186 results on '"Piha-Paul SA"'
Search Results
2. Retreatment with anti-EGFR based therapies in metastatic colorectal cancer: impact of intervening time interval and prior anti-EGFR response
- Author
-
Liu, X, George, GC, Tsimberidou, AM, Naing, A, Wheler, JJ, Kopetz, S, Fu, S, Piha-Paul, SA, Eng, C, Falchook, GS, Janku, F, Garrett, C, Karp, D, Kurzrock, R, Zinner, R, Raghav, K, Subbiah, V, Hess, K, Meric-Bernstam, F, Hong, DS, and Overman, MJ
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Digestive Diseases ,Clinical Research ,Colo-Rectal Cancer ,Adult ,Aged ,Antibodies ,Monoclonal ,Antibodies ,Monoclonal ,Humanized ,Cetuximab ,Colorectal Neoplasms ,Disease-Free Survival ,ErbB Receptors ,Erlotinib Hydrochloride ,Female ,Humans ,Male ,Middle Aged ,Neoplasm Metastasis ,Panitumumab ,Proto-Oncogene Proteins p21(ras) ,Retreatment ,Anti-EGFR treatment ,KRAS-wt CRC ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis ,Epidemiology - Abstract
BackgroundThis retrospective study aims to investigate the activity of retreatment with anti-EGFR-based therapies in order to explore the concept of clonal evolution by evaluating the impact of prior activity and intervening time interval.MethodsEighty-nine KRAS exon 2-wild-type metastatic colorectal patients were retreated on phase I/II clinical trials containing anti-EGFR therapies after progressing on prior cetuximab or panitumumab. Response on prior anti-EGFR therapy was defined retrospectively per physician-records as response or stable disease ≥6 months. Multivariable statistical methods included a multiple logistic regression model for response, and Cox proportional hazards model for progression-free survival.ResultsRetreatment anti-EGFR agents were cetuximab (n = 76) or cetuximab plus erlotinib (n = 13). The median interval time between prior and retreatment regimens was 4.57 months (range: 0.46-58.7). Patients who responded to the prior cetuximab or panitumumab were more likely to obtain clinical benefit to the retreatment compared to the non-responders in both univariate (p = 0.007) and multivariate analyses (OR: 3.38, 95 % CI: 1.27, 9.31, p = 0.019). The clinical benefit rate on retreatment also showed a marginally significant association with interval time between the two anti-EGFR based therapies (p = 0.053). Median progression-free survival on retreatment was increased in prior responders (4.9 months, 95 % CI: 3.6, 6.2) compared to prior non-responders (2.5 months, 95 % CI, 1.58, 3.42) in univariate (p = 0.064) and multivariate analysis (HR: 0.70, 95 % CI: 0.43-1.15, p = 0.156).ConclusionOur data lends support to the concept of clonal evolution, though the clinical impact appears less robust than previously reported. Further work to determine which patients benefit from retreatment post progression is needed.
- Published
- 2015
3. Results of a phase 1 trial combining ridaforolimus and MK-0752 in patients with advanced solid tumours
- Author
-
Piha-Paul, SA, Munster, PN, Hollebecque, A, Argilés, G, Dajani, O, Cheng, JD, Wang, R, Swift, A, Tosolini, A, and Gupta, S
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Clinical Research ,Cancer ,Clinical Trials and Supportive Activities ,Dental/Oral and Craniofacial Disease ,Rare Diseases ,Administration ,Oral ,Adult ,Aged ,Aged ,80 and over ,Antineoplastic Combined Chemotherapy Protocols ,Benzene Derivatives ,Carcinoma ,Squamous Cell ,Drug Administration Schedule ,Europe ,Female ,Head and Neck Neoplasms ,Humans ,Male ,Maximum Tolerated Dose ,Middle Aged ,Molecular Targeted Therapy ,Multimodal Imaging ,Positron-Emission Tomography ,Propionates ,Protein Kinase Inhibitors ,Signal Transduction ,Sirolimus ,Squamous Cell Carcinoma of Head and Neck ,Sulfones ,Time Factors ,Tomography ,X-Ray Computed ,Treatment Outcome ,United States ,Ridaforolimus ,mTOR inhibitor ,MK-0752 ,Notch inhibitor ,Head and neck cancer ,Phase I ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
BackgroundThe phosphatidylinositol 3-kinase/protein kinase-B/mammalian target of rapamycin (PI3K-AKT-mTOR) signalling pathway is aberrantly activated in several cancers. Notch signalling maintains cell proliferation, growth and metabolism in part by driving the PI3K pathway. Combining the mTOR inhibitor ridaforolimus with the Notch inhibitor MK-0752 may increase blockade of the PI3K pathway.MethodsThis phase I dose-escalation study (NCT01295632) aimed to define the dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) of combination oral ridaforolimus (rising doses starting at 20 mg, 5 days/week) and oral MK-0752 (1800 mg once weekly) in patients with solid tumours. No intrapatient dose escalation was permitted.ResultsTwenty eight patients were treated on study. Ridaforolimus doses were escalated from 20 to 30 mg/day. Among 14 evaluable patients receiving ridaforolimus 20 mg, one DLT (grade 2 stomatitis, second episode) was reported. Among eight evaluable patients receiving ridaforolimus 30 mg, three DLTs were reported (one each grade 3 stomatitis, grade 3 diarrhoea, and grade 3 asthenia). The MTD was 20 mg daily ridaforolimus 5 days/week+1800 mg weekly MK-0752. The most common drug-related adverse events included stomatitis, diarrhoea, decreased appetite, hyperglycaemia, thrombocytopenia, asthenia and rash. Two of 15 (13%) patients with head and neck squamous cell carcinoma (HNSCC) had responses: one with complete response and one with partial response. In addition, one patient experienced stable disease ⩾6 months.ConclusionsCombined ridaforolimus and MK-0752 showed activity in HNSCC. However, a high number of adverse events were reported at the MTD, which would require careful management during future clinical development.
- Published
- 2015
4. Neratinib plus fulvestrant plus trastuzumab for HR-positive, HER2-negative, HER2-mutant metastatic breast cancer: outcomes and biomarker analysis from the SUMMIT trial
- Author
-
Jhaveri, K, Eli, LD, Wildiers, H, Hurvitz, SA, Guerrero-Zotano, A, Unni, N, Brufsky, A, Park, H, Waisman, J, Yang, ES, Spanggaard, I, Reid, S, Burkard, ME, Vinayak, S, Prat, A, Arnedos, M, Bidard, F-C, Loi, S, Crown, J, Bhave, M, Piha-Paul, SA, Suga, JM, Chia, S, Saura, C, Garcia-Saenz, JA, Gambardella, V, de Miguel, MJ, Gal-Yam, EN, Raphael, A, Stemmer, SM, Ma, C, Hanker, AB, Ye, D, Goldman, JW, Bose, R, Peterson, L, Bell, JSK, Frazier, A, Diprimeo, D, Wong, A, Arteaga, CL, Solit, DB, Jhaveri, K, Eli, LD, Wildiers, H, Hurvitz, SA, Guerrero-Zotano, A, Unni, N, Brufsky, A, Park, H, Waisman, J, Yang, ES, Spanggaard, I, Reid, S, Burkard, ME, Vinayak, S, Prat, A, Arnedos, M, Bidard, F-C, Loi, S, Crown, J, Bhave, M, Piha-Paul, SA, Suga, JM, Chia, S, Saura, C, Garcia-Saenz, JA, Gambardella, V, de Miguel, MJ, Gal-Yam, EN, Raphael, A, Stemmer, SM, Ma, C, Hanker, AB, Ye, D, Goldman, JW, Bose, R, Peterson, L, Bell, JSK, Frazier, A, Diprimeo, D, Wong, A, Arteaga, CL, and Solit, DB
- Abstract
BACKGROUND: HER2 mutations are targetable alterations in patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC). In the SUMMIT basket study, patients with HER2-mutant MBC received neratinib monotherapy, neratinib + fulvestrant, or neratinib + fulvestrant + trastuzumab (N + F + T). We report results from 71 patients with HR+, HER2-mutant MBC, including 21 (seven in each arm) from a randomized substudy of fulvestrant versus fulvestrant + trastuzumab (F + T) versus N + F + T. PATIENTS AND METHODS: Patients with HR+ HER2-negative MBC with activating HER2 mutation(s) and prior cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) therapy received N + F + T (oral neratinib 240 mg/day with loperamide prophylaxis, intramuscular fulvestrant 500 mg on days 1, 15, and 29 of cycle 1 then q4w, intravenous trastuzumab 8 mg/kg then 6 mg/kg q3w) or F + T or fulvestrant alone. Those whose disease progressed on F + T or fulvestrant could cross-over to N + F + T. Efficacy endpoints included investigator-assessed objective response rate (ORR), clinical benefit rate (RECIST v1.1), duration of response, and progression-free survival (PFS). Plasma and/or formalin-fixed paraffin-embedded tissue samples were collected at baseline; plasma was collected during and at end of treatment. Extracted DNA was analyzed by next-generation sequencing. RESULTS: ORR for 57 N + F + T-treated patients was 39% [95% confidence interval (CI) 26% to 52%); median PFS was 8.3 months (95% CI 6.0-15.1 months). No responses occurred in fulvestrant- or F + T-treated patients; responses in patients crossing over to N + F + T supported the requirement for neratinib in the triplet. Responses were observed in patients with ductal and lobular histology, 1 or ≥1 HER2 mutations, and co-occurring HER3 mutations. Longitudinal circulating tumor DNA sequencing revealed acquisition of additional HER2 alterations, and mutations in genes including PIK3CA, enabling further precision targeting and possible re-respon
- Published
- 2023
5. A Phase I Trial of the MET/ALK/ROS1 Inhibitor Crizotinib Combined with the VEGF Inhibitor Pazopanib in Patients with Advanced Solid Malignancies
- Author
-
Piha-Paul SA, Dumbrava EE, Nair BC, Xiong W, Xu L, Mostorino R, Subbiah V, Tannir N, Fu S, Naing A, Janku F, Karp DD, Patel S, Daw NC, Hong D, Meric-Bernstam F, and Zinner R
- Subjects
crizotinib ,alk/ros1 ,met ,pazopanib ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,vegf ,RC254-282 - Abstract
Sarina A Piha-Paul,1 Ecaterina E Dumbrava,1 Binoj C Nair,1 Wendy Xiong,1 Li Xu,1 Rosa Mostorino,1 Vivek Subbiah,1 Nizar Tannir,2 Siqing Fu,1 Aung Naing,1 Filip Janku,1 Daniel D Karp,1 Shreyaskumar Patel,3 Najat C Daw,4 David Hong,1 Funda Meric-Bernstam,1,5,6 Ralph Zinner7 1Department of Investigational Cancer Therapeutics (A Phase I Clinical Trials Program), University of Texas MD Anderson Cancer Center, Houston, TX, USA; 2Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; 3Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; 4Department of Pediatrics, University of Texas MD Anderson Cancer Center, Houston, TX, USA; 5Department of Breast Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA; 6The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA; 7Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA, USACorrespondence: Sarina A Piha-PaulInvestigational Cancer Therapeutics, MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 455, Houston, TX, 77030, USATel +1 713-563-1055Fax +1 713-792-3535Email spihapau@mdanderson.orgBackground: Crizotinib inhibits ALK, MET and ROS1 tyrosine kinases but the development of resistance to monotherapy is an issue. The anti-angiogenic properties of pazopanib could overcome crizotinib drug resistance. Additionally, the anti-angiogenic properties of crizotinib could augment the clinical efficacy of pazopanib.Methods: We evaluated the safety and responses in patients with advanced solid tumors treated with crizotinib and pazopanib.Results: Eighty-two patients (median age 53 years, range 18– 78 years) were enrolled. The median number of prior systemic therapies was 3 (range, 0– 8). We were able to dose escalate to dose level 8 (crizotinib 250 mg twice daily and pazopanib 800 mg daily) with no MTD identified. Grade 3 or 4 toxicities were seen in 32% of patients with the highest prevalence being fatigue (n=9, 11%), diarrhea (n=6, 7%), vomiting (n=3, 4%), anemia (n=2, 2%) and ALT increased (n=2, 2%). Of the 82 patients, 61 (74%) had measurable disease by RECISTv1.1 and reached first restaging (6 weeks). Partial response (PR) was observed in 6/61 (10%) patients, and stable disease (SD) lasting ≥ 6 months was observed in 10/61 patients (16%) (total = 16/61 (26%) of patients with SD ≥ 6 months/PR).Conclusion: Dose level 6 (crizotinib 200 mg twice daily and pazopanib 600 mg daily) was the most tolerable dosing of the combination and can be used in future studies. We also observed moderate clinical activity in patients with advanced solid tumors that had received numerous prior therapies.Keywords: crizotinib, pazopanib, VEGF, ALK/ROS1, MET
- Published
- 2021
6. Safety, pharmacokinetic, pharmacodynamic and clinical activity of molibresib for the treatment of nuclear protein of the testis carcinoma and other cancers: Results of a Phase I/II open-label, dose escalation study
- Author
-
Cousin, S, Blay, J-Y, Garcia, IB, de Bono, JS, Le Tourneau, C, Moreno, V, Trigo, J, Hann, CL, Azad, AA, Im, S-A, Cassier, PA, French, CA, Italiano, A, Keedy, VL, Plummer, R, Sablin, M-P, Hemming, ML, Ferron-Brady, G, Wyce, A, Khaled, A, Datta, A, Foley, SW, McCabe, MT, Wu, Y, Horner, T, Kremer, BE, Dhar, A, O'Dwyer, PJ, Shapiro, GI, Piha-Paul, SA, Cousin, S, Blay, J-Y, Garcia, IB, de Bono, JS, Le Tourneau, C, Moreno, V, Trigo, J, Hann, CL, Azad, AA, Im, S-A, Cassier, PA, French, CA, Italiano, A, Keedy, VL, Plummer, R, Sablin, M-P, Hemming, ML, Ferron-Brady, G, Wyce, A, Khaled, A, Datta, A, Foley, SW, McCabe, MT, Wu, Y, Horner, T, Kremer, BE, Dhar, A, O'Dwyer, PJ, Shapiro, GI, and Piha-Paul, SA
- Abstract
Molibresib is an orally bioavailable, selective, small molecule BET protein inhibitor. Results from a first time in human study in solid tumors resulted in the selection of a 75 mg once daily dose of the besylate formulation of molibresib as the recommended Phase 2 dose (RP2D). Here we present the results of Part 2 of our study, investigating safety, pharmacokinetics, pharmacodynamics and clinical activity of molibresib at the RP2D for nuclear protein in testis carcinoma (NC), small cell lung cancer, castration-resistant prostate cancer (CRPC), triple-negative breast cancer, estrogen receptor-positive breast cancer and gastrointestinal stromal tumor. The primary safety endpoints were incidence of adverse events (AEs) and serious AEs; the primary efficacy endpoint was overall response rate. Secondary endpoints included plasma concentrations and gene set enrichment analysis (GSEA). Molibresib 75 mg once daily demonstrated no unexpected toxicities. The most common treatment-related AEs (any grade) were thrombocytopenia (64%), nausea (43%) and decreased appetite (37%); 83% of patients required dose interruptions and 29% required dose reductions due to AEs. Antitumor activity was observed in NC and CRPC (one confirmed partial response each, with observed reductions in tumor size), although predefined clinically meaningful response rates were not met for any tumor type. Total active moiety median plasma concentrations after single and repeated administration were similar across tumor cohorts. GSEA revealed that gene expression changes with molibresib varied by patient, response status and tumor type. Investigations into combinatorial approaches that use BET inhibition to eliminate resistance to other targeted therapies are warranted.
- Published
- 2022
7. Abstract PD3-06: Neratinib + fulvestrant for HER2-mutant, HR-positive, metastatic breast cancer: Updated results from the phase 2 SUMMIT trial
- Author
-
Smyth, LM, primary, Piha-Paul, SA, additional, Saura, C, additional, Loi, S, additional, Lu, J, additional, Shapiro, GI, additional, Juric, D, additional, Mayer, IA, additional, Arteaga, C, additional, de la Fuente, M, additional, Brufksy, AM, additional, Mau-Sørensen, M, additional, Arnedos, M, additional, Moreno, V, additional, Sohn, J-H, additional, Schwartzberg, L, additional, Gonzàlez-Farré, X, additional, Cervantes, A, additional, Mann, G, additional, Shahin, S, additional, Cutler, RE, additional, Eli, LD, additional, Xu, F, additional, Bagulho, T, additional, Lalani, AS, additional, Bryce, R, additional, Solit, DB, additional, Hyman, DM, additional, Meric-Bernstam, F, additional, and Baselga, J, additional
- Published
- 2019
- Full Text
- View/download PDF
8. HER kinase inhibition in patients with HER2-and HER3-mutant cancers
- Author
-
Hyman, DM, Piha-Paul, SA, Won, H, Rodon, J, Saura, C, Shapiro, GI, Juric, D, Quinn, DI, Moreno, V, Doger, B, Mayer, IA, Boni, V, Calvo, E, Loi, S, Lockhart, AC, Erinjeri, JP, Scaltriti, M, Ulaner, GA, Patel, J, Tang, J, Beer, H, Selcuklu, SD, Hanrahan, AJ, Bouvier, N, Melcer, M, Murali, R, Schram, AM, Smyth, LM, Jhaveri, K, Li, BT, Drilon, A, Harding, JJ, Iyer, G, Taylor, BS, Berger, MF, Cutler, RE, Xu, F, Butturini, A, Eli, LD, Mann, G, Farrell, C, Lalani, AS, Bryce, RP, Arteaga, CL, Meric-Bernstam, F, Baselga, J, Solit, DB, Hyman, DM, Piha-Paul, SA, Won, H, Rodon, J, Saura, C, Shapiro, GI, Juric, D, Quinn, DI, Moreno, V, Doger, B, Mayer, IA, Boni, V, Calvo, E, Loi, S, Lockhart, AC, Erinjeri, JP, Scaltriti, M, Ulaner, GA, Patel, J, Tang, J, Beer, H, Selcuklu, SD, Hanrahan, AJ, Bouvier, N, Melcer, M, Murali, R, Schram, AM, Smyth, LM, Jhaveri, K, Li, BT, Drilon, A, Harding, JJ, Iyer, G, Taylor, BS, Berger, MF, Cutler, RE, Xu, F, Butturini, A, Eli, LD, Mann, G, Farrell, C, Lalani, AS, Bryce, RP, Arteaga, CL, Meric-Bernstam, F, Baselga, J, and Solit, DB
- Abstract
Somatic mutations of ERBB2 and ERBB3 (which encode HER2 and HER3, respectively) are found in a wide range of cancers. Preclinical modelling suggests that a subset of these mutations lead to constitutive HER2 activation, but most remain biologically uncharacterized. Here we define the biological and therapeutic importance of known oncogenic HER2 and HER3 mutations and variants of unknown biological importance by conducting a multi-histology, genomically selected, 'basket' trial using the pan-HER kinase inhibitor neratinib (SUMMIT; clinicaltrials.gov identifier NCT01953926). Efficacy in HER2-mutant cancers varied as a function of both tumour type and mutant allele to a degree not predicted by preclinical models, with the greatest activity seen in breast, cervical and biliary cancers and with tumours that contain kinase domain missense mutations. This study demonstrates how a molecularly driven clinical trial can be used to refine our biological understanding of both characterized and new genomic alterations with potential broad applicability for advancing the paradigm of genome-driven oncology.
- Published
- 2018
9. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade
- Author
-
Ayers, M, Lunceford, J, Nebozhyn, M, Murphy, E, Loboda, A, Kaufman, DR, Albright, A, Cheng, JD, Kang, SP, Shankaran, V, Piha-Paul, SA, Yearley, J, Seiwert, TY, Ribas, A, and McClanahan, TK
- Published
- 2017
10. Abstract P5-20-06: Phase 1 dose escalation with ZW25, a HER2-targeted bispecific antibody, in patients (pts) with HER2-high breast cancer (BC)
- Author
-
Hamilton, E, primary, Meric-Bernstam, F, additional, Infante, J, additional, Murthy, R, additional, Patnaik, A, additional, Piha-Paul, SA, additional, Tolcher, A, additional, Hausman, D, additional, Royer, N, additional, and Beeram, M, additional
- Published
- 2018
- Full Text
- View/download PDF
11. Phase I dose-escalation study of the mTOR inhibitor sirolimus and the HDAC inhibitor vorinostat in patients with advanced malignancy
- Author
-
Park, H, Garrido-Laguna, I, Naing, A, Fu, S, Falchook, GS, Piha-Paul, SA, Wheler, JJ, Hong, DS, Tsimberidou, AM, Subbiah, V, Zinner, RG, Kaseb, AO, Patel, S, Fanale, MA, Velez-Bravo, VM, Meric-Bernstam, F, Kurzrock, R, and Janku, F
- Subjects
Sirolimus ,Adult ,Male ,Vorinostat ,Maximum Tolerated Dose ,Adolescent ,TOR Serine-Threonine Kinases ,Oncology and Carcinogenesis ,Middle Aged ,phase I ,Hydroxamic Acids ,Histone Deacetylase Inhibitors ,Young Adult ,HDAC ,hemic and lymphatic diseases ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,mTOR ,Humans ,Female ,Aged - Abstract
Preclinical models suggest that histone deacetylase (HDAC) and mammalian target of rapamycin (mTOR) inhibitors have synergistic anticancer activity. We designed a phase I study to determine the safety, maximum tolerated dose (MTD), recommended phase II dose (RP2D), and dose-limiting toxicities (DLTs) of combined mTOR inhibitor sirolimus (1 mg-5 mg PO daily) and HDAC inhibitor vorinostat (100 mg-400 mg PO daily) in patients with advanced cancer. Seventy patients were enrolled and 46 (66%) were evaluable for DLT assessment since they completed cycle 1 without dose modification unless they had DLT. DLTs comprised grade 4 thrombocytopenia (n = 6) and grade 3 mucositis (n = 1). Sirolimus 4 mg and vorinostat 300 mg was declared RP2D because MTD with sirolimus 5 mg caused significant thrombocytopenia. The grade 3 and 4 drug-related toxic effects (including DLTs) were thrombocytopenia (31%), neutropenia (8%), anemia (7%), fatigue (3%), mucositis (1%), diarrhea (1%), and hyperglycemia (1%). Of the 70 patients, 35 (50%) required dose interruption or modification and 61 were evaluable for response. Partial responses were observed in refractory Hodgkin lymphoma (-78%) and perivascular epithelioid tumor (-54%), and stable disease in hepatocellular carcinoma and fibromyxoid sarcoma. In conclusion, the combination of sirolimus and vorinostat was feasible, with thrombocytopenia as the main DLT. Preliminary anticancer activity was observed in patients with refractory Hodgkin lymphoma, perivascular epithelioid tumor, and hepatocellular carcinoma.
- Published
- 2016
12. Abstract S5-07: Preliminary efficacy and safety of pembrolizumab (MK-3475) in patients with PD-L1–positive, estrogen receptor-positive (ER+)/HER2-negative advanced breast cancer enrolled in KEYNOTE-028
- Author
-
Rugo, HS, primary, Delord, J-P, additional, Im, S-A, additional, Ott, PA, additional, Piha-Paul, SA, additional, Bedard, PL, additional, Sachdev, J, additional, Le Tourneau, C, additional, van Brummelen, E, additional, Varga, A, additional, Saraf, S, additional, Pietrangelo, D, additional, Karantza, V, additional, and Tan, A, additional
- Published
- 2016
- Full Text
- View/download PDF
13. Abstract PD5-05: Neratinib for ERBB2 mutant, HER2 non-amplified, metastatic breast cancer: Preliminary analysis from a multicenter, open-label, multi-histology phase II basket trial
- Author
-
Hyman, DM, primary, Piha-Paul, SA, additional, Rodón, J, additional, Saura, C, additional, Puzanov, I, additional, Shapiro, GI, additional, Loi, S, additional, Joensuu, H, additional, Hanrahan, AJ, additional, Modi, S, additional, Lalani, AS, additional, Xu, F, additional, Garza, SJ, additional, Cutler, RE, additional, Bryce, R, additional, Meric-Bernstam, F, additional, Baselga, J, additional, and Solit, DB, additional
- Published
- 2016
- Full Text
- View/download PDF
14. Abstract P4-13-11: Everolimus, letrozole and trastuzumab in estrogen receptor and HER2-positive patients with metastatic breast cancer and other solid tumors: Evaluating synergy and overcoming resistance
- Author
-
Janku, F, primary, Hong, DS, additional, Atkins, JT, additional, Karp, DD, additional, Tsimberidou, AM, additional, Piha-Paul, SA, additional, Subbiah, V, additional, Naing, A, additional, Fu, S, additional, Moulder, SL, additional, Tripathy, D, additional, Meric-Bernstam, F, additional, and Wheler, JJ, additional
- Published
- 2016
- Full Text
- View/download PDF
15. PD09-01: Target-Based Therapeutic Matching in Early-Phase Clinical Trials in Patients with Advanced Breast Cancer and PIK3CA Mutations.
- Author
-
Janku, F, primary, Moulder, SL, additional, Wheler, JJ, additional, Stepanek, V, additional, Falchook, GS, additional, Naing, A, additional, Hong, DS, additional, Fu, S, additional, Piha-Paul, SA, additional, Luthra, R, additional, Tsimberidou, AM, additional, and Kurzrock, R, additional
- Published
- 2011
- Full Text
- View/download PDF
16. Antitumour activity of neratinib in patients with HER2-mutant advanced biliary tract cancers
- Author
-
James J. Harding, Sarina A. Piha-Paul, Ronak H. Shah, Jessica J. Murphy, James M. Cleary, Geoffrey I. Shapiro, David I. Quinn, Irene Braña, Victor Moreno, Mitesh Borad, Sherene Loi, Iben Spanggaard, Haeseong Park, James M. Ford, Mónica Arnedos, Salomon M. Stemmer, Christelle de la Fouchardiere, Christos Fountzilas, Jie Zhang, Daniel DiPrimeo, Casey Savin, S. Duygu Selcuklu, Michael F. Berger, Lisa D. Eli, Funda Meric-Bernstam, Komal Jhaveri, David B. Solit, Ghassan K. Abou-Alfa, Institut Català de la Salut, [Harding JJ] Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. Department of Medicine, Weill Cornell Medical College, New York, NY, USA. [Piha-Paul SA] Department of Investigational Cancer Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA. [Shah RH, Murphy JJ] Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. Kravis Center for Molecular Oncology, Sloan Kettering Institute, New York, NY, USA. [Cleary JM, Shapiro GI] Dana-Farber Cancer Institute, Boston, MA, USA. [Braña I] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Molecular Therapeutic Research Unit – UITM-La Caixa, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Multidisciplinary ,acciones y usos químicos::acciones farmacológicas::mecanismos moleculares de acción farmacológica::inhibidores enzimáticos::inhibidores de proteínas cinasas [COMPUESTOS QUÍMICOS Y DROGAS] ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,General Physics and Astronomy ,Neoplasms::Neoplasms by Site::Digestive System Neoplasms::Biliary Tract Neoplasms [DISEASES] ,General Chemistry ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Otros calificadores::Otros calificadores::/efectos adversos [Otros calificadores] ,Chemical Actions and Uses::Pharmacologic Actions::Molecular Mechanisms of Pharmacological Action::Enzyme Inhibitors::Protein Kinase Inhibitors [CHEMICALS AND DRUGS] ,General Biochemistry, Genetics and Molecular Biology ,Tracte biliar - Càncer - Tractament ,Other subheadings::Other subheadings::/adverse effects [Other subheadings] ,neoplasias::neoplasias por localización::neoplasias del sistema digestivo::neoplasias del tracto biliar [ENFERMEDADES] ,Proteïnes quinases - Inhibidors - Efectes secundaris - Abstract
Antitumour activity; Neratinib; Biliary tract cancers Actividad antitumoral; Neratinib; Cánceres de vías biliares Activitat antitumoral; Neratinib; Càncers de vies biliars 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 (ClinicalTrials.gov: NCT01953926). The primary objective of the BTC cohort, which is now complete, is first objective response rate (ORR) to neratinib 240 mg orally daily. Secondary objectives include confirmed ORR, clinical benefit rate, progression-free survival, duration of response, overall survival, safety and tolerability. Genomic analyses were exploratory. Among 25 treatment-refractory patients (11 cholangiocarcinoma, 10 gallbladder, 4 ampullary cancers), the ORR is 16% (95% CI 4.5–36.1%). The most common HER2 mutations are S310F (n = 11; 48%) and V777L (n = 4; 17%). Outcomes appear 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 are identified at progression in one responder. Diarrhoea is the most common adverse event, with any-grade diarrhoea in 14 patients (56%). Although neratinib demonstrates antitumour activity in patients with refractory BTC harbouring HER2 mutations, the primary endpoint was not met and combinations may be explored. The SUMMIT trial was sponsored/funded by Puma Biotechnology, Inc. Investigators from MSKCC who participated in the trial were also supported in part by a Cancer Center Support Grant (P30 CA008748) and Cycle for Survival. Puma Biotechnology, Inc was involved in the following: study design; data collection, analysis and interpretation of the data; writing of the report; the decision to submit the article for publication. The authors would like to thank all patients and their families for participating in the SUMMIT trial. The authors acknowledge David Hyman (Memorial Sloan Kettering), Richard Bryce (Puma Biotechnology), and Alshad Lalani (Puma Biotechnology) for their important contributions to the original SUMMIT study design, oversight, and interpretation, and Feng Xu (Puma Biotechnology) and Jane Liang (Puma Biotechnology) for statistical and programming support. The authors also thank Lee Miller and Deirdre Carman (Miller Medical Communications Ltd) for medical writing/editing assistance, which was funded by Puma Biotechnology, Inc.
- Published
- 2023
17. First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas
- Author
-
Osama E. Rahma, Darren W.T. Lim, Ravit Geva, Toshikiko Doi, A. Xyrafas, Philippe L. Bedard, Jennifer Marie Mataraza, Alexander M. Lesokhin, Javier Otero, Tira Jing Ying Tan, Angad P Singh, Xinhui Chen, Jason J. Luke, Lisa Nardi, Sarina Anne Piha-Paul, Cinta Hierro, Institut Català de la Salut, [Piha-Paul SA] Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA. [Geva R] Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel. [Tan TJ] Division of Medical Oncology, National Cancer Centre Singapore, Singapore. Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada. [Lim DW] Division of Medical Oncology, National Cancer Centre Singapore, Singapore. [Hierro C] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Molecular Therapeutics Research Unit, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. [Doi T] Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Male ,Cancer Research ,Lymphoma ,Colorectal cancer ,Gastroenterology ,Antineoplastic Agents, Immunological ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Immunology and Allergy ,antibodies ,Immune Checkpoint Inhibitors ,RC254-282 ,Clinical/Translational Cancer Immunotherapy ,clinical trials as topic ,Càncer - Tractament ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Neoplasms::Neoplasms by Histologic Type::Lymphoma [DISEASES] ,Middle Aged ,drug therapy ,Oncology ,Tolerability ,Response Evaluation Criteria in Solid Tumors ,Molecular Medicine ,Adenocarcinoma ,Female ,Adult ,medicine.medical_specialty ,Maximum Tolerated Dose ,Immunology ,therapies ,neoplasias::neoplasias por tipo histológico::linfoma [ENFERMEDADES] ,investigational ,Antibodies, Monoclonal, Humanized ,Quimioteràpia combinada ,neoplasias [ENFERMEDADES] ,Pharmacokinetics ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Internal medicine ,Glucocorticoid-Induced TNFR-Related Protein ,medicine ,Humans ,Adverse effect ,Aged ,Pharmacology ,combination ,Dose-Response Relationship, Drug ,business.industry ,Endometrial cancer ,Cancer ,medicine.disease ,Neoplasms [DISEASES] ,business ,neoplasm - Abstract
BackgroundGWN323 is an IgG1 monoclonal antibody (mAb) against the glucocorticoid-induced tumor necrosis factor receptor-related protein. This first-in-human, open-label phase I/Ib study aimed to investigate the safety and tolerability and to identify the recommended doses of GWN323 with/without spartalizumab, an anti-programmed cell death receptor-1 agent, for future studies. Pharmacokinetics, preliminary efficacy and efficacy biomarkers were also assessed.MethodsPatients (aged ≥18 years) with advanced/metastatic solid tumors with Eastern Cooperative Oncology Group performance status of ≤2 were included. GWN323 (10–1500 mg) or GWN323+spartalizumab (GWN323 10–750 mg+spartalizumab 100–300 mg) were administered intravenously at various dose levels and schedules during the dose-escalation phase. Dose-limiting toxicities (DLTs) were assessed during the first 21 days in a single-agent arm and 42 days in a combination arm. Adverse events (AEs) were graded per National Cancer Institute-Common Toxicity Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors V.1.1.ResultsOverall, 92 patients (single-agent, n=39; combination, n=53) were included. The maximum administered doses (MADs) in the single-agent and combination arms were GWN323 1500 mg every 3 weeks (q3w) and GWN323 750 mg+spartalizumab 300 mg q3w, respectively. No DLTs were observed with single-agent treatment. Three DLTs (6%, all grade ≥3) were noted with combination treatment: blood creatine phosphokinase increase, respiratory failure and small intestinal obstruction. Serious AEs were reported in 30.8% and 34.0%, and drug-related AEs were reported in 82.1% and 77.4% of patients with single-agent and combination treatments, respectively. Disease was stable in 7 patients and progressed in 26 patients with single-agent treatment. In combination arm patients, 1 had complete response (endometrial cancer); 3, partial response (rectal cancer, adenocarcinoma of colon and melanoma); 14, stable disease; and 27, disease progression. GWN323 exhibited a pharmacokinetic profile typical of mAbs with a dose-dependent increase in the pharmacokinetic exposure. Inconsistent decreases in regulatory T cells and increases in CD8+ T cells were observed in the combination arm. Gene expression analyses showed no significant effect of GWN323 on interferon-γ or natural killer-cell signatures.ConclusionsGWN323, as a single agent and in combination, was well tolerated in patients with relapsed/refractory solid tumors. The MAD was 1500 mg q3w for single-agent and GWN323 750 mg+spartalizumab 300 mg q3w for combination treatments. Minimal single-agent activity and modest clinical benefit were observed with the spartalizumab combination.Trial registration numberNCT02740270.
- Published
- 2021
18. Phase 1 Study of Molibresib (GSK525762), a Bromodomain and Extra-Terminal Domain Protein Inhibitor, in NUT Carcinoma and Other Solid Tumors
- Author
-
Geraldine Ferron-Brady, Naomi B. Haas, Sophie Cousin, Christopher L. Carpenter, Christopher A. French, Meg Annan, Victor Moreno, David S. Hong, Peter J. O'Dwyer, Anastasia Wyce, Rabinder K Prinjha, Nigel J. Parr, Irene Brana, Christine L. Hann, Arindam Dhar, Yuehui Wu, Sarina Anne Piha-Paul, P. A. Cassier, Olena Barbash, Johann S. de Bono, John Hilton, Thierry Horner, Sara Duckworth Harward, Mark C. Markowski, Geoffrey I. Shapiro, Institut Català de la Salut, [Piha-Paul SA] University of Texas MD Anderson Cancer Center, Houston, TX. [Hann CL] Johns Hopkins University School of Medicine, Baltimore, MD. [French CA] Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA. [Cousin S] Medical Oncology, Institute Bergonié, Bordeaux, France. [Braña I] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. [Cassier PA] Medical Oncology, Léon Bérard Cancer Center, Lyon, France, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cancer Research ,Nausea ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Pharmacology ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,neoplasias::neoplasias por tipo histológico::neoplasias glandulares y epiteliales::carcinoma [ENFERMEDADES] ,Article ,Neoplasms::Neoplasms by Histologic Type::Neoplasms, Glandular and Epithelial::Carcinoma [DISEASES] ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Carcinoma ,Medicine ,Otros calificadores::Otros calificadores::/farmacocinética [Otros calificadores] ,Adverse effect ,030304 developmental biology ,0303 health sciences ,Farmacocinètica ,business.industry ,Càncer - Tractament ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,medicine.disease ,Dysgeusia ,Oncology ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Toxicity ,Vomiting ,Other subheadings::Other subheadings::Other subheadings::/pharmacokinetics [Other subheadings] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,medicine.symptom ,business - Abstract
Carcinoma; Concentració de fàrmac en plasma; Farmacocinètica Carcinoma; Concentración de fármaco en plasma; Farmacocinética Carcinoma; Plasma drug concentration; Pharmacokinetics Background Bromodomain and extra-terminal domain proteins are promising epigenetic anticancer drug targets. This first-in-human study evaluated the safety, recommended phase II dose, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the bromodomain and extra-terminal domain inhibitor molibresib (GSK525762) in patients with nuclear protein in testis (NUT) carcinoma (NC) and other solid tumors. Methods This was a phase I and II, open-label, dose-escalation study. Molibresib was administered orally once daily. Single-patient dose escalation (from 2 mg/d) was conducted until the first instance of grade 2 or higher drug-related toxicity, followed by a 3 + 3 design. Pharmacokinetic parameters were obtained during weeks 1 and 3. Circulating monocyte chemoattractant protein-1 levels were measured as a pharmacodynamic biomarker. Results Sixty-five patients received molibresib. During dose escalation, 11% experienced dose-limiting toxicities, including six instances of grade 4 thrombocytopenia, all with molibresib 60–100 mg. The most frequent treatment-related adverse events of any grade were thrombocytopenia (51%) and gastrointestinal events, including nausea, vomiting, diarrhea, decreased appetite, and dysgeusia (22%–42%), anemia (22%), and fatigue (20%). Molibresib demonstrated an acceptable safety profile up to 100 mg; 80 mg once daily was selected as the recommended phase II dose. Following single and repeat dosing, molibresib showed rapid absorption and elimination (maximum plasma concentration: 2 hours; t1/2: 3–7 hours). Dose-dependent reductions in circulating monocyte chemoattractant protein-1 levels were observed. Among 19 patients with NC, four achieved either confirmed or unconfirmed partial response, eight had stable disease as best response, and four were progression-free for more than 6 months. Conclusions Once-daily molibresib was tolerated at doses demonstrating target engagement. Preliminary data indicate proof-of-concept in NC. This study (BET115521; NCT01587703) was supported by GlaxoSmithKline (GSK). Financial support for this work was also provided by National Institutes of Health (NIH) Cancer Center Support Grants P30 CA016672 and P30 CA006516, as well as NIH grant R01 CA124633 to CAF.
- Published
- 2019
19. Phase II study of talazoparib in advanced cancers with BRCA1/2, DNA repair, and PTEN alterations.
- Author
-
Piha-Paul SA, Tseng C, Leung CH, Yuan Y, Karp DD, Subbiah V, Hong D, Fu S, Naing A, Rodon J, Javle M, Ajani JA, Raghav KP, Somaiah N, Mills GB, Tsimberidou AM, Zheng X, Chen K, and Meric-Bernstam F
- Abstract
Cancer cells with BRCA1/2 deficiencies are sensitive to poly (ADP-ribose) polymerase (PARP) inhibitors. We evaluated the efficacy of talazoparib in DNA-Damage Repair (DDR)-altered patients. In this phase II trial, patients were enrolled onto one of four cohorts based on molecular alterations: (1) somatic BRCA1/2, (2) other homologous recombination repair pathway, (3) PTEN and (4) germline BRCA1/2. The primary endpoint was a clinical benefit rate (CBR): complete response, partial response or stable disease ≥24 weeks. 79 patients with a median of 4 lines of therapy were enrolled. CBR for cohorts 1-4 were: 32.5%, 19.7%, 9.4% and 30.6%, respectively. PTEN mutations correlated with reduced survival and a trend towards shorter time to progression.Talazoparib demonstrated clinical benefit in selected DDR-altered patients. PTEN mutations/loss patients derived limited clinical benefit. Further study is needed to determine whether PTEN is prognostic or predictive of response to PARP inhibitors., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. A phase I study of TAK-659 and paclitaxel in patients with taxane-refractory advanced solid tumors.
- Author
-
Gouda MA, Shunyakova J, Naing A, Dumbrava E, Hong DS, Yuan Y, Yang P, Myers A, Liang Y, Peng J, Karp D, Tsimberidou AM, Rodon J, Yap TA, Piha-Paul SA, Meric-Bernstam F, and Fu S
- Subjects
- Humans, Female, Middle Aged, Aged, Male, Adult, Drug Resistance, Neoplasm, Taxoids therapeutic use, Taxoids pharmacology, Maximum Tolerated Dose, Syk Kinase metabolism, Paclitaxel therapeutic use, Paclitaxel pharmacology, Paclitaxel administration & dosage, Neoplasms drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology
- Abstract
Background: Paclitaxel resistance limits durability of response in patients with initial clinical benefit. Overexpression of spleen tyrosine kinase (SYK) has been proposed as a possible resistance mechanism. This phase I trial evaluated the safety and preliminary activity of the SYK inhibitor TAK-659 combined with paclitaxel in patients with advanced taxane-refractory solid tumors., Patients and Methods: Patients with advanced solid tumors and prior progression on taxane-based therapy received intravenous infusion of paclitaxel on days 1, 8, and 15 plus oral TAK-659 daily in 28-day cycles. The dose-escalation phase included six cohorts treated at different dose levels; the dose-expansion phase included patients with ovarian cancer treated at the highest dose level. Toxicity was graded using the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. Efficacy was evaluated using Response Evaluation Criteria in Solid Tumors version 1.1., Results: Our study included 49 patients. Maximum tolerated dose was not reached, but higher rates of adverse events were observed at higher dose levels. There were no treatment-related deaths. The most common treatment-related adverse events of any grade were increased aspartate aminotransferase (n = 31; 63%), increased alanine aminotransferase (n = 26; 53%), decreased neutrophil count (n = 26; 53%), and decreased white blood cell count (n = 26; 53%). Most adverse events were either grade 1 or 2. In the 44 patients with evaluable disease, 12 (27%) had stable disease as the best overall response, including three patients with prolonged stable disease, and 4 patients (9%) achieved a partial response., Conclusions: The combination of paclitaxel and TAK-659 showed preliminary activity possibly overcoming resistance to taxane-based therapy as well as a tolerable safety profile in patients with advanced solid tumors., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Initial Chemotherapy for Locally Advanced and Metastatic NUT Carcinoma.
- Author
-
Luo J, Sanchez M, Lee E, Hertzler H, Luong N, Mazzola E, Finstein B, Tamen R, Brisbane G, Nguyen T, Paik PK, Chaft JE, Cheng ML, Khalil H, Piha-Paul SA, Sholl LM, Nishino M, Jänne PA, DuBois SG, Hanna GJ, Shapiro GI, and French CA
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Aged, Aged, 80 and over, Young Adult, Adolescent, Child, Ifosfamide administration & dosage, Ifosfamide therapeutic use, Survival Rate, Nuclear Proteins genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Lung Neoplasms mortality, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
Introduction: NUT carcinoma (NC) is an underdiagnosed and aggressive poorly differentiated or squamous cell cancer. A subset of NC is sensitive to chemotherapy, but the optimal regimen is unknown. Experts have recommended platinum- and ifosfamide-based therapy based on case reports., Methods: Patients with pathologically confirmed NC with known survival outcomes after chemotherapy and consented to participate in a worldwide registry were studied. Results were summarized using descriptive methods., Results: The study included 118 patients with NC. Median age was 34 (range: 1-82) years, 39% were women, and 61% harbored a BRD4::NUTM1 fusion. Patients received platinum (74%) or ifosfamide (26%, including regimens with both, 13%). Of 62 patients with nonmetastatic disease, 40% had a thoracic primary. Compared with platinum-based chemotherapy, patients who received ifosfamide-based chemotherapy had nominally higher progression-free survival (12 mo: 59% [95% CI: 32-87] versus 37% [95% CI: 22-52], hazard ratio = 0.68 [0.32, 1.42], p = 0.3) but not overall survival (OS). Among the 56 patients with metastatic disease, 80% had a thoracic primary. Ifosfamide had an objective response rate (ORR) of 75% (six of eight) and platinum had an ORR of 31% (11 of 36). Nevertheless, there was no difference in progression-free survival or OS. The 3-year OS of the entire cohort was 19% (95% CI: 10%-28%). Of the 11 patients alive greater than 3 years, all presented with nonmetastatic and operable or resectable disease., Conclusion: There is a numerically higher ORR for ifosfamide-based therapy compared with platinum-based therapy, with limited durability. OS at 3 years is only 19%, and development of effective therapies is an urgent unmet need for this patient population., Competing Interests: Disclosure Dr. Luo reports receiving research support to her institution from Erasca, Genentech, Kronos Bio, Novartis, and Revolution Medicines; receiving honoraria from Targeted Oncology, Physicians’ Education Resource, VJ Oncology, Cancer GRACE, and Community Cancer Education, Inc.; having advisory board participation from Astellas, AstraZeneca, and Amgen; receiving personal fees from Erasca, Blueprint Medicines, and Daiichi Sankyo; and having a pending patent filed by Memorial Sloan Kettering related to multimodal features to predict response to immunotherapy (PCT/US2023/115872). Dr. Paik receives compensation for consulting or advisory board participation from Bicara Therapeutics, Inc., EMD Serono, Inc., Novartis, Mirati Therapeutics, and Janssen; and receives honoraria for participation in CME educational programs from IDEOlogy, Excerpta Medica, PeerVoice, Physicians Education Resource, Medscape, Agile, Axis Medical Education, Aptitude Health, MJH, Annenberg Center, Cardinal Health, and Touch Independent Medical Education Ltd. Dr. Chaft has served as a consultant for AstraZeneca, Bristol-Myers Squib, Genentech, Merck, Flame Biosciences, Novartis, Regeneron-Sanofi, Guardant Health, and Janssen; and has received research funding to her institution from AstraZeneca, Bristol-Myers Squib, Genentech, and Merck. Dr. Cheng reports receiving honoraria from Lynx Group, WebMD, and Potomac Center for Medical Education; having consulting or advisory role for AstraZeneca, Boehringer Ingelheim, Mirati Therapeutics, Cepheid, Janssen, and Pfizer; receiving research funding from Palleon Pharmaceuticals (Inst); and receiving travel, accommodations, and expenses from Daiichi Sankyo, AstraZeneca, and Genzyme. Dr. Piha-Paul reports receiving clinical trial research support/grant funding through the institution from AbbVie, Inc., ABM Therapeutics, Inc., Acepodia, Inc., Alkermes, Aminex Therapeutics, BioMarin Pharmaceutical, Inc., Boehringer Ingelheim, Bristol-Myers Squib, Cerulean Pharma, Inc., Chugai Pharmaceutical Co., Ltd., Curis, Inc., Cyclacel Pharmaceuticals, Daiichi Sankyo, Eli Lilly, ENB Therapeutics, Epigenetix Inc., Five Prime Therapeutics, F-Star Beta Limited, F-Star Therapeutics, Gene Quantum, Genmab A/S, Gilead Sciences, Inc., GlaxoSmithKline, Helix BioPharma Corp., Hengrui Pharmaceuticals, Co., Ltd., HiberCell, Inc., Immorna Biotherapeutics, Inc., Immunomedics, Inc., Incyte Corp., Jacobio Pharmaceuticals Co., Ltd., Jiangsu Simcere Pharmaceutical Co., Ltd., Loxo Oncology, Inc., Lytix Biopharma AS, Medimmune, LLC, Medivation, Inc., Merck Sharp and Dohme Corp., Nectin Therapeutics, Ltd., Novartis Pharmaceuticals, Nurix, Pieris Pharmaceuticals, Inc., Pfizer, Phanes Therapeutics, Principia Biopharma, Inc., Puma Biotechnology, Inc., Purinomia Biotech, Inc., Rapt Therapeutics, Inc., Replimune, Roche/Blueprint, Seattle Genetics, Silverback Therapeutics, Shasqi, Inc., Synlogic Therapeutics, Taiho Oncology, Tesaro, Inc., Theradex Oncology, Toragen Therapeutics, Inc., TransThera Bio, Xencor, Inc., ZielBio, Inc., NCI/NIH, and P30CA016672 – Core Grant (CCSG Shared Resources); and having consulting for CRC Oncology. Dr. Sholl reports receiving research support to her institution from Genentech; and consulting from GV20 Therapeutics, Genentech, and Lilly. Dr. Nishino reports having consulting for AstraZeneca; and receiving research grants to the institution from Canon Medical Systems, AstraZeneca, Daiichi Sankyo, and Konica-Minolta. Dr. Jänne reports receiving grants from The Mark Foundation for Cancer Research and the American Cancer Society during the conduct of the study; personal fees outside of submitted work from Boehringer Ingelheim, Pfizer, Roche/Genentech, Chugai Pharmaceuticals, Ignyta, LOXO Oncology, Eli Lilly, SFJ Pharmaceuticals, Voronoi, Daiichi Sankyo, Biocartis, Novartis, Sanofi, Takeda Oncology, Transcenta, Silicon Therapeutics, Syndax, Nuvalent, Bayer, Esai, and Mirati Therapeutics; and grants outside the submitted work from Boehringer Ingelheim, Eli Lilly, Daiichi Sankyo, Takeda Oncology, PUMA, Astellas Pharmaceuticals, and Revolution Medicines. Dr. DuBois reports receiving honoraria or having advisory board participation from Amgen, Bayer, and Jazz Pharmaceuticals; and receiving travel expenses from LOXO Oncology, Roche, and Salarius, all outside the submitted work. Dr. Hanna reports receiving grants/contracts outside the submitted work from ACCRF, Actuate Therapeutics, ASCO CCF, Bicara, Bristol-Myers Squibb, Elevar Therapeutics, Exicure, Gateway for Cancer Research, Genentech, GlaxoSmithKline, ImmunityBio, Kartos, Kite, KSQ, Kura Oncology, Regeneron, Repertoire, Sanofi Genzyme, Secura Bio, and V Foundation; and having advisory role and/or receiving honoraria outside the submitted work from Bicara, Bio-Rad, Boxer Capital, Bristol-Myers Squibb, Coherus, Elevar, Exicure, General Catalyst, Guardian Bio, KSQ, Kura Oncology, Massachusetts Medical Society, Merck, Naveris, Nextech, Prelude, Rain, Regeneron, Remix, Replimune, Sanofi Genzyme, SIRPant, and Surface Oncology. Dr. Shapiro reports receiving personal fees from Merck KGaA/EMD-Serono, Bicycle Therapeutics, Cybrexa Therapeutics, Boehringer Ingelheim, Bayer, ImmunoMet, Concarlo Holdings, Syros, Zentalis, CytomX Therapeutics, Blueprint Medicines, Kymera Therapeutics, Janssen, Xinthera, and Artios; receiving grants from Merck KGaA/EMD-Serono, Tango, Bristol-Myers Squibb, Pfizer, and Eli Lilly; and having a patent for “Dosage regimen for sapacitabine and seliciclib” issued to Cyclacel Pharmaceuticals and GIS and a patent for “Compositions and methods for predicting response and resistance to CDK4/6 inhibition” issued to Liam Cornell and GIS. Dr. French reports receiving research funding from Boehringer Ingelheim and consultant fees from Boehringer Ingelheim. The remaining authors declare no conflict of interest., (Copyright © 2023 International Association for the Study of Lung Cancer. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. First-In-Human Phase I Study of Tinengotinib (TT-00420), a Multiple Kinase Inhibitor, as a Single Agent in Patients With Advanced Solid Tumors.
- Author
-
Piha-Paul SA, Xu B, Dumbrava EE, Fu S, Karp DD, Meric-Bernstam F, Hong DS, Rodon JA, Tsimberidou AM, Raghav K, Ajani JA, Conley AP, Mott F, Fan Y, Fan J, Peng P, Wang H, Ni S, Sun C, Qiang X, Levin WJ, Ngo B, Ru QC, Wu F, and Javle MM
- Subjects
- Male, Humans, Bayes Theorem, Vascular Endothelial Growth Factor A, Maximum Tolerated Dose, Triple Negative Breast Neoplasms drug therapy, Prostatic Neoplasms, Castration-Resistant drug therapy, Neoplasms drug therapy, Neoplasms genetics, Antineoplastic Agents adverse effects, Cholangiocarcinoma drug therapy, Hypertension chemically induced
- Abstract
Purpose: This first-in-human phase I dose-escalation study evaluated the safety, pharmacokinetics, and efficacy of tinengotinib (TT-00420), a multi-kinase inhibitor targeting fibroblast growth factor receptors 1-3 (FGFRs 1-3), Janus kinase 1/2, vascular endothelial growth factor receptors, and Aurora A/B, in patients with advanced solid tumors., Patients and Methods: Patients received tinengotinib orally daily in 28-day cycles. Dose escalation was guided by Bayesian modeling using escalation with overdose control. The primary objective was to assess dose-limiting toxicities (DLTs), maximum tolerated dose (MTD), and dose recommended for dose expansion (DRDE). Secondary objectives included pharmacokinetics and efficacy., Results: Forty-eight patients were enrolled (dose escalation, n = 40; dose expansion, n = 8). MTD was not reached; DRDE was 12 mg daily. DLTs were palmar-plantar erythrodysesthesia syndrome (8 mg, n = 1) and hypertension (15 mg, n = 2). The most common treatment-related adverse event was hypertension (50.0%). In 43 response-evaluable patients, 13 (30.2%) achieved partial response (PR; n = 7) or stable disease (SD) ≥ 24 weeks (n = 6), including 4/11 (36.4%) with FGFR2 mutations/fusions and cholangiocarcinoma (PR n = 3; SD ≥ 24 weeks n = 1), 3/3 (100.0%) with hormone receptor (HR)-positive/HER2-negative breast cancer (PR n = 2; SD ≥ 24 weeks n = 1), 2/5 (40.0%) with triple-negative breast cancer (TNBC; PR n = 1; SD ≥ 24 weeks n = 1), and 1/1 (100.0%) with castrate-resistant prostate cancer (CRPC; PR). Four of 12 patients (33.3%; HR-positive/HER2-negative breast cancer, TNBC, prostate cancer, and cholangiocarcinoma) treated at DRDE had PRs. Tinengotinib's half-life was 28-34 hours., Conclusions: Tinengotinib was well tolerated with favorable pharmacokinetic characteristics. Preliminary findings indicated potential clinical benefit in FGFR inhibitor-refractory cholangiocarcinoma, HER2-negative breast cancer (including TNBC), and CRPC. Continued evaluation of tinengotinib is warranted in phase II trials., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
23. RAF inhibitor re-challenge therapy in BRAF-aberrant pan-cancers: the RE-RAFFLE study.
- Author
-
Nelson BE, Roszik J, Ahmed J, Barretto CMN, Nardo M, Campbell E, Johnson AM, Piha-Paul SA, Oliva ICG, Weathers SP, Cabanillas M, Javle M, Meric-Bernstam F, and Subbiah V
- Subjects
- Humans, Proto-Oncogene Proteins B-raf genetics, Protein Kinase Inhibitors therapeutic use, Retrospective Studies, Prospective Studies, Mutation, Melanoma pathology
- Abstract
Previous studies have shown the clinical benefit of rechallenging the RAF pathway in melanoma patients previously treated with BRAF inhibitors. 44 patients with multiple tumors harboring RAF alterations were rechallenged with a second RAF inhibitor, either as monotherapy or in combination with other therapies, after prior therapy with a first RAF inhibitor. This retrospective observational study results showed that rechallenging with RAFi(s) led to an overall response rate of 18.1% [PR in thyroid (1 anaplastic; 3 papillary), 1 ovarian, 2 melanoma, 1 cholangiocarcinoma, and 1 anaplastic astrocytoma]. The clinical benefit rate was 54.5%; more than 30% of patients had durable responses with PR and SD lasting > 6 months. The median progression-free survival on therapy with second RAF inhibitor in the rechallenge setting either as monotherapy or combination was shorter at 2.7 months (0.9-30.1 m) compared to 8.6 months (6.5-11.5 m) with RAF-1i. However, the median PFS with RAF-2i responders (PFS-2) improved at 12.8 months compared to 11.4 months with RAF-1i responders. The median OS from retreatment with RAF-2i was 15.5 months (11.1-30.8 m). Further prospective studies are needed to validate these results and expand targeted therapy options for RAF-aberrant cancers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. A Relative Bioavailability, Bioequivalence, and Food Effect Study of Niraparib Tablets in Patients with Advanced Solid Tumors.
- Author
-
Falchook G, Patnaik A, Richardson DL, Harvey RD, Sharma MR, Hafez N, Hamilton E, Piha-Paul SA, Barve M, Wise-Draper T, Patel MR, Dowlati A, Pascuzzo J, Tang SC, Faltermeier C, Malinowska IA, Shtessel L, Striha A, and Potocka E
- Subjects
- Humans, Area Under Curve, Biological Availability, Cross-Over Studies, Fasting, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Tablets pharmacokinetics, Therapeutic Equivalency, Antineoplastic Agents pharmacology, Indazoles, Neoplasms drug therapy, Piperidines
- Abstract
Purpose: The poly (ADP-ribose) polymerase inhibitor niraparib is indicated as maintenance treatment in patients with certain subtypes of advanced ovarian cancer, and is being investigated in patients with other solid tumors. Niraparib is available in 100-mg capsules with a starting dosage of 200 or 300 mg/d. This study assessed the relative bioavailability (BA) and bioequivalence (BE) between a 1 × 300-mg tablet relative to 3 × 100-mg niraparib capsules. In addition, the food effect (FE) of a high-fat meal on the pharmacokinetic (PK) properties of tablet-formulated niraparib was investigated., Methods: This was a US-based, 3-stage, open-label, multicenter, single-crossover, randomized-sequence study. Enrolled patients were 18 years and older, with histologically or cytologically confirmed advanced solid tumors (metastatic or local) and disease progression despite standard therapy. Patients were randomly assigned 1:1 to receive niraparib 1 × 300-mg tablet or 3 × 100-mg capsules in the BA and BE stages or 1 × 300-mg tablet in a fasted or fed (high-fat meal) state in the FE stage. Across all study stages, PK parameters were assessed for 7 days after each dose (tablet or capsule) or prandial state (fasted or fed). In the BA stage, patients crossed over to the other treatment after a 7-day washout period, which was extended to 14 days in the BE and FE stages. Tolerability was assessed for patients who received any amount of niraparib., Findings: The BA-, BE-, and FE-evaluable populations comprised 23, 108, and 19 patients, respectively, who completed both treatment periods in each study stage, had sufficient concentration data to accurately estimate PK parameters without niraparib carryover, and did not experience disqualifying events. PK parameters were similar after dosing with tablet or capsule formulations; the 90% CIs of the geometric least square means for C
max , AUC0-t , and AUC0-∞ were within the 0.80 to 1.25 BE limits. In the FE stage, Cmax , AUC0-t , and AUC0-∞ were 11%, 32%, and 28% higher, respectively, in the fed versus fasted state. The safety population included 29, 168, and 28 patients in the BA, BE, and FE stages, respectively, who received niraparib. No new safety signals were identified., Implications: Niraparib tablets were found to be bioequivalent to capsules. A modest (≤32%) FE was observed with a high-fat meal, but was not considered to be clinically meaningful, given niraparib's PK variability., Clinicaltrials: gov identifier: NCT03329001. (Clin Ther. 2024;46:XXX-XXX) © 2024 Elsevier HS Journals, Inc., Competing Interests: Declaration of competing interest This study was funded by GSK (GSK study 213362). GSK contributed to the study design, implementation, data collection, interpretation, and analysis. All authors had full access to the data and had final responsibility for the decision to submit for publication. Medical writing support was provided by Nicholas Thomas, PhD, of Fishawack Indicia Ltd, UK, part of Avalere Health, and funded by GSK., (Copyright © 2024 GSK. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
25. Phase I Study of mTORC1/2 Inhibitor Sapanisertib (CB-228/TAK-228) in Combination with Metformin in Patients with mTOR/AKT/PI3K Pathway Alterations and Advanced Solid Malignancies.
- Author
-
Subbiah V, Coleman N, Piha-Paul SA, Tsimberidou AM, Janku F, Rodon J, Pant S, Dumbrava EEI, Fu S, Hong DS, Zhang S, Sun M, Jiang Y, Roszik J, Song J, Yuan Y, Meric-Bernstam F, and Naing A
- Subjects
- Male, Humans, Female, Middle Aged, Proto-Oncogene Proteins c-akt genetics, Phosphatidylinositol 3-Kinases genetics, Mechanistic Target of Rapamycin Complex 1, AMP-Activated Protein Kinases, TOR Serine-Threonine Kinases genetics, Diarrhea, Adenosine Triphosphate, Metformin adverse effects, Leiomyosarcoma, Exanthema, Adenine analogs & derivatives, Benzoxazoles
- Abstract
Background: Sapanisertib (CB-228/TAK-228) is a potent, selective ATP-competitive, dual inhibitor of mTORC1/2. Metformin is thought to inhibit the mTOR pathway through upstream activation of 5'-AMP-activated protein kinase (AMPK) suggesting combination therapy may enhance antitumor activity of sapanisertib. We report preliminary safety, tolerability, and efficacy from the dose-escalation study of sapanisertib in combination with metformin in patients with advanced solid tumors., Methods: Patients with advanced metastatic solid tumors resistant or refractory to standard treatment, with and without mTOR/AKT/PI3K pathway alterations, received sapanisertib 3 or 4 mg daily together with metformin once to three times daily (500-1,500 mg). All patients underwent 14-day titration period for metformin in cycle 1. Tumor measurements were performed following cycle 2 and subsequently every 8 weeks., Results: A total of 30 patients were enrolled across four cohorts (3 mg/500 mg; 3 mg/1,000 mg, 4 mg/1,000 mg; 4 mg/1,500 mg). 19 were female (63%), median age was 57 (range: 30-77), all were Eastern Cooperative Oncology Group performance status 1. Tumor types included sarcoma (6), breast (4), ovarian (4), head and neck (3), colorectal (2), lung (2), renal cell (2), endometrial (2), gastroesophageal junction (1), prostate (1), stomach (1), urachus (1), and cervical cancer (1). Median number of prior lines of therapy was 4. Most common genomic alterations included PIK3CA (27%), PTEN (17%), AKT1/2 (10%), mTOR (10%). Of 30 patients evaluable for response, 4 patients achieved partial response (PR); 15 patients achieved stable disease (SD) as best response. Disease control rate (PR+SD) was 63%. Of the responders in PR, 3 of 4 patients had documented PTEN mutations (3/5 patients enrolled with PTEN mutations had PR); 2 of 4 of patients in PR had comutations (patient with leiomyosarcoma had both PTEN and TSC; patient with breast cancer had both PTEN and STK11); 1 of 4 patients in PR had AKT and mTOR mutation; tumor types included leiomyosarcoma (n = 2), breast (n = 1), and endometrial cancer (n = 1). Most common treatment-emergent adverse events included nausea, anorexia, diarrhea, and rash. Grade (G) 3-5 treatment-related adverse events included hyperglycemia (4/30; 13%), fatigue (2/30; 7%), hypertriglyceridemia (1/30; 3%), rash (2/20; 7%), diarrhea (2/30; 7%), creatinine increase (1/30; 3%), acidosis (1/30; 3%). No dose-limiting toxicities (DLT) were reported in the 3 mg/500 mg cohort. One of 6 patient had DLT in the 3 mg/1,000 mg cohort (G3 diarrhea) and 2 of 11 patients had DLTs in the 4 mg/1,500 mg cohort (G3 fatigue, G3 rash). 4 mg/1,000 mg was defined as the MTD., Conclusions: The safety profile of mTORC1/2 inhibitor sapanisertib in combination with metformin was generally tolerable, with antitumor activity observed in patients with advanced malignancies harboring PTEN mutations and AKT/mTOR pathway alterations., Significance: Sapanisertib (CB-228/TAK-228) is a potent, selective ATP-competitive, next-generation dual inhibitor of mTORC1/2. Metformin is thought to inhibit the mTOR pathway through upstream activation of AMPK suggesting combination therapy may enhance antitumor activity of sapanisertib. This dose-escalation study of sapanisertib and metformin in advanced solid tumors and mTOR/AKT/PI3K pathway alterations, demonstrates safety, tolerability, and early clinical activity in advanced malignancies harboring PTEN mutations and AKT/mTOR pathway alterations.Clinical trial information: NCT03017833., (© 2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
26. Phase I study of sapanisertib (CB-228/TAK-228/MLN0128) in combination with ziv-aflibercept in patients with advanced solid tumors.
- Author
-
Coleman N, Stephen B, Fu S, Karp D, Subbiah V, Ahnert JR, Piha-Paul SA, Wright J, Fessahaye SN, Ouyang F, Yilmaz B, Meric-Bernstam F, and Naing A
- Subjects
- Humans, Female, Middle Aged, Male, Treatment Outcome, Receptors, Vascular Endothelial Growth Factor therapeutic use, Recombinant Fusion Proteins adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoplasms drug therapy, Neoplasms etiology, Adenine analogs & derivatives, Benzoxazoles
- Abstract
Background: Sapanisertib is a potent ATP-competitive, dual inhibitor of mTORC1/2. Ziv-aflibercept is a recombinant fusion protein comprising human VEGF receptor extracellular domains fused to human immunoglobulin G1. HIF-1α inhibition in combination with anti-angiogenic therapy is a promising anti-tumor strategy. This Phase 1 dose-escalation/expansion study assessed safety/ tolerability of sapanisertib in combination with ziv-aflibercept in advanced solid tumors., Methods: Fifty-five patients with heavily pre-treated advanced metastatic solid tumors resistant or refractory to standard treatment received treatment on a range of dose levels., Results: Fifty-five patients were enrolled and treated across a range of dose levels. Forty were female (73%), median age was 62 (range: 21-79), and ECOG PS was 0 (9, 16%) or 1 (46, 84%). Most common tumor types included ovarian (8), colorectal (8), sarcoma (8), breast (3), cervical (4), and endometrial (4). Median number of prior lines of therapy was 4 (range 2-11). Sapanisertib 4 mg orally 3 days on and 4 days off plus 3 mg/kg ziv-aflibercept IV every 2 weeks on a 28-day cycle was defined as the maximum tolerated dose. Most frequent treatment-related grade ≥2 adverse events included hypertension, fatigue, anorexia, hypertriglyceridemia, diarrhea, nausea, mucositis, and serum lipase increase. There were no grade 5 events. In patients with evaluable disease (n = 50), 37 patients (74%) achieved stable disease (SD) as best response, two patients (4%) achieved a confirmed partial response (PR); disease control rate (DCR) (CR + SD + PR) was 78%., Conclusion: The combination of sapanisertib and ziv-aflibercept was generally tolerable and demonstrated anti-tumor activity in heavily pre-treated patients with advanced malignancies., (© 2024 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
27. Phase 1/2 trial of avelumab combined with utomilumab (4-1BB agonist), PF-04518600 (OX40 agonist), or radiotherapy in patients with advanced gynecologic malignancies.
- Author
-
Knisely A, Ahmed J, Stephen B, Piha-Paul SA, Karp D, Zarifa A, Fu S, Hong DS, Rodon Ahnert J, Yap TA, Tsimberidou AM, Alshawa A, Dumbrava EE, Yang Y, Song J, Meric-Bernstam F, Jazaeri AA, and Naing A
- Subjects
- Humans, Female, Neoplasm Recurrence, Local pathology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Genital Neoplasms, Female drug therapy, Uterine Cervical Neoplasms drug therapy, Immunoglobulin G, Antibodies, Monoclonal, Humanized
- Abstract
Background: Immune checkpoint blockade has shown mixed results in advanced/recurrent gynecologic malignancies. Efficacy may be improved through costimulation with OX40 and 4-1BB agonists. The authors sought to evaluate the safety and efficacy of avelumab combined with utomilumab (a 4-1BB agonist), PF-04518600 (an OX40 agonist), and radiotherapy in patients with recurrent gynecologic malignancies., Methods: The primary end point in this six-arm, phase 1/2 trial was safety of the combination regimens. Secondary end points included the objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors and immune-related Response Evaluation Criteria in Solid Tumors, the disease control rate (DCR), the duration of response, progression-free survival, and overall survival., Results: Forty patients were included (35% with cervical cancer, 30% with endometrial cancer, and 35% with ovarian cancer). Most patients (n = 33; 83%) were enrolled in arms A-C (no radiation). Among 35 patients who were evaluable for efficacy, the ORR was 2.9%, and the DCR was 37.1%, with a median duration of stable disease of 5.4 months (interquartile range, 4.1-7.3 months). Patients with cervical cancer in arm A (avelumab and utomilumab; n = 9 evaluable patients) achieved an ORR of 11% and a DCR of 78%. The median progression-free survival was 2.1 months (95% CI, 1.8-3.5 months), and overall survival was 9.4 months (95% CI, 5.6-11.9 months). No dose-limiting toxicities or grade 3-5 immune-related adverse events were observed., Conclusions: The findings from this trial highlight that, in heavily pretreated patients with gynecologic cancer, even multidrug regimens targeting multiple immunologic pathways, although safe, did not produce significant responses. A DCR of 78% in patients with cervical cancer who received avelumab and utomilumab indicates that further research on this combination in select patients may be warranted., (© 2023 American Cancer Society.)
- Published
- 2024
- Full Text
- View/download PDF
28. A Phase I Trial of Bevacizumab and Temsirolimus in Combination With Valproic Acid in Advanced Solid Tumors.
- Author
-
Nelson BE, Tsimberidou AM, Fu X, Fu S, Subbiah V, Sood AK, Rodon J, Karp DD, Blumenschein G, Kopetz S, Pant S, and Piha-Paul SA
- Subjects
- Female, Humans, Middle Aged, Bevacizumab adverse effects, Valproic Acid adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Ischemia drug therapy, Ischemia etiology, Maximum Tolerated Dose, Mucositis, Neoplasms drug therapy, Neoplasms pathology, Thrombocytopenia drug therapy, Lymphopenia
- Abstract
Background: Preclinical models suggest synergy between anti-angiogenesis therapy, mammalian target of rapamycin (mTOR), and histone deacetylase inhibitors to promote anticancer activity., Methods: This phase I study enrolled 47 patients between April 2012 and 2018 and determined safety, maximum tolerated dose (MTD), and dose-limiting toxicities (DLTs) when combining bevacizumab, temsirolimus, and valproic acid in patients with advanced cancer., Results: Median age of enrolled patients was 56 years. Patients were heavily pretreated with a median of 4 lines of prior therapy. Forty-five patients (95.7%) experienced one or more treatment-related adverse events (TRAEs). Grade 3 TRAEs were lymphopenia (14.9%), thrombocytopenia (8.5%), and mucositis (6.4%). Grade 4 TRAEs included lymphopenia (2.1%) and CNS cerebrovascular ischemia (2.1%). Six patients developed DLTs across 10 dose levels with grade 3 infection, rash, mucositis, bowel perforation, elevated lipase, and grade 4 cerebrovascular ischemia. The MTD was dose level 9 (bevacizumab 5 mg/kg days 1 and 15 intravenously (IV) plus temsirolimus 25 mg days 1, 8, 15, and 22 IV and valproic acid 5 mg/kg on days 1-7 and 15-21 per orally (PO)). Objective response rate (ORR) was 7.9% with confirmed partial response (PRs) in 3 patients (one each in parotid gland, ovarian, and vaginal cancers). Stable disease (SD) ≥+6 months was seen in 5 patients (13.1%). Clinical benefit state (CBR: PR + SD ≥+6 months) was 21%., Conclusion: Combination therapy with bevacizumab, temsirolimus, and valproic acid was feasible, but there were numerous toxicities, which will require careful management for future clinical development (ClinicalTrials.gov Identifier: NCT01552434)., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
29. Everolimus in combination with vandetanib in children, adolescents, and young adults: a phase I study.
- Author
-
Phadnis S, Wang X, Daw NC, Herzog CE, Subbiah IM, Zaky W, Gouda MA, Morani AC, Amini B, Harrison DJ, Piha-Paul SA, Meric-Bernstam F, Gorlick R, Schwartz CL, and Subbiah V
- Subjects
- Humans, Young Adult, Adolescent, Child, Vascular Endothelial Growth Factor A, Sirolimus adverse effects, Piperidines adverse effects, Quinazolines adverse effects, Everolimus adverse effects, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Background: Combined use of inhibitors of mammalian target of rapamycin (mTOR) and vascular endothelial growth factor (VEGF-2) receptors is a potential strategy to overcome resistance to either class of drugs when used alone., Patients and Methods: We designed a phase 1 trial to test the drug combination of a multikinase VEGF receptor 2 inhibitor, vandetanib, and an mTOR inhibitor, everolimus, in a pediatric and young adult patient cohort with advanced cancers. Exceptional responders were probed for tumor mutational profile to explore possible molecular mechanisms of response., Results: Among 21 enrolled patients, clinical benefit was observed in 38% (one patient with partial response and eight patients with stable disease) with a median progression-free survival of 3.3 months. The most common treatment-related adverse event was rash (n = 13). Other treatment-related toxicities included diarrhea, fatigue, hypertension, QT prolongation, hypertriglyceridemia/hypercholesterolemia, transaminitis, thrombocytopenia, and weight loss. None of the patients experienced dose-limiting toxicities. Three exceptional responders were analyzed and were found to harbor genetic alterations including kinase insert domain receptor (KDR) Q472H mutation, EWSR1-CREB3L1, CDKN2A/B loss, and ASPL/ASPSCR1-TFE3 fusion., Conclusions: The combination of vandetanib and everolimus showed early activity and tolerable toxicity profile in pediatric patients with advanced cancers., (Copyright © 2023. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
30. HER2-low expression in patients with advanced or metastatic solid tumors.
- Author
-
Uzunparmak B, Haymaker C, Raso G, Masciari S, Wang L, Lin H, Gorur A, Kirby B, Cimo AM, Kennon A, Ding Q, Urschel G, Yuan Y, Feng G, Rizvi Y, Hussain A, Zhu C, Kim P, Abbadessa G, Subbiah V, Yap TA, Rodon J, Piha-Paul SA, Meric-Bernstam F, and Dumbrava EE
- Subjects
- Humans, Female, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, In Situ Hybridization, Immunohistochemistry, Genomics methods, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms genetics, Breast Neoplasms pathology, Neoplasms, Second Primary
- Abstract
Background: Human epidermal growth factor receptor 2 (HER2)-low is a newly defined category with HER2 1+ or 2+ expression by immunohistochemistry (IHC) and lack of HER2 gene amplification measured by in situ hybridization (ISH). Much remains unknown about the HER2-low status across tumor types and changes in HER2 status between primary and metastatic samples., Patients and Methods: HER2 expression by IHC was evaluated in 4701 patients with solid tumors. We have evaluated the HER2 expression by IHC and amplification by ISH in paired breast and gastric/gastroesophageal (GEJ) primary and metastatic samples. HER2 expression was correlated with ERBB2 genomic alterations evaluated by next-generation sequencing (NGS) in non-breast, non-gastric/GEJ samples., Results: HER2 expression (HER2 IHC 1-3+) was found in half (49.8%) of the cancers, with HER2-low (1 or 2+) found in many tumor types: 47.1% in breast, 34.6% in gastric/GEJ, 50.0% in salivary gland, 46.9% in lung, 46.5% in endometrial, 46% in urothelial, and 45.5% of gallbladder cancers. The concordance evaluation of HER2 expression between primary and metastatic breast cancer samples showed that HER2 3+ remained unchanged in 87.1% with a strong agreement between primary and metastatic samples, with a weighted kappa (Κ) of 0.85 (95% confidence interval 0.79-0.91). ERBB2 alterations were identified in 117 (7.5%) patients with non-breast, non-gastric/GEJ solid tumors who had NGS testing. Of 1436 patients without ERBB2 alterations, 512 (35.7%) showed any level HER2 expression by IHC., Conclusion: Our results show that HER2-low expression is frequently found across tumor types. These findings suggest that many patients with HER2-low solid tumors might benefit from HER2-targeted therapies., (Copyright © 2023. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
31. Neratinib + fulvestrant + trastuzumab for HR-positive, HER2-negative, HER2-mutant metastatic breast cancer: outcomes and biomarker analysis from the SUMMIT trial.
- Author
-
Jhaveri K, Eli LD, Wildiers H, Hurvitz SA, Guerrero-Zotano A, Unni N, Brufsky A, Park H, Waisman J, Yang ES, Spanggaard I, Reid S, Burkard ME, Vinayak S, Prat A, Arnedos M, Bidard FC, Loi S, Crown J, Bhave M, Piha-Paul SA, Suga JM, Chia S, Saura C, Garcia-Saenz JÁ, Gambardella V, de Miguel MJ, Gal-Yam EN, Rapael A, Stemmer SM, Ma C, Hanker AB, Ye D, Goldman JW, Bose R, Peterson L, Bell JSK, Frazier A, DiPrimeo D, Wong A, Arteaga CL, and Solit DB
- Subjects
- Female, Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Fulvestrant, Receptor, ErbB-2, Trastuzumab, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: HER2 mutations are targetable alterations in patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC). In the SUMMIT basket study, patients with HER2-mutant MBC received neratinib monotherapy, neratinib + fulvestrant, or neratinib + fulvestrant + trastuzumab (N + F + T). We report results from 71 patients with HR+, HER2-mutant MBC, including 21 (seven in each arm) from a randomized substudy of fulvestrant versus fulvestrant + trastuzumab (F + T) versus N + F + T., Patients and Methods: Patients with HR+ HER2-negative MBC with activating HER2 mutation(s) and prior cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) therapy received N + F + T (oral neratinib 240 mg/day with loperamide prophylaxis, intramuscular fulvestrant 500 mg on days 1, 15, and 29 of cycle 1 then q4w, intravenous trastuzumab 8 mg/kg then 6 mg/kg q3w) or F + T or fulvestrant alone. Those whose disease progressed on F + T or fulvestrant could cross-over to N + F + T. Efficacy endpoints included investigator-assessed objective response rate (ORR), clinical benefit rate (RECIST v1.1), duration of response, and progression-free survival (PFS). Plasma and/or formalin-fixed paraffin-embedded tissue samples were collected at baseline; plasma was collected during and at end of treatment. Extracted DNA was analyzed by next-generation sequencing., Results: ORR for 57 N + F + T-treated patients was 39% [95% confidence interval (CI) 26% to 52%); median PFS was 8.3 months (95% CI 6.0-15.1 months). No responses occurred in fulvestrant- or F + T-treated patients; responses in patients crossing over to N + F + T supported the requirement for neratinib in the triplet. Responses were observed in patients with ductal and lobular histology, 1 or ≥1 HER2 mutations, and co-occurring HER3 mutations. Longitudinal circulating tumor DNA sequencing revealed acquisition of additional HER2 alterations, and mutations in genes including PIK3CA, enabling further precision targeting and possible re-response., Conclusions: The benefit of N + F + T for HR+ HER2-mutant MBC after progression on CDK4/6is is clinically meaningful and, based on this study, N + F + T has been included in the National Comprehensive Cancer Network treatment guidelines. SUMMIT has improved our understanding of the translational implications of targeting HER2 mutations with neratinib-based therapy., Competing Interests: Disclosure KJ: Consultant/Advisory Board: Novartis, AstraZeneca, Pfizer, BMS, Jounce Therapeutics, Taiho Oncology, Genentech/Roche, Lilly Pharmaceuticals/Loxo Oncology, AbbVie, Eisai, Blueprint Medicines, Seattle Genetics, Daiichi Sankyo, Gilead, Olema Pharmaceuticals, Sun Pharma Advanced Research Company Ltd., Menarini/Stemline, and Scorpion Therapeutics. Research Funding: Novartis, Genentech/Roche, AstraZeneca, Debio Pharmaceuticals, Pfizer, Lilly Pharmaceuticals/Loxo Oncology, Zymeworks, Gilead, Puma Biotechnology, Merck Pharmaceuticals, and Scorpion Therapeutics. HW: His institution received financial compensation on his behalf for advisory boards, lecture fees and/or consultancy fees from Daiichi Sankyo, Gilead, Lilly, Augustine Therapeutics, AstraZeneca, Immutep Pty, MSD, and Roche. He received travel support from Gilead, Daiichi Sankyo, and Pfizer. SAH: Contracted research paid to institution +/- editorial support for authorship: Ambrx, Arvinas, AstraZeneca, Bayer, Celcuity, Cytomx, Daiichi Sankyo, Dantari, Dignitana, Genentech/Roche, G1-Therapeutics, Gilead, Greenwich Life Sciences Inc., GSK, Immunomedics, Eli Lilly, Loxo, Macrogenics, Novartis, OBI Pharma, Orinove, Orum, Pfizer, Phoenix Molecular Designs, Ltd., Pieris, PUMA, Radius, Sanofi, Seattle Genetics/Seagen, and Zymeworks. Speaking: Daiichi Sankyo (2021). AG-Z: Advisory/Consultancy: AstraZeneca, Novartis, MSD, Pierre-Fabre, and Exact Science. Speaker Bureau/Expert testimony: Roche, AstraZeneca, Novartis, MSD, Pfizer, Lilly, and Pierre-Fabre. Research grant/Funding (institution): Pfizer. Travel/Accommodation/Expenses: Roche, Novartis, and Pfizer. NU: Consultant/Advisory Board: Novartis, Eli Lilly, BioTheranostics, Gilead. AB: Consultant: AstraZeneca, Pfizer, Novartis, Lilly, Genentech/Roche, Seagen, Daiichi Sankyo, Merck, Agendia, Sanofi, Puma, Myriad, and Gilead; research support: Agendia and AstraZeneca. HP: Research grants to institution: Adlai Nortye USA, Ambrx, Aprea Therapeutics AB, Array BioPharma, AstraZeneca, BJ Bioscience, Bristol-Myers Squibb, Daiichi Pharmaceutical, Elicio Therapeutics, Exelixis, Fate Therapeutics, Genentech, GlaxoSmithKline, Gossamer Bio, Hutchison MediPharma, ImmuneOncia Therapeutics, ImmunoGen, Mabspace Biosciences, MacroGenics, Merck, Mirati Therapeutics, Novartis Pharmaceuticals, Oncologie, PsiOxus Therapeutics, RePare Therapeutics, Seattle Genetics, Synermore Biologics, TopAlliance Biosciences, Turning Point Therapeutics, Vedanta Biosciences, Xencor. ESY: Research funding: Puma Biotechnology; Advisory boards: Bayer, Clovis; Consultant: AstraZeneca. IS: Institutional Research Funding: Roche/Genentech, Puma Biotechnology, MSD, Merck, AstraZeneca, Incyte, Orion, Genmab, Bristol-Myers Squibb, Bayer/Loxo Oncology, Lilly Pharmaceuticals/Loxo Oncology, Novartis, Pfizer, Amgen, Repare Therapeutics. Honoraria: AstraZeneca. Support for travel and meeting attendance: Roche, Novartis, Merck/Pfizer, Incyte, and AstraZeneca. SR: Consultant/Advisory board: Novartis, AstraZeneca, Gilead, and Daiichi Sankyo. MBh: Consulting/Advisory Board: Daiichi Sankyo, Merck, Pfizer, and AstraZeneca. SV: Funding from Pfizer directly to institution for conducting clinical trial. MA: Consulting or advisory role: Pfizer, AstraZeneca, Menarini, Novartis, Daiichi Sankyo, and Gilead. Speaker’s bureau: Pfizer, Novartis, and Gilead. Research funding: AstraZeneca. F-CB: Consulting or advisory role: Pfizer; AstraZeneca; Lilly; Novartis; Menarini; Sanofi; GSK; Rain Oncology; Caris Life Sciences; GE Healthcare; Exact Sciences; Gilead. Speaker's bureau: Pfizer; Novartis; AstraZeneca; Roche; Lilly; Rain Oncology; Daiichi Sankyo; and Menarini-Stemline. Research Funding: Novartis; Pfizer; Menarini Silicon Biosystems; Prolynx; Merck KGaA; and GE Healthcare. SL: Receives research funding to her institution from Novartis, Bristol Myers Squibb, Merck, Puma Biotechnology, Eli Lilly, Nektar Therapeutics, AstraZeneca, and Seattle Genetics. She has acted as consultant (not compensated) to Seattle Genetics, Novartis, Bristol Myers Squibb, Merck, AstraZeneca, Eli Lilly, Pfizer, Gilead Therapeutics, and Roche-Genentech. She has acted as consultant (paid to her institution) to Aduro Biotech, Novartis, GlaxoSmithKline, Roche-Genentech, AstraZeneca, Silverback Therapeutics, G1 Therapeutics, Puma Biotechnology, Inc., Pfizer, Gilead Therapeutics, Seattle Genetics, Daiichi Sankyo, Merck, Amunix, Tallac Therapeutics, Eli Lilly, and Bristol Myers Squibb. JC: Has taken part in advisory boards for Puma Biotechnology. His unit is the recipient of a peer-reviewed grant from Science Foundation Ireland (an Irish government organization) which is part-funded by Puma Biotechnology Inc. Neither he nor his spouse own Puma Biotechnology stock. MEB: Medical advisory board of Strata Oncology; Research funding from AbbVie, Arcus, Apollomics, Elevation Oncology, Endeavor, Genetech, Puma, Loxo Oncology, and Seagen. SAP-P: Clinical trial research support/grant funding through the institution from AbbVie, Inc., ABM Therapeutics, Inc., Acepodia, Inc., Alkermes, Aminex Therapeutics, Amphivena Therapeutics, Inc., BioMarin Pharmaceutical, Inc., Boehringer Ingelheim, Bristol-Myers Squibb, Cerulean Pharma, Inc., Chugai Pharmaceutical Co., Ltd., Curis, Inc., Cyclacel Pharmaceuticals, Daiichi Sankyo, Eli Lilly, ENB Therapeutics, Epigenetix Inc., Five Prime Therapeutics, F-Star Beta Limited, F-Star Therapeutics, Gene Quantum, Genmab A/S, Gilead Sciences, Inc., GlaxoSmithKline, Helix BioPharma Corp., Hengrui Pharmaceuticals, Co., Ltd., HiberCell, Inc., Immorna Biotherapeutics, Inc., Immunomedics, Inc., Incyte Corp., Jacobio Pharmaceuticals Co., Ltd., Jiangsu Simcere Pharmaceutical Co., Ltd., Lytix Biopharma AS, Medimmune, LLC., Medivation, Inc., Merck Sharp and Dohme Corp., Nectin Therapeutics, Ltd., Novartis Pharmaceuticals, Pieris Pharmaceuticals, Inc., Pfizer, Phanes Therapeutics, Principia Biopharma, Inc., Puma Biotechnology, Inc., Purinomia Biotech, Inc., Rapt Therapeutics, Inc., Replimune, Seattle Genetics, Silverback Therapeutics, Synlogic Therapeutics, Taiho Oncology, Tesaro, Inc., TransThera Bio, ZielBio, Inc., NCI/NIH, P30CA016672—Core Grant (CCSG Shared Resources); and consulting fees from CRC Oncology. JMS: Research grant support: AstraZeneca and Strata Oncology. SC: Advisory/Consultancy: Novartis, F. Hoffmann-La Roche, Pfizer, Eli Lilly, AstraZeneca, Amgen, Gilead, Merck, and Exact Sciences. Research funding to the institution: Novartis, F. Hoffmann-La Roche, Pfizer, Genomic Health/Exact Sciences, AstraZeneca, Genentech, Celgene, Amgen, BMS, Merck, Sanofi, Puma, and Gilead. CS: Consulting, advisory role or travel grants from: AstraZeneca, AX'Consulting, Byondis B.V, Daiichi Sankyo, Eisai, Exact Sciences, Exeter Pharma, F. Hoffmann-La Roche Ltd., Lilly, MediTech, Merck Sharp & Dohme, Novartis, Pfizer, Philips, Pierre-Fabre, PintPharma, Puma Biotechnology, Seagen, and Zymeworks. JAG-S: Consulting or advisory role: Novartis Pharmaceuticals Corporation, AstraZeneca, Lilly, Seagen, Daiichi Sankyo, Gilead Sciences, Exact Sciences, and Stemline Menarini; speakers bureau: Novartis Pharmaceuticals Corporation, Lilly, AstraZeneca, and Stemline Menarini; travel, accommodations, expenses: Daiichi Sankyo, Gilead Sciences, and Exact Sciences. VG: Advisory Board: Boehringer. Research Funding: Bayer, Boehringer, Roche. Institutional Funding: Genentech, Merck Serono, Roche, Beigene, Bayer, Servier, Lilly, Novartis, Takeda, Astellas, Fibrogen, Amcure, Natera, Sierra Oncology, AstraZeneca, Medimmune, BMS, and MSD. ENG-Y: Honoraria from Novartis, Pfizer, Eli Lilly, MSD, and AstraZeneca. CM: Has received research grants from Puma Biotechnology and Pfizer. She received consulting fees from AstraZeneca, Olaris, Novartis, and Sanofi. ABH: Reports stock and other ownership interests in Pfizer (immediate family member) and research funding from Takeda. JWG: has received institutional research funding from Puma Biotechnology Inc. RB: Receives research grant from Puma; has performed consulting for Genentech. JSKB: Employee (and stockholder) of Tempus Labs. LDE, DD, AF, and AW: Employees and stockholders of Puma Biotechnology. CLA: Has received research grants from Pfizer, Lilly, and Takeda. He serves or has served as scientific advisor to Novartis, Lilly, AstraZeneca, Daiichi Sankyo, Merck, Immunomedics, OrigiMed, Sanofi, TAIHO Oncology, and Puma Biotechnology. DBS: Has served as a consultant for/received honorarium from Pfizer, Vividion Therapeutics, Scorpion Therapeutics, FORE Therapeutics, Fog Pharma, Elsie Biotechnologies, Fog Pharma, Rain Oncology, Function Oncology, and BridgeBio. All other authors have declared no conflicts of interest. Data Sharing The authors declare that the data supporting the findings of this study are available within the article. Qualified researchers and study participants may submit requests for other study documentation and clinical trial data to clinicaltrials@pumabiotechnology.com for consideration., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
32. Discordance of HER2 Expression and/or Amplification on Repeat Testing.
- Author
-
DiPeri TP, Kong K, Varadarajan K, Karp DD, Ajani JA, Pant S, Press MF, Piha-Paul SA, Dumbrava EE, and Meric-Bernstam F
- Subjects
- Humans, In Situ Hybridization, Fluorescence, Immunohistochemistry, Biomarkers, Tumor genetics, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Stomach Neoplasms pathology
- Abstract
We sought to assess discordance of HER2 status in patients with HER2-amplified/expressing solid tumors who underwent reevaluation of HER2 status. Patients with metastatic solid tumors and HER2 expression by IHC or amplification by FISH/next-generation sequencing on local testing underwent central HER2 IHC/FISH testing with either archival or fresh biopsies and were evaluated for discordance in HER2 status. 70 patients (12 cancer types) underwent central HER2 reevaluation, including 57 (81.4%) with a new biopsy. In 30 patients with HER2 3+ on local IHC, 21 (70.0%) were 3+, 5 (16.7%) were 2+, 2 (6.7%) were 1+, and 2 (6.7%) had 0 HER2 expression on central IHC. In 15 patients whose cancers were 2+ on local IHC, 2 (13.3%) were 3+, 5 (33.3%) were 2+, 7 (46.7%) were 1+, and 1 (6.7%) had 0 HER2 expression on central IHC. HER2 discordance was seen in 16 of 52 (30.8%) of patients with HER2 overexpression/amplification who underwent a new image-guided biopsy. Discordance was observed in 10 (33.3%) of 30 patients who received intervening HER2-targeted therapy and in 6 (23.8%) of 22 patients who did not. In the 8 patients who had central HER2 assessment from the same archival block used for local testing, none were discordant. Discordance of HER2 status is common in patients with tumors previously identified as HER2-expressing, especially in patients with HER2 2+ tumors. Repeat biomarker evaluation may have value when considering HER2-targeted therapies., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
33. A phase I trial of the pan-ERBB inhibitor neratinib combined with the MEK inhibitor trametinib in patients with advanced cancer with EGFR mutation/amplification, HER2 mutation/amplification, HER3/4 mutation or KRAS mutation.
- Author
-
Piha-Paul SA, Tseng C, Tran HT, Gao M, Karp DD, Subbiah V, Tsimberidou AM, Kawedia JD, Fu S, Pant S, Yap TA, Morris VK, Kee BK, Blum Murphy M, Lim J, and Meric-Bernstam F
- Subjects
- Humans, Middle Aged, Antineoplastic Combined Chemotherapy Protocols, Protein Kinase Inhibitors adverse effects, Genes, erbB, Mutation, ErbB Receptors genetics, Nausea drug therapy, Diarrhea drug therapy, Mitogen-Activated Protein Kinase Kinases, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Proto-Oncogene Proteins p21(ras) genetics, Neoplasms drug therapy
- Abstract
Purpose: Aberrant alterations of ERBB receptor tyrosine kinases lead to tumorigenesis. Single agent therapy targeting EGFR or HER2 has shown clinical successes, but drug resistance often develops due to aberrant or compensatory mechanisms. Herein, we sought to determine the feasibility and safety of neratinib and trametinib in patients with EGFR mutation/amplification, HER2 mutation/amplification, HER3/4 mutation and KRAS mutation., Methods: Patients with actionable somatic mutations or amplifications in ERBB genes or actionable KRAS mutations were enrolled to receive neratinib and trametinib in this phase I dose escalation trial. The primary endpoint was determination of the maximum tolerated dose (MTD) and dose-limiting toxicity (DLT). Secondary endpoints included pharmacokinetic analysis and preliminary anti-tumor efficacy., Results: Twenty patients were enrolled with a median age of 50.5 years and a median of 3 lines of prior therapy. Grade 3 treatment-related toxicities included: diarrhea (25%), vomiting (10%), nausea (5%), fatigue (5%) and malaise (5%). The MTD was dose level (DL) minus 1 (neratinib 160 mg daily with trametinib 1 mg, 5 days on and 2 days off) given 2 DLTs of grade 3 diarrhea in DL1 (neratinib 160 mg daily with trametinib 1 mg daily). The treatment-related toxicities of DL1 included: diarrhea (100%), nausea (55.6%) and rash (55.6%). Pharmacokinetic data showed trametinib clearance was significantly reduced leading to high drug exposures of trametinib. Two patients achieved stable disease (SD) ≥ 4 months., Conclusion: Neratinib and trametinib combination was toxic and had limited clinical efficacy. This may be due to suboptimal drug dosing given drug-drug interactions., Trial Registration Id: NCT03065387., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
34. Phase I Study of SYNB1891, an Engineered E. coli Nissle Strain Expressing STING Agonist, with and without Atezolizumab in Advanced Malignancies.
- Author
-
Luke JJ, Piha-Paul SA, Medina T, Verschraegen CF, Varterasian M, Brennan AM, Riese RJ, Sokolovska A, Strauss J, Hava DL, and Janku F
- Subjects
- Humans, Antibodies, Monoclonal, Humanized, Immunologic Factors therapeutic use, Cytokines therapeutic use, Escherichia coli genetics, Neoplasms drug therapy, Neoplasms genetics
- Abstract
Purpose: SYNB1891 is a live, modified strain of the probiotic Escherichia coli Nissle 1917 (EcN) engineered to produce cyclic dinucleotides under hypoxia, leading to STimulator of INterferon Genes (STING) activation in phagocytic antigen-presenting cells in tumors and activating complementary innate immune pathways., Patients and Methods: This first-in-human study (NCT04167137) enrolled participants with refractory advanced cancers to receive repeat intratumoral injections of SYNB1891 either alone or in combination with atezolizumab, with the primary objective of evaluating the safety and tolerability of both regimens., Results: Twenty-four participants received monotherapy across six cohorts, and 8 participants received combination therapy in two cohorts. Five cytokine release syndrome events occurred with monotherapy, including one that met the criteria for dose-limiting toxicity at the highest dose; no other SYNB1891-related serious adverse events occurred, and no SYNB1891-related infections were observed. SYNB1891 was not detected in the blood at 6 or 24 hours after the first intratumoral dose or in tumor tissue 7 days following the first dose. Treatment with SYNB1891 resulted in activation of the STING pathway and target engagement as assessed by upregulation of IFN-stimulated genes, chemokines/cytokines, and T-cell response genes in core biopsies obtained predose and 7 days following the third weekly dose. In addition, a dose-related increase in serum cytokines was observed, as well as stable disease in 4 participants refractory to prior PD-1/L1 antibodies., Conclusions: Repeat intratumoral injection of SYNB1891 as monotherapy and in combination with atezolizumab was safe and well tolerated, and evidence of STING pathway target engagement was observed., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
35. Multidisciplinary Care of a Large Brain Metastasis in a Patient with Hormone-Receptor-Positive Breast Cancer with Ataxia-Telangiectasia Mutation.
- Author
-
Chelariu-Raicu A, Piha-Paul SA, Chavez-MacGregor M, Johnson J, Sawaya R, McAleer MF, Nguyen A, Hartnett A, Tsimberidou AM, Meric-Bernstam F, and Dumbrava EE
- Abstract
Poly (adenosine diphosphate-ribose) polymerase inhibitors (PARP)i are emerging as standard oncology treatments in various tumor types. The indications will expand as PARPi are being investigated in various breast cancer subtypes. Currently, except for BRCA1/2 mutation carriers with human epidermal growth factor receptor 2 (HER2)-negative breast cancer, there is inadequate identification of predictive biomarkers of response. We present a 57-year-old woman with metastatic breast cancer, hormone-receptor-positive, HER2 negative with a germline ataxia-telangiectasia mutation with a large brain metastasis with clinical benefit to talazoparib. This case report exemplifies the importance of the multidisciplinary management of patients with brain metastases and personalized biomarker selected treatment., Competing Interests: Conflict of Interest: None.
- Published
- 2023
- Full Text
- View/download PDF
36. Impact of Biomarker-Matched Therapies on Outcomes in Patients with Sarcoma Enrolled in Early-Phase Clinical Trials (SAMBA 101).
- Author
-
Carmagnani Pestana R, Moyers JT, Roszik J, Sen S, Hong DS, Naing A, Herzog CE, Fu S, Piha-Paul SA, Rodon J, Yap TA, Karp DD, Tsimberidou AM, Pant S, Zarzour MA, Ratan R, Ravi V, Benjamin RS, Lazar AJ, Wang WL, Daw N, Gill JB, Harrison DJ, Lewis VO, Roland CL, Patel SR, Livingston JA, Somaiah N, Ludwig JA, Conley AP, Hamerschlak N, Gorlick R, Meric-Bernstam F, and Subbiah V
- Subjects
- Humans, Retrospective Studies, Biomarkers, Gastrointestinal Stromal Tumors drug therapy, Gastrointestinal Stromal Tumors genetics, Sarcoma diagnosis, Sarcoma drug therapy, Soft Tissue Neoplasms pathology
- Abstract
Purpose: Developing new therapeutics for any of the more than 100 sarcoma subtypes presents a challenge. After progression from standard therapies, patients with sarcoma may be referred for enrollment in early-phase trials. This study aimed to investigate whether enrollment in biomarker-matched early-phase clinical trials leads to better outcomes for patients with advanced sarcoma., Experimental Design: In this retrospective analysis, investigational treatment characteristics and longitudinal survival outcomes were analyzed in patients with biopsy-confirmed sarcoma enrolled in early-phase trials at MD Anderson Cancer Center from May 2006 to July 2021., Results: Five hundred eighty-seven patients were included [405 soft tissue, 122 bone, 60 gastrointestinal stromal tumor (GIST); median of three prior lines of therapy]. Most common subtypes were leiomyosarcoma (17.2%), liposarcoma (14.0%), and GIST (10.2%). Molecular testing was available for 511 patients (87.1%); 221 patients (37.6%) were treated in matched trials. Overall response rate was 13.1% matched compared with 4.9% in unmatched (P < 0.001); the clinical benefit rate at 6 months was 43.9% vs. 19.9% (P < 0.001). Progression-free survival was longer for patients in matched trials (median, 5.5 vs. 2.4 months; P < 0.001), and overall survival was also superior for patients in matched trials (median, 21.5 vs. 12.3 months; P < 0.001). The benefit of enrollment in matched trials was maintained when patients with GIST were excluded from the analysis., Conclusions: Enrollment in biomarker-matched early-phase trials is associated with improved outcomes in heavily pretreated patients with metastatic sarcoma. Molecular testing of tumors from patients with advanced sarcoma and enrollment in matched trials is a reasonable therapeutic strategy., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
37. Multicenter Phase II Trial of the WEE1 Inhibitor Adavosertib in Refractory Solid Tumors Harboring CCNE1 Amplification.
- Author
-
Fu S, Yao S, Yuan Y, Previs RA, Elias AD, Carvajal RD, George TJ, Yuan Y, Yu L, Westin SN, Xing Y, Dumbrava EE, Karp DD, Piha-Paul SA, Tsimberidou AM, Ahnert JR, Takebe N, Lu K, Keyomarsi K, and Meric-Bernstam F
- Subjects
- Female, Humans, Carcinoma, Ovarian Epithelial drug therapy, Bayes Theorem, Oncogene Proteins genetics, Cyclin E, Protein-Tyrosine Kinases genetics, Cell Cycle Proteins genetics, Antineoplastic Agents therapeutic use, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics
- Abstract
Purpose: Preclinical cancer models harboring CCNE1 amplification were more sensitive to adavosertib treatment, a WEE1 kinase inhibitor, than models without amplification. Thus, we conducted this phase II study to assess the antitumor activity of adavosertib in patients with CCNE1 -amplified, advanced refractory solid tumors., Patients and Methods: Patients aged ≥ 18 years with measurable disease and refractory solid tumors harboring CCNE1 amplification, an Eastern Cooperative Oncology Group performance status of 0-1, and adequate organ function were studied. Patients received 300 mg of adavosertib once daily on days 1 through 5 and 8 through 12 of a 21-day cycle. The trial followed Bayesian optimal phase II design. The primary end point was objective response rate (ORR)., Results: Thirty patients were enrolled. The median follow-up duration was 9.9 months. Eight patients had partial responses (PRs), and three had stable disease (SD) ≥ 6 months, with an ORR of 27% (95% CI, 12 to 46), a SD ≥ 6 months/PR rate of 37% (95% CI, 20 to 56), a median progression-free survival duration of 4.1 months (95% CI, 1.8 to 6.4), and a median overall survival duration of 9.9 months (95% CI, 4.8 to 15). Fourteen patients with epithelial ovarian cancer showed an ORR of 36% (95% CI, 13 to 65) and SD ≥ 6 months/PR of 57% (95% CI, 29 to 82), a median progression-free survival duration of 6.3 months (95% CI, 2.4 to 10.2), and a median overall survival duration of 14.9 months (95% CI, 8.9 to 20.9). Common treatment-related toxicities were GI, hematologic toxicities, and fatigue., Conclusion: Adavosertib monotherapy demonstrates a manageable toxicity profile and promising clinical activity in refractory solid tumors harboring CCNE1 amplification, especially in epithelial ovarian cancer. Further study of adavosertib, alone or in combination with other therapeutic agents, in CCNE1 -amplified epithelial ovarian cancer is warranted.
- Published
- 2023
- Full Text
- View/download PDF
38. Antitumour activity of neratinib in patients with HER2-mutant advanced biliary tract cancers.
- Author
-
Harding JJ, Piha-Paul SA, Shah RH, Murphy JJ, Cleary JM, Shapiro GI, Quinn DI, Braña I, Moreno V, Borad M, Loi S, Spanggaard I, Park H, Ford JM, Arnedos M, Stemmer SM, de la Fouchardiere C, Fountzilas C, Zhang J, DiPrimeo D, Savin C, Duygu Selcuklu S, Berger MF, Eli LD, Meric-Bernstam F, Jhaveri K, Solit DB, and Abou-Alfa GK
- Subjects
- Humans, Female, Receptor, ErbB-2 genetics, Diarrhea chemically induced, Antineoplastic Combined Chemotherapy Protocols adverse effects, Treatment Outcome, Quinolines pharmacology, Quinolines therapeutic use, Biliary Tract Neoplasms drug therapy, Biliary Tract Neoplasms genetics, Biliary Tract Neoplasms chemically induced, Breast Neoplasms etiology
- 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 (ClinicalTrials.gov: NCT01953926). The primary objective of the BTC cohort, which is now complete, is first objective response rate (ORR) to neratinib 240 mg orally daily. Secondary objectives include confirmed ORR, clinical benefit rate, progression-free survival, duration of response, overall survival, safety and tolerability. Genomic analyses were exploratory. Among 25 treatment-refractory patients (11 cholangiocarcinoma, 10 gallbladder, 4 ampullary cancers), the ORR is 16% (95% CI 4.5-36.1%). The most common HER2 mutations are S310F (n = 11; 48%) and V777L (n = 4; 17%). Outcomes appear 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 are identified at progression in one responder. Diarrhoea is the most common adverse event, with any-grade diarrhoea in 14 patients (56%). Although neratinib demonstrates antitumour activity in patients with refractory BTC harbouring HER2 mutations, the primary endpoint was not met and combinations may be explored., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
39. Predictors of Oncologic Outcome in Patients Receiving Phase I Investigational Therapy for Recurrent or Metastatic Cervical Cancer.
- Author
-
Son J, Lin HY, Fu S, Biter AB, Dumbrava EE, Karp DD, Naing A, Pant S, Piha-Paul SA, Rodon J, Subbiah V, Tsimberidou AM, Yap TA, Frumovitz MM, Jazaeri AA, Ramirez PT, Westin SN, Yuan Y, Meric-Bernstam F, and Hong DS
- Abstract
Introduction: We aimed to identify clinical, pathologic, and treatment factors that are predictive of response and survival in patients with cervical cancer referred to phase I clinical trials., Methods: Patients with cervical cancer who received at least one dose of a phase I investigational agent at our institution between 2014 and 2022 were included. The log-rank test was used to analyze differences in progression-free survival (PFS) and overall survival (OS), and multivariable regression analysis was performed., Results: We included 65 patients with a median age of 41 years (range, 20-74), 3 prior therapies (range, 1-7), and 67.7% squamous carcinoma. The rate of distant metastasis at trial entry was 84.6%. The most common molecular alterations included PIK3CA (46.5%), PD-L1+ (46.2%), EPH (30.0%), and CREBBP (23.1%); 23.1% had received a prior checkpoint inhibitor. Phase I trials were for immunotherapy (58.5%) or targeted therapy (41.5%). The rate of biomarker matching was 21.5%. For all patients, median PFS was 3.6 months (95% CI, 2.0-5.2) and OS was 9.3 months (95% CI, 7.0-10.6). Factors at study entry associated with worse survival were presence of bone metastasis (PFS 1.6 vs 4.4 months: hazard ratio [HR], 2.8; p = 0.001; OS 3.8 vs 10.0 months: HR, 3.9; p < 0.0001) and absolute lymphocyte count below 1000/μL (PFS 1.8 vs 5.2 months: HR, 2.9; p = 0.0004; OS 7.0 vs 10.6 months: HR, 3.2; p = 0.0009). Factors associated only with worse OS were absolute neutrophil count above 4700/μL, hemoglobin below 10.5 g/dL, and smoking status. Grade 3+ treatment-related adverse events were seen in 16.9% of cases., Conclusion: Bone metastasis and absolute lymphocyte count below normal range at phase I study entry portend poor survival in patients with recurrent or metastatic cervical cancer., Competing Interests: Conflicts of Interest: The authors declare no conflict of interest directly relating to this study. Unelated conflicts of interest are listed in Supplemental Material (available online)., (© 2023.)
- Published
- 2023
- Full Text
- View/download PDF
40. Complex I inhibitor of oxidative phosphorylation in advanced solid tumors and acute myeloid leukemia: phase I trials.
- Author
-
Yap TA, Daver N, Mahendra M, Zhang J, Kamiya-Matsuoka C, Meric-Bernstam F, Kantarjian HM, Ravandi F, Collins ME, Francesco MED, Dumbrava EE, Fu S, Gao S, Gay JP, Gera S, Han J, Hong DS, Jabbour EJ, Ju Z, Karp DD, Lodi A, Molina JR, Baran N, Naing A, Ohanian M, Pant S, Pemmaraju N, Bose P, Piha-Paul SA, Rodon J, Salguero C, Sasaki K, Singh AK, Subbiah V, Tsimberidou AM, Xu QA, Yilmaz M, Zhang Q, Li Y, Bristow CA, Bhattacharjee MB, Tiziani S, Heffernan TP, Vellano CP, Jones P, Heijnen CJ, Kavelaars A, Marszalek JR, and Konopleva M
- Subjects
- Animals, Mice, Histone Deacetylase Inhibitors therapeutic use, Oxidative Phosphorylation, Humans, Antineoplastic Agents adverse effects, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Neoplasms pathology
- Abstract
Although targeting oxidative phosphorylation (OXPHOS) is a rational anticancer strategy, clinical benefit with OXPHOS inhibitors has yet to be achieved. Here we advanced IACS-010759, a highly potent and selective small-molecule complex I inhibitor, into two dose-escalation phase I trials in patients with relapsed/refractory acute myeloid leukemia (NCT02882321, n = 17) and advanced solid tumors (NCT03291938, n = 23). The primary endpoints were safety, tolerability, maximum tolerated dose and recommended phase 2 dose (RP2D) of IACS-010759. The PK, PD, and preliminary antitumor activities of IACS-010759 in patients were also evaluated as secondary endpoints in both clinical trials. IACS-010759 had a narrow therapeutic index with emergent dose-limiting toxicities, including elevated blood lactate and neurotoxicity, which obstructed efforts to maintain target exposure. Consequently no RP2D was established, only modest target inhibition and limited antitumor activity were observed at tolerated doses, and both trials were discontinued. Reverse translational studies in mice demonstrated that IACS-010759 induced behavioral and physiological changes indicative of peripheral neuropathy, which were minimized with the coadministration of a histone deacetylase 6 inhibitor. Additional studies are needed to elucidate the association between OXPHOS inhibition and neurotoxicity, and caution is warranted in the continued development of complex I inhibitors as antitumor agents., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
41. Manufacturing-dependent change in biological activity of the TLR4 agonist GSK1795091 and implications for lipid A analog development.
- Author
-
Steeghs N, Hansen AR, Hanna GJ, Garralda E, Park H, Strauss J, Adam M, Campbell G, Carver J, Easton R, Mays K, Skrdla P, Struemper H, Washburn ML, Matheny C, and Piha-Paul SA
- Subjects
- Humans, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Cytokines, Lipid A therapeutic use, Toll-Like Receptor 4 agonists, Neoplasms drug therapy, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
A phase I trial (NCT03447314; 204686) evaluated the safety and efficacy of GSK1795091, a Toll-like receptor 4 (TLR4) agonist, in combination with immunotherapy (GSK3174998 [anti-OX40 monoclonal antibody], GSK3359609 [anti-ICOS monoclonal antibody], or pembrolizumab) in patients with solid tumors. The primary endpoint was safety; other endpoints included efficacy, pharmacokinetics, and pharmacodynamics (PD). Manufacturing of GSK1795091 formulation was modified during the trial to streamline production and administration, resulting in reduced PD (cytokine) activity. Fifty-four patients received GSK1795091 with a combination partner; 32 received only the modified GSK1795091 formulation, 15 received only the original formulation, and seven switched mid-study from the original to the modified formulation. Despite the modified formulation demonstrating higher systemic GSK1795091 exposure compared with the original formulation, the transient, dose-dependent elevations in cytokine and chemokine concentrations were no longer observed (e.g., IP-10, IL10, IL1-RA). Most patients (51/54; 94%) experienced ≥1 treatment-emergent adverse event (TEAE) during the study. Safety profiles were similar between formulations, but a higher incidence of TEAEs associated with immune responses (chills, fatigue, pyrexia, nausea, and vomiting) were observed with the original formulation. No conclusions can be made regarding GSK1795091 anti-tumor activity due to the limited data collected. Manufacturing changes were hypothesized to have caused the change in biological activity in this study. Structural characterization revealed GSK1795091 aggregate size in the modified formulation to be twice that in the original formulation, suggesting a negative correlation between GSK1795091 aggregate size and PD activity. This may have important clinical implications for future development of structurally similar compounds., (© 2022 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2022
- Full Text
- View/download PDF
42. Clinical activity of checkpoint inhibitors in angiosarcoma: A retrospective cohort study.
- Author
-
Ravi V, Subramaniam A, Zheng J, Amini B, Trinh VA, Joseph J, Mennel RG, Bishop AJ, Sturgis EM, Goepfert RP, Yalamanchili S, Botello G, Stephen B, Piha-Paul SA, Patel AB, Lazar AJ, Conley AP, Benjamin RS, Patel SR, Futreal PA, Somaiah N, and Naing A
- Subjects
- Humans, Immunotherapy, Progression-Free Survival, Prospective Studies, Retrospective Studies, Hemangiosarcoma
- Abstract
Background: Systemic treatments for angiosarcoma remains an area of unmet clinical need. The authors conducted this retrospective study to assess the clinical activity of checkpoint inhibitors in patients with angiosarcoma. The primary objective was to assess the objective response rate, and the secondary objective was to assess the progression-free and overall survival durations and disease control rate., Methods: Patient data were obtained using The University of Texas MD Anderson Cancer Center Tumor Registry database. The final study population was refined to only include patients who had undergone pembrolizumab monotherapy. The objective response rate was evaluated using RECIST/irRECIST version 1.1. Progression-free survival and overall survival were defined as the time from the initiation of immunotherapy to disease progression or recurrence, death, or last follow-up and to death or last follow-up, respectively., Results: The final cohort comprised 25 patients. Most patients had metastatic disease (72%) and had undergone at least two lines of systemic therapy (80%) before starting pembrolizumab. The objective response rate was 18%, whereas the disease control rate was 59%. The median progression-free survival duration was 6.2 months and was not significantly different between the cutaneous (4.7 months) and visceral angiosarcoma (6.2 months) groups (p = .42). The median overall survival duration was 72.6 months. Toxicities were recorded for eight patients, with fatigue, anemia, constipation, and rash being the most common., Conclusions: Pembrolizumab shows durable clinical activity in angiosarcoma. These findings suggest that checkpoint inhibition as monotherapy or combination therapy is likely to have a high probability of success.© 2022 American Cancer Society., Lay Summary: This is the largest retrospective study to assess the clinical activity of checkpoint inhibitor monotherapy in angiosarcomas. The study includes an adequate number of patients with visceral angiosarcoma that enabled to obtain meaningful clinical insights that were previously unavailable. Our findings indicate an improvement in progression-free survival with pembrolizumab that is comparable to other active agents in angiosarcoma. Pembrolizumab monotherapy in angiosarcomas also has a favorable tolerability profile. Our findings emphasize the need for prospective studies to evaluate the activity of pembrolizumab monotherapy and combination therapy., (© 2022 American Cancer Society.)
- Published
- 2022
- Full Text
- View/download PDF
43. Safety and Tolerability of MEDI0562, an OX40 Agonist mAb, in Combination with Durvalumab or Tremelimumab in Adult Patients with Advanced Solid Tumors.
- Author
-
Goldman JW, Piha-Paul SA, Curti B, Pedersen KS, Bauer TM, Groenland SL, Carvajal RD, Chhaya V, Kirby G, McGlinchey K, Hammond SA, Streicher K, Townsley DM, Chae YK, Voortman J, Marabelle A, and Powderly J
- Subjects
- Adult, Antibodies, Monoclonal, Antibodies, Monoclonal, Humanized, Humans, Ki-67 Antigen, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoplasms drug therapy, Neoplasms etiology
- Abstract
Purpose: Combination therapies targeting immunologic checkpoints have shown promise in treating multiple tumor types. We report safety and tolerability of MEDI0562, a humanized IgG1K OX40 mAb, in combination with durvalumab (anti-PD-L1), or tremelimumab (anti-CTLA-4), in adult patients with previously treated advanced solid tumors., Patients and Methods: In this phase I, multicenter, open-label study, patients received escalating doses of MEDI0562 (2.25, 7.5, or 22.5 mg) every 2 weeks in combination with durvalumab (1,500 mg) or tremelimumab (75 or 225 mg) every 4 weeks, intravenously, until unacceptable toxicity or progressive disease. Tumor assessments were performed every 8 weeks. The primary objective was to evaluate safety and tolerability., Results: Among the 27 and 31 patients who received MEDI0562 + durvalumab or MEDI0562 + tremelimumab, 74.1% and 67.7% reported a treatment-related adverse event (AE), and 22.2% and 19.4% experienced a treatment-emergent AE that led to discontinuation, respectively. The MTD of MEDI0562 + durvalumab was 7.5 mg MEDI0562 + 1,500 mg durvalumab; the maximum administered dose of MEDI0562 + tremelimumab was 22.5 mg MEDI0562 + 225 mg tremelimumab. Three patients in the MEDI0562 + durvalumab arm had a partial response. The mean percentage of Ki67+CD4+ and Ki67+CD8+ memory T cells increased by >100% following the first dose of MEDI0562 + durvalumab or tremelimumab in all dose cohorts. A decrease in OX40+FOXP3 regulatory T cells was observed in a subset of patients with available paired biopsies., Conclusions: Following dose escalation, moderate toxicity was observed in both treatment arms, with no clear efficacy signals demonstrated., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
44. Patient-reported symptom burden in patients with rare cancers receiving pembrolizumab in a phase II Clinical Trial.
- Author
-
Mendoza TR, Hong DS, Peterson CB, Stephen B, Dumbrava E, Pant S, Tsimberidou AM, Yap TA, Sheshadri A, Altan M, George G, Castillo L, Rodriguez E, Gong J, Subbiah V, Janku F, Fu S, Piha-Paul SA, Ahnert JR, Karp DD, Cleeland C, Meric-Bernstam F, and Naing A
- Subjects
- Antibodies, Monoclonal, Humanized, Diarrhea, Fatigue chemically induced, Female, Humans, Longitudinal Studies, Male, Middle Aged, Pain, Patient Reported Outcome Measures, Prospective Studies, Severity of Illness Index, Exanthema, Neoplasms diagnosis, Neoplasms drug therapy
- Abstract
Patients with rare solid tumors treated on early phase trials experience toxicities from their tumors and treatments. However, limited data exist to describe the detailed symptom burden suffered by these patients, particularly those with rare solid tumors treated with immunotherapy. We performed a prospective longitudinal study to capture patient-reported symptom burden. Patients completed the validated MD Anderson Symptom Inventory (MDASI)-Immunotherapy with 20 symptoms including 7 immunotherapy-specific items and 6 interference items at baseline and weekly thereafter for up to 9 weeks. Symptoms and interference were rated on 0-10 scales (0 = none or no interference, 10 = worst imaginable or complete interference). Group-based trajectory modelling determined higher and lower symptom groups. A total of 336 MDASI questionnaires were completed by 53 patients (mean age 55.4y, 53% male) with advanced rare cancers receiving pembrolizumab in a Phase II clinical trial. Symptoms reported as most severe over the course of the treatment over 9 weeks were fatigue [mean (M) = 3.8, SD = 2.3], pain (M = 3.7, SD = 2.9), disturbed sleep (M = 2.7, SD = 2.3), drowsiness (M = 2.6, SD = 2.0) and lack of appetite (M = 2.5, SD = 2.1). Pain in the abdomen (M = 2.2, SD = 2.4), rash (M = 1.1, SD = 1.8) and diarrhea (M = 0.9, SD = 1.5) were less severe. Interference with walking was rated the highest (M = 3.4, SD = 2.8) and relations with others was rated the lowest (M = 2.1, SD = 2.6). Using a composite score based on the five most severe symptoms (fatigue, pain, lack of appetite, feeling drowsy and sleep disturbance), 43% were classified into the high symptom burden group. Using a score based on immunotherapy-specific symptoms (e.g., rash, diarrhea) 33% of patients were included in the high symptom group. Symptom burden stayed relatively stable in the high- and low-symptom burden patient groups from baseline through 9 weeks. Some patients with rare malignancies experienced high symptom burden even at baseline. In patients with rare cancers, symptom trajectories stayed relatively stable over nine weeks of treatment with pembrolizumab.Trial registration: ClinicalTrials.gov identifier: NCT02721732., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
45. Efficacy and safety of pembrolizumab for patients with previously treated advanced vulvar squamous cell carcinoma: Results from the phase 2 KEYNOTE-158 study.
- Author
-
Shapira-Frommer R, Mileshkin L, Manzyuk L, Penel N, Burge M, Piha-Paul SA, Girda E, Lopez Martin JA, van Dongen MGJ, Italiano A, Xu L, Jin F, Norwood K, and Ott PA
- Subjects
- Antibodies, Monoclonal, Humanized adverse effects, B7-H1 Antigen metabolism, Female, Humans, Carcinoma, Squamous Cell drug therapy, Vulvar Neoplasms drug therapy
- Abstract
Objective: Treatment options for advanced vulvar cancer are limited. We evaluated pembrolizumab monotherapy in patients with advanced vulvar squamous cell carcinoma (SCC) enrolled in the phase 2 multicohort, open-label KEYNOTE-158 study (NCT02628067)., Methods: Eligible patients had histologically or cytologically documented advanced vulvar SCC with prior treatment failure, measurable disease per RECIST v1.1, ECOG performance status 0-1, and a tumor sample available for biomarker analysis. Pembrolizumab 200 mg was administered intravenously Q3W for up to 35 cycles (approximately 2 years). The primary endpoint was objective response rate (ORR) per RECIST v1.1 by independent central radiologic review in all patients and subgroups based on PD-L1 combined positive score (≥1 [PD-L1-positive] versus <1 [PD-L1-negative])., Results: 101 patients were enrolled. Median time from first dose to data cutoff was 36.0 months. The ORR (95% CI) was 10.9% (5.6%-18.7%) among all patients, 9.5% (4.2%-17.9%) among the 84 patients with PD-L1-positive tumors, and 28.6% (3.7%-71.0%) among the 7 patients with PD-L1-negative tumors. Among patients with a response, median DOR was 20.4 (range, 2.1+ to 28.0) months. Median (95% CI) PFS and OS were 2.1 (2.0-2.1) and 6.2 (4.9-9.4) months, respectively. Treatment-related AEs occurred in 50.5% of patients (grade 3-5, 11.9%) and led to discontinuation of treatment in 5.0% of patients. Two deaths were considered treatment-related (hepatitis, n = 2)., Conclusions: Pembrolizumab monotherapy was associated with durable responses in a subset of patients with vulvar SCC. Responses occurred regardless of tumor PD-L1 status. No new safety signals emerged; overall, pembrolizumab was well tolerated., Competing Interests: Declaration of competing interest Ronnie Shapira-Frommer: study funding to the institution from Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA (MSD), to support study conduct; honoraria for serving as a speaker from MSD, BMS, AstraZeneca, Novartis, and Roche; personal fees for advisory boards from MSD, Clovis Oncology, and VBL Therapeutics. Linda Mileshkin: study funding to the institution from MSD to support study conduct. Ludmila Manzyuk: study funding to the institution from MSD to support study conduct. Nicolas Penel: study funding to the institution from MSD to support study conduct. Sarina A. Piha-Paul: study funding to the institution from MSD to support study conduct; clinical trial research support from AbbVie, Inc., ABM Therapeutics, Inc., Acepodia, Inc., Alkermes, Aminex Therapeutics, Amphivena Therapeutics, Inc., BioMarin Pharmaceutical, Inc., Boehringer Ingelheim, Bristol Myers Squibb, Cerulean Pharma, Inc., Chugai Pharmaceutical Co., Ltd., Curis, Inc., Cyclacel Pharmaceuticals, Daiichi Sankyo, Eli Lilly, ENB Therapeutics, Five Prime Therapeutics, F-Star Beta Limited, F-Star Therapeutics, Ltd., Gene Quantum, Genmab A/S, GlaxoSmithKline, Helix BioPharma Corp., HiberCell, Inc., Immunomedics, Inc., Incyte Corp., Jacobio Pharmaceuticals Co., Ltd., Lytix Biopharma AS, MedImmune, LLC, Medivation, Inc., Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA; Novartis Pharmaceuticals, Pieris Pharmaceuticals, Inc., Pfizer, Principia Biopharma, Inc., Puma Biotechnology, Inc., Rapt Therapeutics, Inc., Seattle Genetics, Silverback Therapeutics, Synlogic, Taiho Oncology, Tesaro, Inc., TransThera Bio, and NCI/NIH (CCSG Shared Resources Grant# P30Ca016672). Matthew Burge: honoraria and study funding to the institution from MSD to support study conduct. Eugenia Girda: study funding to the institution from MSD to support study conduct. Jose A. Lopez Martin: study funding to the institution from MSD to support study conduct; grants, personal fees, and non-financial support from MSD during the conduct of the study; grants, personal fees, and non-financial support from BMS; grants from Merck-Serono; grants and personal fees from Pfizer; personal fees from Bayer|grants and personal fees from Lilly; grants, personal fees, and non-financial support from PharmaMar; grants, personal fees, and non-financial support from Roche; grants and personal fees from Novartis; and personal fees from Pierre-Fabre; current employee of PharmaMar (beginning after this study was conducted). Marloes G.J. van Dongen: study funding to the institution from MSD to support study conduct. Antoine Italiano: study funding to the institution from MSD to support study conduct, grants from MSD, grants from BMS, grants and personal fees from Bayer, personal fees from Springworks, grants from AstraZeneca, grants and personal fees from Roche, grants from Ipsen, grants from PharmaMar. Lei Xu: employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. Owns stock in Merck & Co., Inc., Kenilworth, NJ, USA. Fan Jin: employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. Owns stock in Merck & Co., Inc., Kenilworth, NJ, USA. Kevin Norwood: employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA. Owns stock in Merck & Co., Inc., Kenilworth, NJ, USA. Patrick Ott: study funding to the institution from MSD to support study conduct; grants from BMS, Genentech, Celldex, Cytomx, Pfizer, Neon Therapeutics, Armo Biosciences, AstraZeneca, Xencor, and Oncorus; personal fees from Alexion, Amgen, BMS, Genentech, Celldex, Cytomx, Pfizer, Novartis, Neon Therapeutics (now BioNTechUS), and Array., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
46. Evaluation of pharmacokinetics and safety of talazoparib in patients with advanced cancer and varying degrees of hepatic impairment.
- Author
-
Guo C, Yu Y, Chakrabarti J, Piha-Paul SA, Moroose R, Plotka A, Shi H, Durairaj C, Wang DD, and Wainberg ZA
- Subjects
- Humans, Liver Diseases complications, Liver Diseases drug therapy, Neoplasms drug therapy, Neoplasms pathology, Phthalazines adverse effects, Phthalazines pharmacokinetics
- Abstract
Aim: This phase I study investigated talazoparib pharmacokinetics (PK) and safety in patients with advanced solid tumours and varying degrees of hepatic function., Methods: Patients with advanced solid tumours and normal hepatic function or varying degrees of hepatic impairment (mild, moderate or severe, based on National Cancer Institute Organ Dysfunction Working Group classification) received talazoparib 0.5 mg once daily for 22 calendar days. Plasma and urine samples after single and multiple doses were collected and analysed for talazoparib using validated assays. Plasma PK data from all patients were analysed using the population PK method. Plasma and urine PK parameters in PK-evaluable patients were calculated using noncompartmental analysis (NCA). Safety was monitored in all enrolled patients., Results: Thirty-eight patients were enrolled; 37 had ≥1 PK concentration, among which 17 were evaluable for NCA. Population PK analysis (n = 37) indicated no significant impact of hepatic function on apparent clearance (CL/F) of talazoparib. Baseline creatinine clearance was the only significant covariate on CL/F (α = 0.05). NCA of data (n = 17) showed no clear trend for increase in exposure on day 22 with worsening hepatic function. Talazoparib protein binding was comparable in patients with varying hepatic function. Talazoparib was generally well tolerated, and the safety profile observed in this study was consistent with the known safety profile of the drug., Conclusions: Hepatic impairment (mild, moderate or severe) has no impact on the PK of talazoparib. No dose modification is recommended for patients with advanced solid tumours and various degrees of hepatic impairment, and this labelling language has been approved by the US Food and Drug Administration and the European Medicines Agency., (© 2022 The Authors. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2022
- Full Text
- View/download PDF
47. Longitudinal Monitoring of Circulating Tumor DNA to Predict Treatment Outcomes in Advanced Cancers.
- Author
-
Gouda MA, Huang HJ, Piha-Paul SA, Call SG, Karp DD, Fu S, Naing A, Subbiah V, Pant S, Dustin DJ, Tsimberidou AM, Hong DS, Rodon J, Meric-Bernstam F, and Janku F
- Subjects
- Biomarkers, Tumor genetics, Humans, Liquid Biopsy, Treatment Outcome, Circulating Tumor DNA genetics, Neoplasms genetics
- Abstract
Purpose: The response to cancer therapies is typically assessed with radiologic imaging 6-10 weeks after treatment initiation. Circulating tumor DNA (ctDNA), however, has a short half-life, and dynamic changes in ctDNA quantity may allow for earlier assessment of the therapeutic response., Methods: Patients with advanced solid tumors referred to the Department of Investigational Cancer Therapeutics at The University of Texas MD Anderson Cancer Center were invited to participate in a liquid biopsy protocol for which serial blood samples were collected before, during, and after systemic therapy. We isolated ctDNA from serially collected plasma samples at baseline, mid-treatment, and first restaging. Genomically informed droplet digital polymerase chain reaction (ddPCR) was performed, and ctDNA quantities were reported as aggregate variant allele frequencies for all detected molecular aberrations., Results: We included 204 patients receiving 260 systemic therapies. The ctDNA detection rate was higher in progressors (patients with progressive disease) compared with nonprogressors (patients with stable disease, partial responses, or complete responses) at all time points ( P < .009). Moreover, ctDNA detection was associated with a shorter median time-to-treatment failure ( P ≤ .001). Positive delta and slope values for changes in ctDNA quantity were more frequent in progressors ( P ≤ .03 and P < .001, respectively) and were associated with a shorter median time-to-treatment failure ( P ≤ .014 and P < .001, respectively). Increasing ctDNA quantity was predictive of clinical and/or radiologic progressive disease in 73% of patients (median lead time, 23 days)., Conclusion: Detection of ctDNA and early dynamic changes in its quantity can predict the clinical outcomes of systemic therapies in patients with advanced solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
48. Report of the First International Symposium on NUT Carcinoma.
- Author
-
French CA, Cheng ML, Hanna GJ, DuBois SG, Chau NG, Hann CL, Storck S, Salgia R, Trucco M, Tseng J, Stathis A, Piekarz R, Lauer UM, Massard C, Bennett K, Coker S, Tontsch-Grunt U, Sos ML, Liao S, Wu CJ, Polyak K, Piha-Paul SA, and Shapiro GI
- Subjects
- Cell Cycle Proteins, Humans, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oncogene Proteins, Fusion genetics, Transcription Factors genetics, Carcinoma genetics, Sarcoma, Ewing
- Abstract
NUT carcinoma is a rare, aggressive cancer defined by rearrangements of the NUTM1 gene. No routinely effective treatments of NUT carcinoma exist, despite harboring a targetable oncoprotein, most commonly BRD4-NUT. The vast majority of cases are fatal. Poor awareness of the disease is a major obstacle to progress in the treatment of NUT carcinoma. While the incidence likely exceeds that of Ewing sarcoma, and BRD4-NUT heralded the bromodomain and extra-terminal domain (BET) inhibitor class of selective epigenetic modulators, NUT carcinoma is incorrectly perceived as "impossibly rare," and therefore receives comparatively little private or governmental funding or prioritization by pharma. To raise awareness, propagate scientific knowledge, and initiate a consensus on standard and targeted treatment of NUT carcinoma, we held the First International Symposium on NUT Carcinoma on March 3, 2021. This virtual event had more than eighty attendees from the Americas, Europe, Asia, and Australia. Patients with NUT carcinoma and family members were represented and shared perspectives. Broadly, the four areas discussed by experts in the field included (1) the biology of NUT carcinoma; (2) standard approaches to the treatment of NUT carcinoma; (3) results of clinical trials using BET inhibitors; and (4) future directions, including novel BET bromodomain inhibitors, combinatorial approaches, and immunotherapy. It was concluded that standard chemotherapeutic approaches and first-generation BET bromodomain inhibitors, the latter complicated by a narrow therapeutic window, are only modestly effective in a minority of cases. Nonetheless, emerging second-generation targeted inhibitors, novel rational synergistic combinations, and the incorporation of immuno-oncology approaches hold promise to improve the prognosis of this disease., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
49. Clinical characteristics and outcomes of phase I cancer patients with CCNE1 amplification: MD Anderson experiences.
- Author
-
Yao S, Meric-Bernstam F, Hong D, Janku F, Naing A, Piha-Paul SA, Tsimberidou AM, Karp D, Subbiah V, Yap TA, Ahnert JR, Pant S, Dumbrava EEI, Wathoo C, Campbell E, Yu L, Yamamura Y, and Fu S
- Subjects
- Clinical Trials, Phase I as Topic, Cyclin E genetics, Humans, Middle Aged, Mutation, Oncogene Proteins genetics, Retrospective Studies, Gene Amplification, Neoplasms genetics
- Abstract
Cyclin E is frequently encoded by CCNE1 gene amplification in various malignancies. We reviewed the medical records of patients with solid tumors displaying CCNE1 amplification to determine the effect of this amplification for future therapeutic development. We reviewed the medical records of patients with advanced solid tumors harboring CCNE1 amplification who were seen at the phase I clinic between September 1, 2012, and December 31, 2019. Among 79 patients with solid tumors harboring CCNE1 amplification, 56 (71%) received phase 1 clinical trial therapy, 39 (49%) had 3 or more concurrent genomic aberrances, and 52 (66%) had a concurrent TP53 mutation. The median overall survival (OS) after patients' initial phase I visit was 8.9 months and after their initial metastasis diagnosis was 41.4 months. We identified four factors associated with poor risk: age < 45 years, body mass index ≥ 25 kg/m
2 , presence of the TP53 mutation, and elevated LDH > upper limit of normal. In patients treated with gene aberration-related therapy, anti-angiogenic therapy led to significantly longer OS after their initial phase I trial therapy than those who did not: 26 months versus 7.4 months, respectively (P = 0.04). This study provided preliminary evidence that CCNE1 amplification was associated with frequent TP53 mutation and aggressive clinical outcomes. Survival benefit was observed in patients who received antiangiogenic therapy and gene aberration-related treatment, supporting the future development of a personalized approach to combine gene aberration-related therapy with antiangiogenesis for the treatment of advanced malignancies harboring CCNE1 amplification., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
50. Efficacy of pembrolizumab in patients with advanced cancer of unknown primary (CUP): a phase 2 non-randomized clinical trial.
- Author
-
Raghav KP, Stephen B, Karp DD, Piha-Paul SA, Hong DS, Jain D, Chudy Onwugaje DO, Abonofal A, Willett AF, Overman M, Smaglo B, Huey RW, Meric-Bernstam F, Varadhachary GR, and Naing A
- Subjects
- Adult, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Humans, Progression-Free Survival, B7-H1 Antigen, Neoplasms, Unknown Primary drug therapy
- Abstract
Background: Cancer of unknown primary (CUP) is an aggressive rare malignancy with limited treatment options. Data regarding clinical activity of immune checkpoint inhibitors in CUP is lacking. Therefore, we evaluated the efficacy of pembrolizumab, a programmed cell death-1 inhibitor, in patients with CUP., Methods: The study was designed as a phase 2 basket trial for independent rare tumor cohorts including CUP. Adult patients with CUP who had progressed on previous systemic therapy, performance status 0/1 and measurable disease per Response Evaluation Criteria in Solid Tumors (RECIST V.1.1) were eligible. Patients received pembrolizumab (200 mg) intravenously every 21 days. Twenty-nine patients were enrolled and treated between August 2016 and June 2020. The primary endpoint was non-progression rate (NPR) at 27 weeks (NPR-27) per immune-related RECIST. Key prespecified secondary endpoints were confirmed objective response rate (ORR), safety, duration of response (DoR), progression-free survival (PFS) and overall survival (OS). Pretreatment biopsies were examined for biomarkers of response (programmed cell death ligand-1 (PD-L1) expression and tumor infiltrating lymphocytes (TILs))., Results: Among 25 (of 29 enrolled) eligible and evaluable patients, 14 (56%) had poorly differentiated carcinoma. Patients received a median of two lines of therapy prior to enrollment. Median follow-up was 27.3 months. NPR-27 was observed in seven patients (28.0% (95% CI: 12.1 to 49.4)). ORR was 20.0% (95% CI: 6.8 to 40.7) with five patients achieving immune-related partial response with median DoR of 14.7 months (95% CI: 9.8 to 19.6). Median PFS and OS were 4.1 (95% CI: 3.1 to 5.1) and 11.3 (95% CI: 5.5 to 17.1) months, respectively. Treatment-related adverse events of any and grade ≥3 were seen in 19 (76%) and 4 (16%) patients, respectively. One (4%) patient had grade 3 immune-related acute kidney injury requiring treatment discontinuation. Neither PD-L1 nor TILs were associated with NPR-27. Both positive PD-L1 staining (44.4% vs 6.3%; p=0.040) and intense TIL infiltration (44.4% vs 6.3%; p=0.040) were associated with response., Conclusion: Pembrolizumab showed encouraging efficacy in patients with CUP with acceptable safety profile., Trial Registration Number: NCT02721732., Competing Interests: Competing interests: KPR reports research support from Bayer, AstraZeneca, and Daiichi outside the submitted work; SAP-P reports research support from AbbVie, ABM Therapeutics, Acepodia, Alkermes, Aminex Therapeutics, Amphivena Therapeutics, BioMarin Pharmaceutical, Boehringer Ingelheim, Bristol Myers Squibb, Cerulean Pharma, Chugai Pharmaceutical Co., Curis, Daiichi Sankyo, Eli Lilly, ENB Therapeutics, Five Prime Therapeutics, Gene Quantum, Genmab A/S, GlaxoSmithKline, Helix BioPharma Corp., Incyte Corp., Jacobio Pharmaceuticals Co., Medimmune, LLC., Medivation, Merck Sharp & Dohme Corp., Novartis Pharmaceuticals, Pieris Pharmaceuticals, Pfizer; Principia Biopharma, Puma Biotechnology, Rapt Therapeutics, Seattle Genetics, Silverback Therapeutics, Taiho Oncology, Tesaro, TransThera Bio, NCI/NIH, P30CA016672 – Core Grant (CCSG Shared Resources) outside the submitted work; DSH reports research support from AbbVie, Adaptimmune, Adlai Nortye, Amgen, AstraZeneca, Bayer, Bristol Myers Squibb, Daichi-Sankyo, Eisai, Eli Lilly, EMD Sereno, Erasca, Fate Therapeutics, Genentech, Genmab, GlaxoSmithKline, Ignyta, Infinity, Kite, Kyowa, LOXO, Merck, MedImmune, Millenium, Mirati, miRNA, Molecular TeMpLaTeS, Mologen, NaVier, nci-cep, Novartis, Numab, Pfizer, Seattle Genetics, Takeda, Turning Point, Vernstam, VM Oncology, and other support from Adaptimmune, Amgen, AstraZeneca, Bayer, Genentech, GlaxoSmithKline, Infinity, Numab, Pfizer, Seattle Genetics, Alpha Insights, Acuta, Axiom, Baxter, Boxer Capital, COG, Ecor1, GLG, Group H, Guidepoint, HCW Precision, Janssen, Merrimack, Medscape, Prime Oncology, STCube, Tavistock, Trieza Therapeutics, Molecular Match, Oncoresponse, Presagia, AACR, ASCO, Celgene, Eli Lilly, SITC, and Phillips, outside of the submitted work; MO reports research support from Merck Sharp & Dohme Corp, AbbVie, Agilvax, Takeda Pharmaceuticals (Japan), Acrotech Biopharma, Janssen Research & Development LLC, Pfizer outside the submitted work; FM-B reports research support from Aileron Therapeutics, AstraZeneca, Bayer Healthcare Pharmaceutical, Calithera Biosciences, Curis, CytomX Therapeutics, Daiichi Sankyo Co., Debiopharm International, eFFECTOR Therapeutics, Genentech, Guardant Health, Klus Pharma, Millennium Pharmaceuticals, Novartis, Puma Biotechnology, Taiho Pharmaceutical Co.; consulting fees from Aduro BioTech, Alkermes, AstraZeneca, DebioPharm, eFFECTOR Therapeutics, F. Hoffman-La Roche, Genentech, IBM Watson, Jackson Laboratory, Kolon Life Science, OrigiMed, PACT Pharma, Parexel International, Pfizer, Samsung Bioepis, Seattle Genetics, Tyra Biosciences, Xencor, Zymeworks; has served on advisory committees for Immunomedics, Inflection Biosciences, Mersana Therapeutics, Puma Biotechnology, Seattle Genetics, Silverback Therapeutics, Spectrum Pharmaceuticals, Zentalis; receives honoraria from Chugai Biopharmaceuticals, Mayo Clinic, Rutgers Cancer Institute of New Jersey; and support for travel and accommodation from Beth Israel Deaconess Medical Center outside the submitted work; AN reports research support from NCI, EMD Serono, MedImmune, Healios Onc. Nutrition, Atterocor/Millendo, Amplimmune, ARMO BioSciences, Karyopharm Therapeutics, Incyte, Novartis, Regeneron, Merck, Bristol Myers Squibb, Pfizer, CytomX Therapeutics, Neon Therapeutics, Calithera BioSciences, TopAlliance BioSciences, Eli Lilly, Kymab, PsiOxus, Arcus Biosciences, NeoImmuneTech, ImmuneOncia, and Surface Oncology, non-financial support for travel and accommodation from ARMO BioSciences, has served as an advisory board member for Novartis, CytomX Therapeutics, Genome and Company, STCube Pharmaceuticals, OncoSec KEYNOTE-695, and Kymab, reports research funding for his spouse from Immune Deficiency Foundation, Jeffery Modell Foundation and chao physician-scientist, and Baxalta, and his spouse has served as an advisory board member for Takeda, CSL, Behring, Horizon, and Pharming outside the submitted work. BSt, DDK, DJ, DOCO, AA, AFW, BSm, RWH, GRV declare no competing interests., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.