and UCL - (SLuc) Service d'oncologie médicale
- Subjects
Diarrhea ,EXPRESSION ,Cancer Research ,BILE-ACID SYNTHESIS ,Science & Technology ,Maximum Tolerated Dose ,IDENTIFICATION ,CASCADE ,POTENT ,Liver Neoplasms ,HEPATOCELLULAR CARCINOMAS ,Receptors, Fibroblast Growth Factor ,SIGNAL ,Bile Acids and Salts ,Oncology ,FXR ,Neoplasms ,FGFR4 ,Humans ,Pharmacology (medical) ,Female ,Receptor, Fibroblast Growth Factor, Type 4 ,Protein Kinase Inhibitors ,Life Sciences & Biomedicine ,SUPPRESSION - Abstract
INTRODUCTION: Fibroblast growth factor receptor (FGFR)-4/FGF19 pathway dysregulation is implicated in hepatobiliary and other solid tumors. INCB062079, an oral, selective, FGFR4 inhibitor, inhibits growth in FGF19/FGFR4-driven liver cancer models. METHODS: This was a two-part, phase I study (NCT03144661) in previously treated patients with advanced solid tumors. The primary objective was to determine safety, tolerability, and maximum tolerated dose (MTD), while secondary objectives included pharmacokinetics, pharmacodynamics (plasma FGF19; bile acid salts/7α-hydroxy-4-cholesten-3-one [C4] levels), and preliminary efficacy. In Part 1, patients received INCB062079 starting at 10 mg once daily, with 3 + 3 dose escalation. Part 2 (dose expansion) was not conducted because of study termination. RESULTS: Twenty-three patients were treated (hepatobiliary, n = 11; ovarian, n = 9; other, n = 3). Among six patients receiving 15 mg twice daily, two patients had dose-limiting toxicities (DLTs; grade 3 diarrhea, grade 3 transaminitis). Both had high pretreatment C4 concentrations, prompting a protocol amendment requiring pretreatment C4 concentrations
- Published
- 2023
74. Neo‐adjuvant FOLFIRINOX in borderline resectable and locally advanced pancreatic adenocarcinoma
- Author
-
Sally Temraz, Farah Nassar, Miza Salim Hammoud, Deborah Mukherji, Eileen M. O'Reilly, Haifa Dbouk, Fadi Farhat, Maya Charafeddine, Walid Faraj, Mohammad J. Khalifeh, Ghassan K. Abou‐Alfa, and Ali Shamseddine
- Subjects
Pancreatic Neoplasms ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Leucovorin ,Humans ,Prospective Studies ,Fluorouracil ,General Medicine ,Middle Aged ,Adenocarcinoma ,Neoadjuvant Therapy - Abstract
Surgery and systemic therapy provide the best option for long-term cancer control in localized resectable pancreas cancer. The present study assessed the efficacy and safety of neoadjuvant treatment with FOLFIRINOX in patients with borderline resectable (BR) and locally advanced (LA) pancreas cancer (PDAC).This was a prospective noninterventional observational trial of neoadjuvant FOLFIRINOX in BR and LA PDAC. The primary objective was the R0/R1 surgical resection rate. Secondary objectives included progression free survival (PFS) and overall survival (OS), tolerability, and toxicity.Forty-nine patients were enrolled between 2013 and 2019; the majority had LA disease (59.2%). Median age was 61 years, and median Ca 19-9 level pretreatment was 523.4 μmol/L. Following neoadjuvant FOLFIRINOX, 11 patients (22.5%) underwent surgical resection, the majority of which were BR at diagnosis (72.7%). Median OS and PFS for the entire group were 25 (95% CI: 17.2-32.8) and 12 months (95% CI: 9.7-13.3), respectively. Median PFS in BR patients was 14 (95% CI: 10.5-17.5) compared to 12 months (95% CI: 5.2-18.8) in patients with LA patients. Median OS and PFS were not reached in patients who underwent surgical resection as compared to 22 (95% CI: 18.6-25.4) and 9 months (95% CI: 4.2-13.9) in those who did not, respectively. Grade 3/4 neutropenia, leukopenia, neuropathy, nausea/vomiting, and diarrhea occurred in 6.3%, 2.1%, 10.4%, 4.2%, and 8.3%, respectively.Neoadjuvant FOLFIRINOX is an active regimen for patients with LA/BR PDAC with a resection rate of 22.5%. These results are in line with prior data.
- Published
- 2022
75. First-In-Human Effects of PPT1 Inhibition Using the Oral Treatment with GNS561/Ezurpimtrostat in Patients with Primary and Secondary Liver Cancers
- Author
-
James J. Harding, Ahmad Awada, Gael Roth, Thomas Decaens, Philippe Merle, Nuria Kotecki, Chantal Dreyer, Christelle Ansaldi, Madani Rachid, Soraya Mezouar, Agnes Menut, Eloïne Nadeige Bestion, Valérie Paradis, Philippe Halfon, Ghassan K. Abou-Alfa, and Eric Raymond
- Subjects
Oncology ,Hepatology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Research Article - Abstract
Introduction: GNS561/Ezurpimtrostat is a first-in-class, orally bioavailable, small molecule that blocks cancer cell proliferation by inhibiting late-stage autophagy and dose-dependent build-up of enlarged lysosomes by interacting with the palmitoyl-protein thioesterase 1 (PPT1). Methods: This phase I, open-label, dose-escalation trial (3 + 3 design) explored two GNS561 dosing schedules: one single oral intake 3 times a week (Q3W) and twice daily (BID) continuous oral administration in patients with advanced hepatocellular carcinoma, cholangiocarcinoma, and pancreatic adenocarcinoma or colorectal adenocarcinomas with liver metastasis. The primary objective was to determine GNS561 recommended phase II dose (RP2D) and schedule. Secondary objectives included evaluation of the safety/tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity of GNS561. Results: Dose escalation ranged from 50 to 400 mg Q3W to 200–300 mg BID. Among 26 evaluable patients for safety, 20 were evaluable for efficacy and no dose-limiting toxicity was observed. Adverse events (AEs) included gastrointestinal grade 1–2 events, primarily nausea and vomiting occurred in 13 (50%) and 14 (54%) patients, respectively, and diarrhea in 11 (42%) patients. Seven grade 3 AEs were reported (diarrhea, decreased appetite, fatigue, alanine aminotransferase, and aspartate aminotransferase increased). Q3W administration was associated with limited exposure and the BID schedule was preferred. At 200 mg BID GNS561, plasma and liver concentrations were comparable to active doses in animal models. Liver trough concentrations were much higher than in plasma a median time of 28 days of administration with a mean liver to plasma ratio of 9,559 (Min 149–Max 25,759), which is in accordance with rat preclinical data observed after repeated administration. PPT1 expression in cancer tissues in the liver was reduced upon GNS561 exposure. There was no complete or partial response. Five patients experienced tumor stable diseases (25%), including one minor response (−23%). Conclusion: Based on a favorable safety profile, exposure, and preliminary signal of activity, oral GNS561 RP2D was set at 200 mg BID. Studies to evaluate the antitumor activity of GNS561 in hepatocarcinoma cells and intrahepatic cholangiocarcinoma are to follow NCT 03316222.
- Published
- 2022
76. Nivolumab versus sorafenib in advanced hepatocellular carcinoma (CheckMate 459): a randomised, multicentre, open-label, phase 3 trial
- Author
-
Thomas Yau, Joong-Won Park, Richard S Finn, Ann-Lii Cheng, Philippe Mathurin, Julien Edeline, Masatoshi Kudo, James J Harding, Philippe Merle, Olivier Rosmorduc, Lucjan Wyrwicz, Eckart Schott, Su Pin Choo, Robin Kate Kelley, Wolfgang Sieghart, Eric Assenat, Renata Zaucha, Junji Furuse, Ghassan K Abou-Alfa, Anthony B El-Khoueiry, Ignacio Melero, Damir Begic, Gong Chen, Jaclyn Neely, Tami Wisniewski, Marina Tschaika, and Bruno Sangro
- Subjects
Male ,Carcinoma, Hepatocellular ,Nivolumab ,Oncology ,Liver Neoplasms ,Humans ,Female ,Middle Aged ,Sorafenib ,Aged - Abstract
Single-agent nivolumab showed durable responses, manageable safety, and promising survival in patients with advanced hepatocellular carcinoma in the phase 1-2 CheckMate 040 study. We aimed to investigate nivolumab monotherapy compared with sorafenib monotherapy in the first-line setting for patients with advanced hepatocellular carcinoma.In this randomised, open-label, phase 3 trial done at medical centres across 22 countries and territories in Asia, Australasia, Europe, and North America, patients at least 18 years old with histologically confirmed advanced hepatocellular carcinoma not eligible for, or whose disease had progressed after, surgery or locoregional treatment; with no previous systemic therapy for hepatocellular carcinoma, with Child-Pugh class A and Eastern Cooperative Oncology Group performance status score of 0 or 1, and regardless of viral hepatitis status were randomly assigned (1:1) via an interactive voice response system to receive nivolumab (240 mg intravenously every 2 weeks) or sorafenib (400 mg orally twice daily) until disease progression or unacceptable toxicity. The primary endpoint was overall survival assessed in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of study drug. This completed trial is registered with ClinicalTrials.gov, NCT02576509.Between Jan 11, 2016, and May 24, 2017, 743 patients were randomly assigned to treatment (nivolumab, n=371; sorafenib, n=372). At the primary analysis, the median follow-up for overall survival was 15·2 months (IQR 5·7-28·0) for the nivolumab group and 13·4 months (5·7-25·9) in the sorafenib group. Median overall survival was 16·4 months (95% CI 13·9-18·4) with nivolumab and 14·7 months (11·9-17·2) with sorafenib (hazard ratio 0·85 [95% CI 0·72-1·02]; p=0·075; minimum follow-up 22·8 months); the protocol-defined significance level of p=0·0419 was not reached. The most common grade 3 or worse treatment-related adverse events were palmar-plantar erythrodysaesthesia (1 [1%] of 367 patients in the nivolumab group vs 52 [14%] of patients in the sorafenib group), aspartate aminotransferase increase (22 [6%] vs 13 [4%]), and hypertension (0 vs 26 [7%]). Serious treatment-related adverse events were reported in 43 (12%) patients receiving nivolumab and 39 (11%) patients receiving sorafenib. Four deaths in the nivolumab group and one death in the sorafenib group were assessed as treatment related.First-line nivolumab treatment did not significantly improve overall survival compared with sorafenib, but clinical activity and a favourable safety profile were observed in patients with advanced hepatocellular carcinoma. Thus, nivolumab might be considered a therapeutic option for patients in whom tyrosine kinase inhibitors and antiangiogenic drugs are contraindicated or have substantial risks.Bristol Myers Squibb in collaboration with Ono Pharmaceutical.
- Published
- 2022
77. Mouse characteristics that affect establishing xenografts from hepatocellular carcinoma patient biopsies in the United States
- Author
-
Chenhui Zou, Imane El Dika, Koen O. A. Vercauteren, Marinela Capanu, Joanne Chou, Jinru Shia, Jill Pilet, Corrine Quirk, Gadi Lalazar, Linda Andrus, Mohammad Kabbani, Amin Yaqubie, Danny Khalil, Taha Mergoub, Luis Chiriboga, Charles M. Rice, Ghassan K. Abou‐Alfa, and Ype P. de Jong
- Subjects
endocrine system ,Cancer Research ,Carcinoma, Hepatocellular ,human alpha1‐antitrypsin ,endocrine system diseases ,Biopsy ,Liver Neoplasms ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Xenograft Model Antitumor Assays ,digestive system ,nitisinone ,United States ,digestive system diseases ,Fah−/− mice ,Disease Models, Animal ,Mice ,Oncology ,Animals ,Heterografts ,Humans ,Radiology, Nuclear Medicine and imaging ,hormones, hormone substitutes, and hormone antagonists ,RC254-282 ,PDX - Abstract
Background Hepatocellular carcinoma (HCC) patient‐derived xenograft (PDX) models hold potential to advance knowledge in HCC biology to help improve systemic therapies. Beside hepatitis B virus‐associated tumors, HCC is poorly established in PDX. Methods PDX formation from fresh HCC biopsies were obtained and implanted intrahepatically or in subrenal capsule (SRC). Mouse liver injury was induced in immunodeficient Fah−/− mice through cycling off nitisinone after HCC biopsy implantation, versus continuous nitisinone as non‐liver injury controls. Mice with macroscopically detectable PDX showed rising human alpha1‐antitrypsin (hAAT) serum levels, and conversely, no PDX was observed in mice with undetectable hAAT. Results Using rising hAAT as a marker for PDX formation, 20 PDX were established out of 45 HCC biopsy specimens (44%) reflecting the four major HCC etiologies most commonly identified at Memorial SloanKettering similar to many other institutions in the United States. PDX was established only in severely immunodeficient mice lacking lymphocytes and NK cells. Implantation under the renal capsule improved PDX formation two‐fold compared to intrahepatic implantation. Two out of 18 biopsies required murine liver injury to establish PDX, one associated with hepatitis C virus and one with alcoholic liver disease. PDX tumors were histologically comparable to biopsy specimens and 75% of PDX lines could be passaged. Conclusions Using cycling off nitisinone‐induced liver injury, HCC biopsies implanted under the renal capsule of severely immunodeficient mice formed PDX with 57% efficiency as determined by rising hAAT levels. These findings facilitate a more efficient make‐up of PDX for research into subset‐specific HCC.
- Published
- 2021
78. Exposure-Response Analyses of Tremelimumab Monotherapy or in Combination with Durvalumab in Patients with Unresectable Hepatocellular Carcinoma
- Author
-
Xuyang Song, Robin Kate Kelley, Anis A. Khan, Nathan Standifer, Diansong Zhou, KyoungSoo Lim, Rajesh Krishna, Lu Liu, Kun Wang, Patricia McCoon, Alejandra Negro, Philip He, Megan Gibbs, John F. Kurland, and Ghassan K. Abou-Alfa
- Subjects
Cancer Research ,Treatment Outcome ,Rare Diseases ,Oncology ,Clinical Research ,Carcinoma ,Liver Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Oncology and Carcinogenesis ,Humans ,Hepatocellular ,Oncology & Carcinogenesis ,Cancer - Abstract
Purpose:A novel single-dose regimen of 300 mg tremelimumab in combination with durvalumab [Single Tremelimumab Regular Interval Durvalumab (STRIDE)] has demonstrated a favorable benefit-risk profile in the phase I/II Study 22 (NCT02519348) and phase III HIMALAYA study (NCT03298451). This study evaluated the pharmacokinetics, exposure–response, and exposure–pharmacodynamics relationships of tremelimumab in patients with unresectable hepatocellular carcinoma (uHCC).Patients and Methods:A previous tremelimumab population pharmacokinetic model was validated using data from parts 2 and 3 of Study 22. Exposure–response analyses explored relationships of tremelimumab exposure with efficacy and safety. Pharmacokinetics and pharmacodynamics relationships were evaluated using linear and nonlinear regression models.Results:The observed pharmacokinetics of tremelimumab in uHCC were consistent with predictions; no significant covariates were identified. Tremelimumab exposure was not significantly associated with adverse events, objective response rate, or progression-free survival. Overall survival (OS) was longer for patients with tremelimumab exposure, minimum serum drug concentration (Cmin1) ≥ median versus Cmin1 < median (18.99 months vs. 10.97 months), but this exposure-survival analysis might be confounded with baseline characteristics of albumin level and neutrophil to lymphocyte ratio, which had a significant impact on OS (P = 0.0004 and 0.0001, respectively). The predicted Cmin1 of tremelimumab in STRIDE regimen (12.9 μg/mL) was greater than the estimated concentration of tremelimumab eliciting half-maximal increases (EC50 = 5.24 μg/mL) in CD8+Ki67+ T-cell counts.Conclusions:Our findings support novel insights into tremelimumab pharmacokinetics and exposure–response relationships in HCC and support the clinical utility of the STRIDE regimen in patients with uHCC.
- Published
- 2022
79. Liposomal irinotecan plus fluorouracil and leucovorin versus fluorouracil and leucovorin for metastatic biliary tract cancer after progression on gemcitabine plus cisplatin (NIFTY): a multicentre, open-label, randomised, phase 2b study
- Author
-
Myoung Joo Kang, Ghassan K. Abou-Alfa, Jaekyung Cheon, Kyu-Pyo Kim, Baek-Yeol Ryoo, Il Hwan Kim, Kyung Won Kim, Byung Woog Kang, Jae Ho Jeong, Ji Sung Lee, Hyewon Ryu, and Changhoon Yoo
- Subjects
Cisplatin ,medicine.medical_specialty ,business.industry ,Hazard ratio ,Neutropenia ,medicine.disease ,Gastroenterology ,digestive system diseases ,Gemcitabine ,Oncology ,Fluorouracil ,Internal medicine ,medicine ,Clinical endpoint ,Liposomal Irinotecan ,business ,Adverse effect ,medicine.drug - Abstract
Summary Background The prognosis of patients with advanced biliary tract cancer who have progressed on gemcitabine plus cisplatin is dismal. We aimed to investigate the efficacy and safety of second-line liposomal irinotecan plus fluorouracil and leucovorin in patients with metastatic biliary tract cancer that has progressed on gemcitabine plus cisplatin. Methods This multicentre, open-label, randomised, phase 2b (NIFTY) study was done at five academic institutions in South Korea and included patients aged 19 years or older with histologically or cytologically confirmed metastatic biliary tract cancer that had progressed on first-line gemcitabine plus cisplatin and an Eastern Cooperative Oncology Group performance status of 0 or 1. By use of an interactive web-based response system integrated with an electronic data capture system, patients were randomly assigned (1:1) using permuted blocks (block size 4) to receive either intravenous liposomal irinotecan (70 mg/m2 for 90 min) plus intravenous leucovorin (400 mg/m2 for 30 min) and intravenous fluorouracil (2400 mg/m2 for 46 h) every 2 weeks or leucovorin and fluorouracil only every 2 weeks, and were stratified by primary tumour site, previous surgery with curative intent, and participating centre. Study treatment was continued until the patient had disease progression or unacceptable toxicities, or withdrew consent. The primary endpoint was blinded independent central review (BICR)-assessed progression-free survival. The primary endpoint and safety were assessed in the full analysis set and the safety analysis set, respectively, both of which comprised all randomly assigned patients who received at least one dose of the study treatment. This trial is registered with ClinicalTrials.gov , NCT03524508 , and enrolment is complete. Findings Between Sept 5, 2018, and Feb 18, 2020, 193 patients were screened for eligibility, of whom 174 (88 in the liposomal irinotecan plus fluorouracil and leucovorin group and 86 in the fluorouracil plus leucovorin group) were enrolled and included in the full analysis and safety analysis sets. At a median follow-up of 11·8 months (IQR 7·7–18·7), the median BICR-assessed progression-free survival was significantly longer in the liposomal irinotecan plus fluorouracil and leucovorin group (7·1 months, 95% CI 3·6–8·8) than in the fluorouracil and leucovorin group (1·4 months, 1·2–1·5; hazard ratio 0·56, 95% CI 0·39–0·81; p=0·0019). The most common grade 3–4 adverse events were neutropenia (21 [24%] of 88 in the liposomal irinotecan plus fluorouracil and leucovorin group vs one [1%] of 86 in the fluorouracil and leucovorin group) and fatigue or asthenia (11 [13%] vs three [3%]). Serious adverse events occurred in 37 (42%) patients receiving liposomal irinotecan plus fluorouracil and leucovorin and 21 (24%) patients receiving fluorouracil and leucovorin. There were no treatment-related deaths. Interpretation Adding liposomal irinotecan to fluorouracil and leucovorin significantly improved BICR-assessed progression-free survival in patients with advanced biliary tract cancer. Liposomal irinotecan plus fluorouracil and leucovorin could be considered a standard-of-care second-line therapy for advanced biliary tract cancer. Funding Servier and HK inno.N Translation For the Korean translation of the abstract see Supplementary Materials section.
- Published
- 2021
80. Safety, Efficacy, and Pharmacodynamics of Tremelimumab Plus Durvalumab for Patients With Unresectable Hepatocellular Carcinoma: Randomized Expansion of a Phase I/II Study
- Author
-
Philip He, Robin Kate Kelley, Ho Yeong Lim, Takuji Okusaka, Thomas Yau, David Wai-Meng Tai, Ghassan K. Abou-Alfa, Masafumi Ikeda, Bruno Sangro, Yoon-Koo Kang, Silvia Damian, Johanna C. Bendell, Mitesh J. Borad, William P. Harris, Tae-You Kim, Shukui Qin, Alejandra Negro, Mallory Makowsky, Nathan Standifer, Masatoshi Kudo, Ann-Lii Cheng, Jordi Bruix, Antonio Gasbarrini, and Wei Peng Yong
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Durvalumab ,0302 clinical medicine ,Monoclonal ,80 and over ,Humanized ,6.2 Cellular and gene therapies ,Cancer ,Aged, 80 and over ,Liver Disease ,Liver Neoplasms ,Middle Aged ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,HIV/AIDS ,Female ,Patient Safety ,Biotechnology ,medicine.drug ,Liver Cancer ,Adult ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,medicine.drug_class ,Clinical Trials and Supportive Activities ,Clinical Sciences ,Oncology and Carcinogenesis ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Antibodies ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Aged ,business.industry ,Carcinoma ,Evaluation of treatments and therapeutic interventions ,Hepatocellular ,medicine.disease ,Orphan Drug ,030104 developmental biology ,Phase i ii ,Pharmacodynamics ,Digestive Diseases ,business ,Tremelimumab - Abstract
PURPOSE This phase I/II study evaluated tremelimumab (anticytotoxic T-lymphocyte–associated antigen-4 monoclonal antibody) and durvalumab (antiprogrammed death ligand-1 monoclonal antibody) as monotherapies and in combination for patients with unresectable hepatocellular carcinoma (HCC), including a novel regimen featuring a single, priming dose of tremelimumab (ClinicalTrials.gov identifier: NCT02519348 ). PATIENTS AND METHODS Patients with HCC who had progressed on, were intolerant to, or refused sorafenib were randomly assigned to receive T300 + D (tremelimumab 300 mg plus durvalumab 1,500 mg [one dose each during the first cycle] followed by durvalumab 1,500 mg once every 4 weeks), durvalumab monotherapy (1,500 mg once every 4 weeks), tremelimumab monotherapy (750 mg once every 4 weeks [seven doses] and then once every 12 weeks), or T75 + D (tremelimumab 75 mg once every 4 weeks plus durvalumab 1,500 mg once every 4 weeks [four doses] followed by durvalumab 1,500 mg once every 4 weeks). Safety was the primary end point. Secondary end points included objective response rate (ORR) by Response Evaluation Criteria in Solid Tumors v1.1 and overall survival; exploratory end points included circulating lymphocyte profiles. RESULTS A total of 332 patients were enrolled (T300 + D, n = 75; durvalumab, n = 104; tremelimumab, n = 69; and T75 + D, n = 84). Tolerability was acceptable across arms, with grade ≥ 3 treatment-related adverse events occurring in 37.8%, 20.8%, 43.5%, and 24.4%, respectively. Confirmed ORRs (95% CI) were 24.0% (14.9 to 35.3), 10.6% (5.4 to 18.1), 7.2% (2.4 to 16.1), and 9.5% (4.2 to 17.9), respectively. An early expansion of CD8+ lymphocytes was associated with response across arms, with highest proliferating CD8+ lymphocyte levels occurring in the T300 + D arm. The median (95% CI) overall survival was 18.7 (10.8 to 27.3), 13.6 (8.7 to 17.6), 15.1 (11.3 to 20.5), and 11.3 (8.4 to 15.0) months in the T300 + D, durvalumab, tremelimumab, and T75 + D arms, respectively. CONCLUSION All regimens were found to be tolerable and clinically active; however, the T300 + D regimen demonstrated the most encouraging benefit-risk profile. The unique pharmacodynamic activity and association with ORR of the T300 + D regimen further support its continued evaluation in HCC.
- Published
- 2021
81. Pertuzumab and trastuzumab for HER2-positive, metastatic biliary tract cancer (MyPathway): a multicentre, open-label, phase 2a, multiple basket study
- Author
-
John D. Hainsworth, Ghassan K. Abou-Alfa, Arisha Patel, Howard A. Burris, Christopher Sweeney, Ron Bose, Vaikunth Cuchelkar, David R. Spigel, Razelle Kurzrock, Katja Schulze, Nilofer S. Azad, Jonathan Levy, Milind Javle, Charles Swanton, Ben George, Claire F. Friedman, Funda Meric-Bernstam, Mitesh J. Borad, and Yong Wang
- Subjects
Male ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Receptor, ErbB-2 ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,Loading dose ,Antineoplastic Agents, Immunological ,Trastuzumab ,Internal medicine ,medicine ,Humans ,Progression-free survival ,Neoplasm Metastasis ,Adverse effect ,Survival rate ,Response Evaluation Criteria in Solid Tumors ,Aged ,business.industry ,Middle Aged ,Progression-Free Survival ,United States ,Survival Rate ,Regimen ,Biliary Tract Neoplasms ,Oncology ,Female ,Pertuzumab ,business ,medicine.drug - Abstract
Summary Background Systemic therapies for metastatic biliary tract cancers are few, and patients have a median overall survival of less than 1 year. MyPathway evaluates the activity of US Food and Drug Administration-approved therapies in non-indicated tumours with potentially actionable molecular alterations. In this study, we present an analysis of patients with metastatic biliary tract cancers with HER2 amplification, overexpression, or both treated with a dual anti-HER2 regimen, pertuzumab plus trastuzumab, from MyPathway. Methods MyPathway is a non-randomised, multicentre, open-label, phase 2a, multiple basket study. Patients aged 18 years and older with previously treated metastatic biliary tract cancers with HER2 amplification, HER2 overexpression, or both and an Eastern Cooperative Oncology Group performance status of 0–2 were enrolled from 23 study sites in the USA and received intravenous pertuzumab (840 mg loading dose, then 420 mg every 3 weeks) plus trastuzumab (8 mg/kg loading dose, then 6 mg/kg every 3 weeks). The primary endpoint was investigator-assessed objective response rate according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. The primary outcome and adverse events were analysed in all patients who received at least one dose of pertuzumab and trastuzumab. This trial is registered with ClinicalTrials.gov , NCT02091141 , and is ongoing. Findings 39 patients enrolled in the MyPathway HER2 biliary tract cancer cohort between Oct 28, 2014, and May 29, 2019, were evaluable for anti-tumour activity by the March 10, 2020, data cutoff date. Median follow-up was 8·1 months (IQR 2·7–15·7). Nine of 39 patients achieved a partial response (objective response rate 23% [95% CI 11–39]). Grade 3–4 treatment-emergent adverse events were reported in 18 (46%) of 39 patients, most commonly increased alanine aminotransferase and increased aspartate aminotransferase (each five [13%] of 39). Treatment-related grade 3 adverse events were reported in three (8%) of 39 patients, including increased alanine aminotransferase, aspartate aminotransferase, blood alkaline phosphatase, and blood bilirubin. Serious treatment-emergent adverse events were observed in ten (26%) of 39 patients, of which only abdominal pain occurred in more than one patient (two [5%] of 39). There were no treatment-related serious adverse events, treatment-related grade 4 events, or deaths. Interpretation Treatment was well tolerated in patients with previously treated HER2-positive metastatic biliary tract cancer. The response rate is promising for the initiation of randomised, controlled trials of pertuzumab plus trastuzumab in this patient population. Funding F Hoffmann-La Roche–Genentech.
- Published
- 2021
82. Tremelimumab and durvalumab in the treatment of unresectable, advanced hepatocellular carcinoma
- Author
-
Tiago Biachi de Castria, Danny N Khalil, James J Harding, Eileen M O'Reilly, and Ghassan K Abou-Alfa
- Subjects
Cancer Research ,Oncology ,General Medicine - Abstract
Liver cancer is the third most common cause of cancer-related mortality worldwide, with over 780,000 deaths in 2018. About 90% of liver cancer cases are hepatocellular carcinoma (HCC), a prototype of inflammation-driven cancer, leading to a robust rationale for the exploration of immune therapy. Previously approved agents for first-line therapy, such as sorafenib, lenvatinib and bevacizumab combined with atezolizumab, have focused on angiogenesis. HIMALAYA was the first trial to demonstrate the benefit of dual immune checkpoint inhibitors, representing a new treatment option in this scenario.Liver cancer is the third most common cause of cancer-related mortality worldwide, with over 780,000 deaths in 2018. About 90% of liver cancer cases originate in liver cells and are referred to as hepatocellular carcinoma (HCC). Systemic treatment (medications) is the mainstay for patients with advanced disease who are not suitable for resection or liver transplant and aims to improve survival and quality of life. HIMALAYA was the first study to demonstrate the benefit of using a combination of two immunotherapy medications for initial treatment.
- Published
- 2022
83. Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Nicolas A. Giraldo, Esther Drill, Baby A. Satravada, Imane El Dika, A. Rose Brannon, Josephine Dermawan, Abhinita Mohanty, Kerem Ozcan, Debyani Chakravarty, Ryma Benayed, Efsevia Vakiani, Ghassan K. Abou-Alfa, Ritika Kundra, Nikolaus Schultz, Bob T. Li, Michael F. Berger, James J. Harding, Marc Ladanyi, Eileen M. O'Reilly, William Jarnagin, Chad Vanderbilt, Olca Basturk, and Maria E. Arcila
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Gallbladder carcinoma (GBC) is an uncommon and aggressive disease, which remains poorly defined at a molecular level. Here, we aimed to characterize the molecular landscape of GBC and identify markers with potential prognostic and therapeutic implications. Experimental Design: GBC samples were analyzed using the MSK-IMPACT (Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets) platform (targeted NGS assay that analyzes 505 cancer-associated genes). Variants with therapeutic implications were identified using OncoKB database. The associations between recurrent genetic alterations and clinicopathologic characteristics (Fisher exact tests) or overall survival (univariate Cox regression) were evaluated. P values were adjusted for multiple testing. Results: Overall, 244 samples (57% primary tumors and 43% metastases) from 233 patients were studied (85% adenocarcinomas, 10% carcinomas with squamous differentiation, and 5% neuroendocrine carcinomas). The most common oncogenic molecular alterations appeared in the cell cycle (TP53 63% and CDKN2A 21%) and RTK_RAS pathways (ERBB2 15% and KRAS 11%). No recurrent structural variants were identified. There were no differences in the molecular landscape of primary and metastasis samples. Variants in SMAD4 and STK11 independently associated with reduced survival in patients with metastatic disease. Alterations considered clinically actionable in GBC or other solid tumor types (e.g., NTRK1 fusions or oncogenic variants in ERBB2, PIK3CA, or BRCA1/2) were identified in 35% of patients; 18% of patients with metastatic disease were treated off-label or enrolled in a clinical trial based on molecular findings. Conclusions: GBC is a genetically diverse malignancy. This large-scale genomic analysis revealed alterations with potential prognostic and therapeutic implications and provides guidance for the development of targeted therapies.
- Published
- 2022
84. P18 Pemigatinib for previously treated locally advanced or metastatic cholangiocarcinoma: final results from FIGHT-202
- Author
-
Arndt Vogel, Vaibhav Sahai, Antoine Hollebecque, Gina M Vaccaro, Davide Melisi, Raed MAl Rajabi, Andrew S Paulson, Mitesh J Borad, David Gallinson, Adrian G Murphy, Do-Youn Oh, Efrat Dotan, Daniel V Catenacci, Eric Van Cutsem, Christine F Lihou, Huiling Zhen, Luisa Veronese, and Ghassan K Abou-Alfa
- Published
- 2022
85. Assessment of pegylated arginine deiminase and modified FOLFOX6 in patients with advanced hepatocellular carcinoma: Results of an international, single‐arm, phase 2 study
- Author
-
Chia Jui Yen, James J. Harding, Stacey A. Cohen, Mehmet Akce, Yin Hsun Feng, Shukui Qin, Wen Tsung Huang, Imane El Dika, Ghassan K. Abou-Alfa, Tim Meyer, Dae Won Lee, Amanda Johnston, John S. Bomalaski, Debashis Sarker, Benjamin R. Tan, Baek Yeol Ryoo, Eileen M. O'Reilly, Yen Yang Chen, Ho Yeong Lim, Ching Liang Ho, and Tsai Sheng Yang
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Hydrolases ,Leucovorin ,Phases of clinical research ,Neutropenia ,Gastroenterology ,Polyethylene Glycols ,FOLFOX ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Aged ,Aged, 80 and over ,business.industry ,Liver Neoplasms ,Hepatitis C ,Middle Aged ,Hepatitis B ,medicine.disease ,digestive system diseases ,Oxaliplatin ,Oncology ,Response Evaluation Criteria in Solid Tumors ,Hepatocellular carcinoma ,Female ,Fluorouracil ,business ,medicine.drug - Abstract
Background Arginine starvation depletes the micronutrients required for DNA synthesis and interferes with both thymidylate synthetase activity and DNA repair pathways in preclinical models of hepatocellular carcinoma (HCC). Pegylated arginine deiminase (ADI-PEG 20), an arginine degrader, potentiates the cytotoxic activity of platinum and pyrimidine antimetabolites in HCC cellular and murine models. Methods This was a global, multicenter, open-label, single-arm, phase 2 trial of ADI-PEG 20 and modified 5-fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) in patients who had HCC with Child-Pugh A cirrhosis and disease progression on ≥2 prior lines of treatment. The primary objective was the objective response rate assessed according to Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary objectives were to estimate progression-free survival, overall survival, safety, and tolerability. Eligible patients were treated with mFOLFOX6 intravenously biweekly at standard doses and ADI-PEG-20 intramuscularly weekly at 36 mg/m2 . Results In total, 140 patients with advanced HCC were enrolled. The median patient age was 62 years (range, 30-85 years), 83% of patients were male, 76% were of Asian race, 56% had hepatitis B viremia, 10% had hepatitis C viremia, 100% had received ≥2 prior lines of systemic therapy, and 39% had received ≥3 prior lines of systemic therapy. The objective response rate was 9.3% (95% confidence interval [CI], 5.0%-15.4%), with a median response duration of 10.2 months (95% CI, 5.8 months to not reached). The median progression-free survival was 3.8 months (95% CI, 1.8-6.3 months), and the median overall survival was 14.5 months (95% CI, 13.6-20.9 months). The most common grade ≥3 treatment-related events were neutropenia (32.9%), white blood cell count decrease (20%), platelet count decrease (19.3%), and anemia (9.3%). Conclusions Concurrent mFOLFOX6 plus ADI-PEG 20 exhibited limited antitumor activity in patients with treatment-refractory HCC. The study was terminated early, and no further evaluation of the combination will be pursued. Lay summary Arginine is an important nutrient for hepatocellular carcinoma (HCC). The depletion of arginine with pegylated arginine deiminase (ADI-PEG 20), an arginine degrader, appeared to make chemotherapy (FOLFOX) work better in animal models of HCC and in patients with HCC on an early phase clinical trial. To formally test this hypothesis in the clinical setting, a large, global, phase 2 clinical trial was conducted of ADI-PEG 20 and FOLFOX in the treatment of patients with refractory HCC. The study showed limited activity of ADI-PEG 20 and FOLFOX in advanced HCC and was stopped early.
- Published
- 2021
86. ASO Visual Abstract: Recurrence of Hepatocellular Carcinoma After Complete Radiologic Response to Trans-Arterial embolization: A Retrospective Study on Patterns, Treatments, and Prognosis
- Author
-
Crisanta H. Ilagan, Debra A. Goldman, Mithat Gönen, Victoria G. Aveson, Michelle Babicky, Vinod P. Balachandran, Jeffrey A. Drebin, William R. Jarnagin, Alice C. Wei, T. Peter Kingham, Ghassan K. Abou-Alfa, Karen T. Brown, and Michael I. D’Angelica
- Subjects
Carcinoma, Hepatocellular ,Oncology ,Liver Neoplasms ,Humans ,Surgery ,Chemoembolization, Therapeutic ,Prognosis ,Embolization, Therapeutic ,Retrospective Studies - Published
- 2022
87. Tremelimumab plus Durvalumab in Unresectable Hepatocellular Carcinoma
- Author
-
Ghassan K. Abou-Alfa, George Lau, Masatoshi Kudo, Stephen L. Chan, Robin Kate Kelley, Junji Furuse, Wattana Sukeepaisarnjaroen, Yoon-Koo Kang, Tu Van Dao, Enrico N. De Toni, Lorenza Rimassa, Valeriy Breder, Alexander Vasilyev, Alexandra Heurgué, Vincent C. Tam, Kabir Mody, Satheesh Chiradoni Thungappa, Yuriy Ostapenko, Thomas Yau, Sergio Azevedo, María Varela, Ann-Lii Cheng, Shukui Qin, Peter R. Galle, Sajid Ali, Michelle Marcovitz, Mallory Makowsky, Philip He, John F. Kurland, Alejandra Negro, and Bruno Sangro
- Published
- 2022
88. Treatment With Liposomal Irinotecan Plus Fluorouracil and Leucovorin for Patients With Previously Treated Metastatic Biliary Tract Cancer
- Author
-
Jaewon Hyung, Ilhwan Kim, Kyu-pyo Kim, Baek-Yeol Ryoo, Jae Ho Jeong, Myoung Joo Kang, Jaekyung Cheon, Byung Woog Kang, Hyewon Ryu, Ji Sung Lee, Kyung Won Kim, Ghassan K. Abou-Alfa, and Changhoon Yoo
- Subjects
Cancer Research ,Oncology - Abstract
ImportanceThe NIFTY trial demonstrated the benefit of treatment with second-line liposomal irinotecan (nal-IRI) plus fluorouracil (FU) and leucovorin (LV) for patients with advanced biliary tract cancer (BTC).ObjectiveTo report the updated efficacy outcomes from the NIFTY trial with extended follow-up of 1.3 years with reperformed masked independent central review (MICR) with 3 newly invited radiologists.Design, Setting, and ParticipantsThe NIFTY trial was a randomized, multicenter, open-label, phase 2b clinical trial conducted between September 5, 2018, and December 31, 2021, at 5 tertiary referral centers in South Korea. Patients with advanced BTC whose disease progressed while receiving first-line gemcitabine plus cisplatin with at least 1 measurable lesion per Response Evaluation Criteria in Solid Tumors, version 1.1, were eligible. Data analysis was completed on May 9, 2022.InterventionsPatients were randomized 1:1 to receive LV, 400 mg/m2, bolus and FU, 2400 mg/m2, for a 46-hour infusion intravenously every 2 weeks with or without nal-IRI, 70 mg/m2, before LV intravenously. Patients were treated until disease progression or unacceptable toxic effects.Main Outcomes and MeasuresPrimary end point was progression-free survival (PFS) as assessed by MICR. Secondary end points were PFS as assessed by the investigator, overall survival, and objective response rate.ResultsA total of 178 patients (75 women [42.1%]; median [IQR] age, 64 [38-84] years) were randomly assigned, and 174 patients were included in the full analysis set (88 patients [50.6%] in the nal-IRI plus FU/LV group vs 86 patients [49.4%] in the FU/LV alone group). In this updated analysis, the median MICR-assessed PFS was 4.2 months (95% CI, 2.8-5.3) for the nal-IRI plus FU/LV group and 1.7 months (95% CI, 1.4-2.6) for the FU/LV alone group (hazard ratio, 0.61; 95% CI, 0.44-0.86; P = .004), in contrast to the 7.1 and 1.4 months reported in the previous study, respectively. The discordance rate for tumor progression date between the MICR and investigators was 17.8% (vs 30% in the previous study).Conclusions and RelevanceThe NIFTY randomized clinical trial demonstrated significant improvement in PFS with treatment with nal-IRI plus FU/LV compared with FU/LV alone for patients with advanced BTC after progression to gemcitabine plus cisplatin. The combination of nal-IRI plus FU/LV could be considered as a second-line treatment option for patients with previously treated advanced BTC.Trial Registrationclinicaltrials.gov Identifier: NCT03524508
- Published
- 2023
89. Abstract 6421: Molecular profiles and single cell analysis identify immunogenic pancreatic ductal adenocarcinoma (iPDAC)
- Author
-
Wungki Park, Catherine O'Connor, Shigeaki Umeda, Roshan Sharma, Yingjie Zhu, Elias-Ramzey Karnoub, Anna Varghese, Kevin C. Soares, Alejandro Jimemez, Asli Yavas, Kenneth H. Yu, Balachandran P. Vinod, Joanne F. Chou, Danny N. Khalil, Kelsen David, Hulya Sahin Ozkan, Olca Basturk, Marinela Capanu, Tal Nawy, Michael F. Berger, Ghassan K. Abou-Alfa, Jorge S. Reis-Filho, Ronan Chaligne, Nadeem Riaz, Dana Pe'er, Christine Iacobuzio-Donahue, and Eileen M. O'Reilly
- Subjects
Cancer Research ,Oncology - Abstract
Most pancreatic ductal adenocarcinomas (PDAC) are lethal and resistant to immunotherapy. Thus, identifying the immunogenic subgroup (iPDAC) and therapeutic targets can save lives. Herein, we present molecular features of iPDAC. 3 cohorts (A, B, C) from 288 patients whose sequenced tumors (MSK-IMPACT) were classified by homologous recombination deficiency groups. MSI-H were excluded. Survival, tumor mutation burden, genomic instability score, and enriched pathways for each cohort are included in Table 1. Patients in A (BRCA1/2/PALB2) had longer survivals vs B/C. 61 samples were selected for bulk RNAseq analysis for A vs C. Gene Ontology was enriched for upregulated humoral, T cell, and neutrophil immunity. CIBERSORT suggested higher infiltration of gamma delta T (Tgd) cells (p=0.039) and neutrophils (p=0.012), but lower Treg (p=0.001). Multidimensional insights in cellular components of cancer, immune, stroma, and neural genes were obtained by single nuclear RNA (snRNAseq) analysis from 30 biopsies for A vs C. 10x Genomics Chromium platform for library and Scanpy for computational analysis after Cell Ranger pipelines were used. 61,868 nuclei were profiled from 18 (13 baseline and 5 matched longitudinal) samples after quality evaluation. UMAP accurately clustered cells from each patient. Long-term survivors (LTS) had heterogenous baseline immune cell infiltrates of plasma cells, neutrophils, and CD8 (+) cytotoxic T cells. In matched samples of LTS, evolution of more prominent CD8 (+) T cells, macrophage, plasma cell, and neutrophil were observed. Single nucleus T-Cell Receptor sequencing for clonal trajectory inference will be done to determine the associated single cell molecular features contributing to iPDAC and identify novel targets for future intervention. Table 1. Cohort (Total: N=288) A: core HRD (BRCA1/2/PALB2) B: non-core HRD (ATM, BARD1, BLM, CHEK2, RAD50, RAD51C, RTEL1, MUTYH) C: others without HR-gene alterations Number (%) 48 (16.6) 19 (6.5) 221 (76) Median overall survival (95% confidence Interval) 33 months (3.6-64) 16 (11- not reached) 16 (14-18) Tumor Mutation Burden (TMB) 4.4 3.5 3.9 Genomic Instability Score (GIS, HRD score) 26 12 13 Gene Ongology term, enrichment score, adjusted p-value Adaptive immune response, GO:0002250, 0.49, 1.69e-10 Not included Reference to cohort A Humoral immune response, GO:0006959, 0.58, 1.67e-9 T cell activation, GO:0042110, 0.44, 2.75e-8 Neutrophil chemotaxis, GO:0030593, 0.73, 4.3e-10 Citation Format: Wungki Park, Catherine O'Connor, Shigeaki Umeda, Roshan Sharma, Yingjie Zhu, Elias-Ramzey Karnoub, Anna Varghese, Kevin C. Soares, Alejandro Jimemez, Asli Yavas, Kenneth H. Yu, Balachandran P. Vinod, Joanne F. Chou, Danny N. Khalil, Kelsen David, Hulya Sahin Ozkan, Olca Basturk, Marinela Capanu, Tal Nawy, Michael F. Berger, Ghassan K. Abou-Alfa, Jorge S. Reis-Filho, Ronan Chaligne, Nadeem Riaz, Dana Pe'er, Christine Iacobuzio-Donahue, Eileen M. O'Reilly. Molecular profiles and single cell analysis identify immunogenic pancreatic ductal adenocarcinoma (iPDAC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6421.
- Published
- 2023
90. Non-Surgical Locoregional Therapies Alone or in Combination with Systemic Therapy in Patients with Hepatocellular Carcinoma
- Author
-
Perla Chami, William Jarnagin, Ghassan K. Abou-Alfa, James Harding, Neal Kim, Haibo Lin, Maria El Homsi, Christopher Crane, and Carla Hajj
- Subjects
Cancer Research ,Oncology - Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer, representing the third-leading cause of cancer-related deaths worldwide. Curative intent treatment options for patients with HCC include liver transplantation, resection and ablation of small lesions. Other potentially curative therapies include cryoablation, microwave ablation and percutaneous alcohol injection. For locally advanced disease, different arterially directed therapies including transarterial chemoembolization and selective internal radiation therapy, plus external beam radiation including three-dimensional conformal radiation therapy, intensity-modulated radiation therapy, stereotactic body radiation therapy and proton beam therapy, are available or studied. Systemic therapies based on checkpoint inhibitors and tyrosine kinase inhibitors are available for the management of metastatic HCC and sometimes for locally advanced disease. Combinations of locoregional therapies with systemic drugs are currently the subject of several clinical trials.
- Published
- 2023
91. Genome-scale profiling of circulating cell-free DNA signatures for early detection of hepatocellular carcinoma in cirrhotic patients
- Author
-
Lei, Chen, Ghassan K, Abou-Alfa, Bo, Zheng, Jing-Feng, Liu, Jian, Bai, Lu-Tao, Du, Yun-Song, Qian, Rong, Fan, Xiao-Long, Liu, Lin, Wu, Jin-Lin, Hou, Hong-Yang, Wang, and Chuan-Xin, Wang
- Subjects
Liver Cirrhosis ,Carcinoma, Hepatocellular ,business.industry ,Liver Neoplasms ,Genome scale ,Early detection ,Cell Biology ,Computational biology ,Biology ,medicine.disease ,Circulating Cell-Free DNA ,Text mining ,Hepatocellular carcinoma ,Biomarkers, Tumor ,medicine ,Humans ,Profiling (information science) ,business ,Cell-Free Nucleic Acids ,Letter to the Editor ,Molecular Biology - Published
- 2021
92. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma
- Author
-
Thomas Yau, Jaclyn Neely, Ignacio Melero, Yun Shen, Richard S. Finn, Zachary Boyd, Marina Tschaika, Hao Tracy Tang, Bruno Sangro, Samir Wadhawan, Junji Furuse, Ann-Lii Cheng, Ghassan K. Abou-Alfa, Joong-Won Park, and Anthony B. El-Khoueiry
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,LAG3 ,Hepatocellular carcinoma ,CD8 Antigens ,Programmed Cell Death 1 Receptor ,Ipilimumab ,Inflammation ,Article ,B7-H1 Antigen ,Biomarkers, Pharmacological ,Immune system ,Antigens, CD ,Internal medicine ,Biomarkers, Tumor ,Humans ,Medicine ,Immune Checkpoint Inhibitors ,Hepatology ,business.industry ,Liver Neoplasms ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Survival Analysis ,Lymphocyte Activation Gene 3 Protein ,Nivolumab ,STAT1 Transcription Factor ,Programmed death ligand 1 (PD-L1) ,Female ,Inflammatory gene expression signatures ,Drug Monitoring ,medicine.symptom ,business ,Liver cancer ,medicine.drug - Abstract
Background & Aims: Nivolumab, a programmed death (PD)-1 (PD-1) inhibitor, led to durable responses, manageable safety, and increased survival in patients with advanced hepatocellular carcinoma (HCC). In our retrospective analysis, we studied the immunobiology and potential associations between biomarkers and outcomes with nivolumab in HCC. Methods: Fresh and archival tumour samples from dose-escalation and dose-expansion phases of the CheckMate 040 trial were analysed by immunohistochemistry and RNA sequencing to assess several inflammatory gene expression signatures, including CD274 (PD-ligand 1 [PD-L1]), CD8A, LAG3, and STAT1. Biomarkers were assessed for association with clinical outcomes (best overall response by blinded independent central review per RECIST v1.1 and overall survival [OS]). Results: Complete or partial tumour responses were observed in PD-L1–positive and PD-L1–negative patients treated with nivolumab monotherapy. Median OS was 28.1 (95% CI 18.2–n.a.) vs. 16.6 months (95% CI 14.2–20.2) for patients with tumour PD-L1 ≥1% vs., Graphical Abstract
- Published
- 2020
93. Targeting HER2 mutant advanced biliary tract cancers with neratinib: results from the SUMMIT ‘basket’ trial
- Author
-
James Harding, Sarina Piha-Paul, Ronak Shah, Jessica Murphy, James Cleary, Geoffrey Shapiro, David Quinn, Irene Braña, Victor Moreno, Mitesh Borad, Sherene Loi, Iben Spanggaard, Haeseong Park, James Ford, Monica Arnedos, Salomon Stemmer, Christelle de la Fouchardiere, Christos Fountzilas, Jie Zhang, Daniel DiPrimeo, Casey Savin, S. Duygu Selcuklu, Michael Berger, Lisa Eli, Funda Meric-Bernstam, Komal Jhaveri, David Solit, and Ghassan K Abou-Alfa
- Abstract
HER2 mutations are infrequent genomic events in biliary tract cancers (BTCs). Neratinib, an irreversible, pan-HER, oral tyrosine kinase inhibitor, interferes with constitutive receptor kinase activation and has activity in HER2-mutant tumours. SUMMIT is an open-label, single-arm, multi-cohort, phase 2, ‘basket’ trial of neratinib in patients with solid tumours harbouring oncogenic HER2 somatic mutations (NCT01953926). The primary objective of the BTC cohort was objective response rate (ORR). Among 25 treatment-refractory patients (11 cholangiocarcinoma, 10 gallbladder, 4 ampullary cancers), the ORR was 16%. The most common HER2 mutations were S310F (n = 11; 48%) and V777L (n = 4; 17%). Outcomes appeared worse for ampullary tumours or those with co-occurring oncogenic TP53 and CDKN2A alterations. Loss of amplified HER2 S310F and acquisition of multiple previously undetected oncogenic co-mutations were identified at progression in one responder. Neratinib demonstrated anti-tumour activity in patients with refractory BTC harbouring HER2 mutations. Future studies of rational combinations are warranted.
- Published
- 2022
94. Progression-Free Survival in Patients With Cholangiocarcinoma With or Without FGF/FGFR Alterations: A FIGHT-202 Post Hoc Analysis of Prior Systemic Therapy Response
- Author
-
Kristen Bibeau, Luis Féliz, Christine F. Lihou, Haobo Ren, and Ghassan K. Abou-Alfa
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE Oncogenic fibroblast growth factor receptor (FGFR) gene alterations have been described in patients with cholangiocarcinoma (CCA). This post hoc analysis assessed progression-free survival (PFS) in patients who had received first- or second-line systemic therapy for advanced/metastatic CCA before enrollment in the phase II FIGHT-202 study (ClinicalTrials.gov identifier: NCT02924376 ). PATIENTS AND METHODS Patients with locally advanced or metastatic CCA with FGFR2 fusions/rearrangements (n = 107), other FGF/FGFR alterations (n = 20), or no FGF/FGFR alterations (n = 18) and documented disease progression after at least one systemic cancer therapy before enrollment in FIGHT-202 were assessed. Prior therapy and disease response data were collated from electronic case report forms. PFS was calculated for each prior line of systemic cancer therapy. RESULTS Among patients with FGFR2 fusions/rearrangements, other FGF/ FGFR alterations, and no FGF/ FGFR alterations, respectively, the median PFS with prior first-line systemic therapy was 5.5 months (95% CI, 4.0 to 8.0; n = 102), 4.4 months (2.7 to 7.1; n = 19), and 2.8 months (1.6 to 11.3; n = 16); the median PFS with prior second-line systemic therapy was 4.2 months (3.0 to 5.3; n = 39), 3.0 months (1.1 to 9.9; n = 8), and 5.9 months (2.4 to 12.5; n = 6). The median PFS was 7.0 months (4.9 to 11.1) for patients with FGFR2 fusions/rearrangements (n = 65) with second-line pemigatinib received during the FIGHT-202 trial. CONCLUSION In patients with CCA and FGFR2 fusions or rearrangements, second-line treatment with pemigatinib may be associated with longer PFS compared with second-line treatment with systemic therapy received before study enrollment; however, a prospective controlled trial is required to confirm this. The results support the therapeutic potential of pemigatinib previously demonstrated in FIGHT-202.
- Published
- 2022
95. Circulating tumor and invasive cell expression profiling predicts effective therapy in pancreatic cancer
- Author
-
Kenneth H. Yu, Jennifer Park, Avni Mittal, Ghassan K. Abou‐Alfa, Imane El Dika, Andrew S. Epstein, David H. Ilson, David P. Kelsen, Geoffrey Y. Ku, Jia Li, Wungki Park, Anna M. Varghese, Joanne F‐L Chou, Marinela Capanu, Brandon Cooper, Andrew Bartlett, Devan McCarthy, Vineet Sangar, Brian McCarthy, and Eileen M. O'Reilly
- Subjects
Pancreatic Neoplasms ,Cancer Research ,Oncology ,Paclitaxel ,Albumins ,Antineoplastic Combined Chemotherapy Protocols ,Leucovorin ,Humans ,Fluorouracil ,Prospective Studies ,Adenocarcinoma ,Deoxycytidine - Abstract
Pancreatic adenocarcinoma (PDAC) remains a refractory disease; however, modern cytotoxic chemotherapeutics can induce tumor regression and extend life. A blood-based, pharmacogenomic, chemosensitivity assay using gene expression profiling of circulating tumor and invasive cells (CTICs) to predict treatment response was previously developed. The combination regimen of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel (G/nab-P) are established frontline approaches for treating advanced PDAC; however, there are no validated biomarkers for treatment selection. A similar unmet need exists for choosing second-line therapy.The chemosensitivity assay was evaluated in metastatic PDAC patients presenting for frontline treatment. A prospective study enrolled patients (n = 70) before receiving either FOLFIRINOX or G/nab-P at a 1:1 ratio. Six milliliters of peripheral blood was collected at baseline and at time of disease progression. CTICs were isolated, gene-expression profiling was performed, and the assay was used to predict effective and ineffective chemotherapeutic agents. Treating physicians were blinded to the assay prediction results.Patients receiving an effective regimen as predicted by the chemosensitivity assay experienced significantly longer median progression-free survival (mPFS; 7.8 months vs. 4.2 months; hazard ratio [HR], 0.35; p = .0002) and median overall survival (mOS; 21.0 months vs. 9.7 months; HR, 0.40; p = .005), compared with an ineffective regimen. Assay prediction for effective second-line therapy was explored. The entire study cohort experienced favorable outcomes compared with historical controls, 7.1-month mPFS and 12.3-month mOS.Chemosensitivity assay profiling is a promising tool for guiding therapy in advanced PDAC. Further prospective validation is under way (clinicaltrials.gov NCT03033927).
- Published
- 2022
96. Racial and ethnic disparities in early treatment with immunotherapy for advanced HCC in the United States
- Author
-
Joseph C. Ahn, Marie Lauzon, Michael Luu, Mazen Noureddin, Walid Ayoub, Alexander Kuo, Vinay Sundaram, Kambiz Kosari, Nicholas Nissen, Jun Gong, Andrew Hendifar, Lewis R. Roberts, Ghassan K. Abou‐Alfa, Amit G. Singal, and Ju Dong Yang
- Subjects
Carcinoma, Hepatocellular ,Hepatology ,Liver Neoplasms ,Ethnicity ,Humans ,Immunotherapy ,Healthcare Disparities ,United States - Abstract
Immunotherapy has emerged as an effective treatment for patients with advanced-stage HCC. We aimed to investigate the efficacy of immunotherapy for advanced HCC in a nationwide cohort and racial and ethnic disparities in access to immunotherapy.We used the US National Cancer Database to identify patients with tumor-node-metastasis stage 3 or 4 HCC between 2017 and 2018. We performed multivariable Cox regression to identify factors associated with overall survival (OS) and logistic regression to identify factors associated with receipt of immunotherapy. Of the 3,990 patients treated for advanced HCC, 3,248 (81.4%) patients received chemotherapy and 742 (18.6%) patients received immunotherapy as a first-line treatment. Immunotherapy was associated with improved OS compared with chemotherapy (adjusted HR: 0.76, 95% CI: 0.65-0.88) after adjusting for covariates. There were racial and ethnic disparities in access to immunotherapy, with Hispanic (adjusted OR [aOR]: 0.63, 95% CI: 0.46-0.83) and Black patients (aOR: 0.71, 95% CI: 0.54-0.89) less likely to receive immunotherapy compared with White patients. There was a significant interaction between race-ethnicity and facility type, with higher disparity observed in nonacademic centers (interaction p = 0.004).Immunotherapy was associated with improved OS compared with chemotherapy in advanced HCC. There are significant disparities in early access to immunotherapy, likely due to differential access to clinical trials and experimental therapies. A comprehensive approach to monitoring and eliminating racial-ethnic disparities in the management of advanced HCC is urgently needed.
- Published
- 2022
97. KDR genetic predictor of toxicities induced by sorafenib and regorafenib
- Author
-
Julia C. F. Quintanilha, Susan Geyer, Amy S. Etheridge, Alessandro Racioppi, Kelli Hammond, Daniel J. Crona, Carol E. Peña, Sawyer B. Jacobson, Federica Marmorino, Daniele Rossini, Chiara Cremolini, Hanna K. Sanoff, Ghassan K. Abou-Alfa, and Federico Innocenti
- Subjects
Pharmacology ,Pyridines ,Phenylurea Compounds ,Neoplasms ,Genetics ,Molecular Medicine ,Humans ,Sorafenib ,Vascular Endothelial Growth Factor Receptor-2 ,Article - Abstract
No biomarkers are available to predict toxicities induced by VEGFR TKIs. This study aimed to identify markers of toxicities induced by these drugs using a discovery-validation approach. The discovery set included 140 sorafenib-treated cancer patients (TARGET study) genotyped for SNPs in 56 genes. The most significant SNPs associated with grade ≥2 hypertension, diarrhea, dermatologic toxicities, and composite toxicity (any one of the toxicities) were tested for association with grade ≥2 toxicity in a validation set of 201 sorafenib-treated patients (Alliance/CALGB 80802). The validated SNP was tested for association with grade ≥2 toxicity in 107 (LCCC 1029) and 82 (Italian cohort) regorafenib-treated patients. SNP-toxicity associations were evaluated using logistic regression, and a meta-analysis between the studies was performed by inverse variance. Variant rs4864950 in KDR increased the risk of grade ≥2 composite toxicity in TARGET, Alliance/CALGB 80802, and the Italian cohort (meta-analysis p=6.79×10(−4), OR=2.01, 95% CI 1.34–3.01). We identified a predictor of toxicities induced by VEGFR TKIs.
- Published
- 2022
98. Efficacy and safety of cabozantinib for patients with advanced hepatocellular carcinoma based on albumin-bilirubin grade
- Author
-
S. Hazra, Zhong-Zhe Lin, Yen-Hsun Chen, Ghassan K. Abou-Alfa, Suvajit Sen, Tim Meyer, Rebecca A. Miksad, Jillian Youkstetter, Ann-Lii Cheng, Stefano Kim, Anthony B. El-Khoueiry, Irfan Cicin, Heinz-Josef Klümpen, Robin Kate Kelley, Oncology, and CCA - Cancer Treatment and Quality of Life
- Subjects
Sorafenib ,Oral ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Cabozantinib ,Pyridines ,Population ,Placebo ,Gastroenterology ,chemistry.chemical_compound ,Liver Function Tests ,Internal medicine ,medicine ,80 and over ,Humans ,Anilides ,education ,Adverse effect ,Serum Albumin ,Aged ,Retrospective Studies ,education.field_of_study ,business.industry ,Hazard ratio ,Carcinoma ,Liver Neoplasms ,Bilirubin ,Hepatocellular ,Middle Aged ,medicine.disease ,Survival Analysis ,Treatment Outcome ,Oncology ,chemistry ,Hepatocellular carcinoma ,Administration ,Female ,Liver function ,business ,medicine.drug - Abstract
Background Albumin-bilirubin (ALBI) grade is an objective measure of liver function for patients with hepatocellular carcinoma (HCC). The tyrosine kinase inhibitor cabozantinib is approved for patients with advanced HCC who have received prior sorafenib based on the phase 3 CELESTIAL trial (NCT01908426). Cabozantinib improved overall survival (OS) and progression-free survival (PFS) versus placebo in patients with previously treated HCC. Methods Patients were randomised 2:1 to receive cabozantinib 60 mg or placebo orally every day. Clinical outcomes in patients with ALBI grade 1 or 2 at baseline were evaluated in CELESTIAL. ALBI scores were retrospectively calculated based on baseline serum albumin and total bilirubin, with an ALBI grade of 1 defined as ≤ −2.60 score and a grade of 2 as a score of > −2.60 to ≤ −1.39. Results Cabozantinib improved OS and PFS versus placebo in both ALBI grade 1 (hazard ratio [HR] [95% CI]: 0.63 [0.46–0.86] and 0.42 [0.32–0.56]) and ALBI grade 2 (HR [95% CI]: 0.84 [0.66–1.06] and 0.46 [0.37–0.58]) subgroups. Adverse events were consistent with those in the overall population. Rates of grade 3/4 adverse events associated with hepatic decompensation were generally low and were more common among patients in the ALBI grade 2 subgroup. Discussion These results provide initial support of cabozantinib in patients with advanced HCC irrespective of ALBI grade 1 or 2. Trial registration number ClinicalTrials.gov number, NCT01908426.
- Published
- 2022
99. Dermatologic Adverse Events Associated with Selective Fibroblast Growth Factor Receptor Inhibitors: Overview, Prevention, and Management Guidelines
- Author
-
Milan J. Anadkat, Benjamin H. Kaffenberger, Kathleen Guindon, Ghassan K. Abou-Alfa, Jonathan S. Leventhal, Mario E. Lacouture, and Vincent Sibaud
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Morpholines ,Antineoplastic Agents ,Guidelines ,03 medical and health sciences ,0302 clinical medicine ,Erdafitinib ,Internal medicine ,medicine ,Humans ,Pyrroles ,Adverse effect ,Calciphylaxis ,Bladder cancer ,business.industry ,Drug‐related side effects and adverse events ,Onycholysis ,medicine.disease ,Receptors, Fibroblast Growth Factor ,Dermatologic ,Fibroblast growth factor receptor ,Discontinuation ,Bile Ducts, Intrahepatic ,Pyrimidines ,030104 developmental biology ,Bile Duct Neoplasms ,Symptom Management and Supportive Care ,030220 oncology & carcinogenesis ,Quality of Life ,business ,Tyrosine kinase - Abstract
Fibroblast growth factor receptor (FGFR) tyrosine kinases, which are expressed on the cell membrane, are involved in a wide range of biological functions such as cell proliferation, survival, migration, and differentiation. The identification of FGFR fusions and other alterations in a wide range of solid tumors, including cholangiocarcinoma and bladder cancer, has resulted in the development of several selective FGFR inhibitors for use in these indications, for example, infigratinib, erdafitinib, derazantinib, pemigatinib, and futibatinib. In addition to the typical adverse events associated with tyrosine kinases, the FGFR inhibitors appear to give rise to a number of adverse events affecting the skin. Here we describe these skin events, which include the more common nail adverse events (e.g., onycholysis), palmar–plantar erythrodysesthesia syndrome, and stomatitis, as well as less common reactions such as calciphylaxis. This review aims to provide oncologists with an understanding of these dermatologic events and proposes guidelines for the management of treatment‐emergent dermatologic adverse events. Awareness of possible adverse events associated with specific drugs should allow physicians to educate patients as to what to expect and implement effective management plans at the earliest possible opportunity, thereby preventing premature discontinuation while maintaining patient quality of life. Implications for Practice Identification of fibroblast growth factor receptor (FGFR) aberrations in cholangiocarcinoma and bladder cancer led to development of selective FGFR inhibitors for these indications, based on clinical benefit and safety profiles. The most frequent adverse events (AEs) include those affecting skin, hair, and nails, a unique class effect of these agents. These are usually mild to moderate in severity. This work reviewed skin AEs reported with FGFR inhibitors and provides management guidelines for physicians, aiming to increase awareness of skin events and provide effective treatment strategies. Early intervention and effective management may improve treatment adherence, optimize outcomes, and improve quality of life., This review provides oncologists with an understanding of dermatologic adverse events related to the use of FGFR inhibitors and proposes guidelines for treatment.
- Published
- 2020
100. Infigratinib in patients with advanced cholangiocarcinoma with FGFR2 gene fusions/translocations: the PROOF 301 trial
- Author
-
Angela Lamarca, Teresa Macarulla, Do Youn Oh, Ghassan K. Abou-Alfa, Ivan Borbath, Sameek Roychowdhury, Milind Javle, Amit Pande, Shalini Makawita, Allen Cohn, Lipika Goyal, Terry Cho, Rachna T. Shroff, Michael Howland, Saeed Sadeghi, and Ai Li
- Subjects
Cisplatin ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Chromosomal translocation ,General Medicine ,Malignancy ,medicine.disease ,Gemcitabine ,law.invention ,Targeted therapy ,Clinical trial ,Randomized controlled trial ,law ,Internal medicine ,medicine ,business ,Gene ,medicine.drug - Abstract
Cholangiocarcinoma is an aggressive malignancy with poor overall survival. Approximately 15% of intrahepatic cholangiocarcinomas contain FGFR alterations. Infigratinib is an oral FGFR 1–3 kinase inhibitor. Favorable results from a Phase II trial of infigratinib in advanced/metastatic FGFR-altered cholangiocarcinomas has led to its further investigation in the front-line setting. In this article we describe the design, objectives and rationale for PROOF 301, a Phase III multicenter, open label, randomized trial of infigratinib in comparison to standard of care gemcitabine and cisplatin in advanced/metastatic cholangiocarcinoma with FGFR2 translocations. The results of this study have the potential to define a new role for a chemotherapy-free, targeted therapy option in the front-line setting for these patients. Clinical Trial Registration: NCT03773302 (ClincalTrials.gov)
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.