25 results on '"Alain Mita"'
Search Results
2. 651 A phase 1, multicenter, open-label, dose-escalation, safety, pharmacodynamic, pharmacokinetic study of Q702 with a cohort expansion at the RP2D in patients with advanced solid tumors
- Author
-
Angela Alistar, Alain Mita, Devalingam Mahalingam, Anthony El-Khoueiry, Jeongjun Kim, Hyunji Ahn, Jinho Choi, Seung-Hee Ryu, Jiye Ahn, Bae Jung Choi, Hwankyu Kang, Yeong-In Yang, Seung-Joo Lee, Borami Jeon, Stefan Proniuk, and Kiyean Nam
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. 523 Preliminary clinical experience with XmAb20717, a PD-1 x CTLA-4 bispecific antibody, in patients with advanced solid tumors
- Author
-
Adil Daud, Ezra Cohen, Matthew Reilley, Siwen Hu-Lieskovan, Shubham Pant, Raphael Clynes, Roger Cohen, Anthony Shields, Mark Stein, Alain Mita, Bartosz Chmielowski, Yana Najjar, Elaine Shum, Joaquina Baranda, R Donald Harvey, Catherine Fleener, Ying Ding, Sowmya Chollate, Jolene Shorr, Barbara Hickingbottom, and Lei Bao
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. 525 Preliminary safety, PK/PD and efficacy results from a first-in-human phase I/IIa clinical trial of BNT411, a systemic Toll-like receptor 7 agonist in patients with solid tumors
- Author
-
Hendrik-Tobias Arkenau, Emiliano Calvo, Alain Mita, Young Kwang Chae, Devalingam Mahalingam, Ugur Sahin, Elena Garralda, Oezlem Tuereci, Vladimir Galvao, Stefan Symeonides, Maria Miguel, Hariz Hassan, Annette Baumhauer, Timo Völker, Marie-Cristine Kühnle, Roman Rösemann, and Stefan Strobl
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
5. Avelumab for platinum-ineligible/refractory recurrent and/or metastatic squamous cell carcinoma of the head and neck: phase Ib results from the JAVELIN Solid Tumor trial
- Author
-
Nicolas Penel, Ulrich Keilholz, Ani Balmanoukian, Manish R Patel, Sanjay Goel, Keun-Wook Lee, Alain Mita, Christophe Le Tourneau, Deborah J Wong, Patrick Schöffski, Marcis Bajars, Amaury Daste, Hans Juergen Grote, Dongli Zhou, Michael S Gordon, Martin Gutierrez, Damien Vansteene, and Joël Guigay
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
6. Avelumab (anti–PD-L1) as first-line switch-maintenance or second-line therapy in patients with advanced gastric or gastroesophageal junction cancer: phase 1b results from the JAVELIN Solid Tumor trial
- Author
-
Hyun Cheol Chung, Hendrik-Tobias Arkenau, Jeeyun Lee, Sun Young Rha, Do-Youn Oh, Lucjan Wyrwicz, Yoon-Koo Kang, Keun-Wook Lee, Jeffrey R. Infante, Sung Sook Lee, Margaret Kemeny, Ulrich Keilholz, Bohuslav Melichar, Alain Mita, Ruth Plummer, Denis Smith, Arnold B. Gelb, Huiling Xiong, Janet Hong, Vikram Chand, and Howard Safran
- Subjects
Avelumab ,Metastatic ,Gastric ,Esophagogastric junction ,Adenocarcinoma ,Maintenance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background We evaluated the antitumor activity and safety of avelumab, a human anti–PD-L1 IgG1 antibody, as first-line switch-maintenance (1 L-mn) or second-line (2 L) treatment in patients with advanced gastric/gastroesophageal cancer (GC/GEJC) previously treated with chemotherapy. Methods In a phase 1b expansion cohort, patients without (1 L-mn) or with (2 L) disease progression following first-line chemotherapy for advanced GC/GEJC received avelumab 10 mg/kg intravenously every 2 weeks. Endpoints included best overall response, progression-free survival (PFS), overall survival (OS), and safety. Results Overall, 150 patients were enrolled (1 L-mn, n = 90; 2 L, n = 60) and median follow-up in the 1 L-mn and 2 L subgroups was 36.0 and 33.7 months, respectively. The confirmed objective response rate was 6.7% in both subgroups (95% CI, 2.5–13.9% and 1.8–16.2%, respectively), including complete responses in 2.2% of the 1 L-mn subgroup (n = 2). In the 1 L-mn and 2 L subgroups, median duration of response was 21.4 months (95% CI, 4.0–not estimable) and 3.5 months (95% CI, 2.8–8.3) and disease control rates were 56.7 and 28.3%, respectively. Median PFS in the 1 L-mn and 2 L subgroups was 2.8 months (95% CI, 2.3–4.1) and 1.4 months (95% CI, 1.3–1.5), with 6-month PFS rates of 23.0% (95% CI, 14.7–32.4%) and 7.9% (95% CI, 2.6–17.2%), and median OS was 11.1 months (95% CI, 8.9–13.7) and 6.6 months (95% CI, 5.4–9.4), respectively. In the 1 L-mn subgroup, median OS measured from start of 1 L chemotherapy was 18.7 months (95% CI, 15.4–20.6). Across both subgroups, 20.7% had an infusion-related reaction of any grade. Other common treatment-related adverse events (TRAEs) of any grade included fatigue (10.0%) and nausea (6.7%). Treatment-related serious adverse events occurred in 4.0% of patients. Overall, 8.7% had a grade ≥3 TRAE, including 1 treatment-related death. Conclusion Avelumab showed clinical activity and an acceptable safety profile in patients with GC/GEJC. Trial registration ClinicalTrials.gov NCT01772004; registered 21 January 2013.
- Published
- 2019
- Full Text
- View/download PDF
7. 407 Preliminary safety, pharmacokinetics/pharmacodynamics, and antitumor activity of XmAb20717, a PD-1 x CTLA-4 bispecific antibody, in patients with advanced solid tumors
- Author
-
Adil Daud, Rom Leidner, Ezra Cohen, Matthew Reilley, Siwen Hu-Lieskovan, Raphael Clynes, Roger Cohen, John Thompson, Mark Stein, Alain Mita, Bartosz Chmielowski, Yana Najjar, Elaine Shum, R Donald Harvey, Catherine Fleener, Ying Ding, Sowmya Chollate, Hector Avina, Jolene Shorr, and Barbara Hickingbottom
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
8. A phase 2 study of an oral mTORC1/mTORC2 kinase inhibitor (CC-223) for non-pancreatic neuroendocrine tumors with or without carcinoid symptoms.
- Author
-
Edward Wolin, Alain Mita, Amit Mahipal, Tim Meyer, Johanna Bendell, John Nemunaitis, Pam N Munster, Luis Paz-Ares, Ellen H Filvaroff, Shaoyi Li, Kristen Hege, Hans de Haan, and Monica Mita
- Subjects
Medicine ,Science - Abstract
Second-generation mammalian target of rapamycin (mTOR) inhibitors such as CC-223 may have theoretical advantages over first-generation drugs by inhibiting TOR kinase in mTOR complex 1 (mTORC1) and 2 (mTORC2), potentially improving clinical efficacy for well-differentiated neuroendocrine tumors (NET).Enrolled patients had metastatic, well-differentiated NET of non-pancreatic gastrointestinal or unknown origin, with/without carcinoid symptoms, had failed ≥1 systemic chemotherapy, and were taking a somatostatin analog (SSA). Oral once-daily CC-223 was administered in 28-day cycles starting at 45 mg (n = 24), with a subsequent cohort starting at 30 mg (n = 23). Objectives were to evaluate tolerability, preliminary efficacy, and pharmacokinetic and biomarker profiles of CC-223. Forty-seven patients completed the study, with mean treatment duration of 378 days and mean dose of 26 mg; 26% of patients remained on the starting dose. Most frequent grade ≥3 toxicities were diarrhea (38%), fatigue (21%), and stomatitis (11%). By investigator, 3 of 41 evaluable patients (7%) showed partial response (PR) and 34 (83%) had stable disease (SD) for a disease control rate (DCR) of 90% (95% confidence interval [CI] 76.9-97.3%). Duration of PR was 125-401 days; median SD duration was 297 days (min-max, 50-1519 days). Median progression-free survival was 19.5 months (95% CI 10.4-28.5 months). Carcinoid symptoms of flushing, diarrhea, or both improved in 50%, 41%, and 39% of affected patients, respectively. For the first time, this study describes that a second-generation mTOR pathway inhibitor can result in highly durable tumor regression and control of NET carcinoid symptoms. The manageable safety profile, high DCR, and durable response, coupled with reduction in carcinoid symptoms refractory to SSA, make CC-223 a promising agent for further development.
- Published
- 2019
- Full Text
- View/download PDF
9. Case Report of CCDC149–ALK Fusion: A Novel Genetic Alteration and a Clinically Relevant Target in Metastatic Papillary Thyroid Carcinoma
- Author
-
Hannah Lee, Vimal Krishnan, Lori J. Wirth, Carmelo Nucera, Mariza Venturina, Peter M. Sadow, Alain Mita, and Wendy Sacks
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism - Published
- 2022
10. Figure S1A from Multicenter Phase I Study of Erdafitinib (JNJ-42756493), Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients with Advanced or Refractory Solid Tumors
- Author
-
Josep Tabernero, Jeffrey Infante, Hong Xie, Trilok Parekh, Italo Poggesi, Ademi Santiago-Walker, Bob Zhong, Martha Gonzalez, Alexander Spira, Ramaswamy Govindan, Victor Moreno, Emiliano Calvo, Mark Awad, Nancy Chan, Andres Cervantes, Alain Mita, Cinta Hierro, Antoine Italiano, and Rastislav Bahleda
- Abstract
Supplementary Figure S1. Mean concentration-time erdafitinib profiles after a single dose (cycle 1 day 1 [C1D1]) and at steady-state (cycle 2 day 1 [C2D1]) after daily dosing (Panel A) or intermittent dosing (Panel B). 9 mg QD Part 1 at Cycle 2 Day 1 (C2D1) was not reported as n=1.
- Published
- 2023
11. Data from Multicenter Phase I Study of Erdafitinib (JNJ-42756493), Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients with Advanced or Refractory Solid Tumors
- Author
-
Josep Tabernero, Jeffrey Infante, Hong Xie, Trilok Parekh, Italo Poggesi, Ademi Santiago-Walker, Bob Zhong, Martha Gonzalez, Alexander Spira, Ramaswamy Govindan, Victor Moreno, Emiliano Calvo, Mark Awad, Nancy Chan, Andres Cervantes, Alain Mita, Cinta Hierro, Antoine Italiano, and Rastislav Bahleda
- Abstract
Purpose:Here, we report results of the first phase I study of erdafitinib, a potent, oral pan-FGFR inhibitor.Patients and Methods:Patients age ≥18 years with advanced solid tumors for which standard antineoplastic therapy was no longer effective were enrolled (NCT01703481). Parts 2 to 4 employed molecular screening for activating FGFR genomic alterations. In patients with such alterations, two selected doses/schedules identified during part 1 dose-escalation [9 mg once daily and 10 mg intermittently (7 days on/7 days off), as previously published (Tabernero JCO 2015;33:3401-8)] were tested.Results:The study included 187 patients. The most common treatment-related adverse events were hyperphosphatemia (64%), dry mouth (42%), and asthenia (28%), generally grade 1/2 severity. All cases of hyperphosphatemia were grade 1/2 except for 1 grade 3 event. Skin, nail, and eye changes were observed in 43%, 33%, and 28% of patients, respectively (mostly grade 1/2 and reversible after temporary dosing interruption). Urothelial carcinoma and cholangiocarcinoma were most responsive to erdafitinib, with objective response rates (ORR) of 46.2% (12/26) and 27.3% (3/11), respectively, in response-evaluable patients with FGFR mutations or fusions. All patients with urothelial carcinoma and cholangiocarcinoma who responded to erdafitinib carried FGFR mutations or fusions. Median response duration was 5.6 months for urothelial carcinoma and 11.4 months for cholangiocarcinoma. ORRs in other tumor types were Conclusions:Erdafitinib shows tolerability and preliminary clinical activity in advanced solid tumors with genomic changes in the FGFR pathway, at two different dosing schedules and with particularly encouraging responses in urothelial carcinoma and cholangiocarcinoma.
- Published
- 2023
12. Supplementary Data from Multicenter Phase I Study of Erdafitinib (JNJ-42756493), Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients with Advanced or Refractory Solid Tumors
- Author
-
Josep Tabernero, Jeffrey Infante, Hong Xie, Trilok Parekh, Italo Poggesi, Ademi Santiago-Walker, Bob Zhong, Martha Gonzalez, Alexander Spira, Ramaswamy Govindan, Victor Moreno, Emiliano Calvo, Mark Awad, Nancy Chan, Andres Cervantes, Alain Mita, Cinta Hierro, Antoine Italiano, and Rastislav Bahleda
- Abstract
Supplementary Table S1. Arithmetic Mean {plus minus} SD Plasma Erdafitinib Pharmacokinetic Parameters after the First Erdafitinib Administration (Cycle 1 Day 1) or at Steady-State (Cycle 2 Day 1) in Patients Assigned to the Continuous Daily Dosing or the Intermittent (7 Days on/7 Days off) Dosing Regimen (Part 1 and Part 2 of the Study)
- Published
- 2023
13. Phase 1 study to determine the safety and dosing of autologous PBMCs modified to present HPV16 antigens (SQZ- PBMC-HPV) in HLA-A*02+ patients with HPV16+ Solid Tumors
- Author
-
Antonio Jimeno, Joaquina Baranda, Wade T. Iams, Jong Chul Park, Monica Mita, Michael S. Gordon, Matthew Taylor, Neesha Dhani, Alexis D. Leal, Prakash Neupane, Cathy Eng, Oladapo Yeku, Alain Mita, Justin C. Moser, Marcus Butler, Scott M. Loughhead, Julia Jennings, Nathan R. Miselis, Rui-Ru Ji, Nitya Nair, Martin Kornacker, Ricardo F. Zwirtes, Howard Bernstein, and Armon Sharei
- Subjects
Pharmacology ,Oncology ,Pharmacology (medical) - Abstract
Purpose We conducted a dose escalation Phase 1 study of autologous PBMCs loaded by microfluidic squeezing (Cell Squeeze® technology) with HPV16 E6 and E7 antigens (SQZ-PBMC-HPV), in HLA-A*02 + patients with advanced/metastatic HPV16 + cancers. Preclinical studies in murine models had shown such cells resulted in stimulation and proliferation of antigen specific CD8 + cells, and demonstrated antitumor activity. Methods Administration of SQZ-PBMC-HPV was every 3 weeks. Enrollment followed a modified 3 + 3 design with primary objectives to define safety, tolerability, and the recommended Phase 2 dose. Secondary and exploratory objectives were antitumor activity, manufacturing feasibility, and pharmacodynamic evaluation of immune responses. Results Eighteen patients were enrolled at doses ranging from 0.5 × 106to 5.0 × 106live cells/kg. Manufacture proved feasible and required 6live cells/kg with double priming was chosen as the recommended Phase 2 dose. Multiple participants exhibited pharmacodynamic changes consistent with immune responses supporting the proposed mechanism of action for SQZ-PBMC-HPV, including patients previously refractory to checkpoint inhibitors.
- Published
- 2022
14. 759 Phase 1b study of LNS8801 in combination with pembrolizumab in patients with secondary resistance to immune checkpoint inhibitors
- Author
-
Jordi Rodon, Marya Chaney, Justine Cohen, Tina Garyantes, Jessica Lin, Patricia Lorusso, Alain Mita, Monica Mita, Carolyn Muller, Christopher Natale, Marlana Orloff, Kyriakos Papadopoulos, and Sapna Patel
- Published
- 2022
15. Case Report of
- Author
-
Hannah, Lee, Vimal, Krishnan, Lori J, Wirth, Carmelo, Nucera, Mariza, Venturina, Peter M, Sadow, Alain, Mita, and Wendy, Sacks
- Subjects
Adult ,Iodine Radioisotopes ,Proto-Oncogene Proteins B-raf ,Thyroid Cancer, Papillary ,Mutation ,Humans ,Female ,Anaplastic Lymphoma Kinase ,Thyroid Neoplasms ,Carcinoma, Papillary - Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer, usually with an indolent course.
- Published
- 2022
16. Abstract 2255: Patient pharmacodynamic biomarker and pk evaluation results from an ongoing phase I dose-escalation study of q702, an axl, mer and csf1r kinase inhibitor in patients with advanced solid tumors
- Author
-
Bae Jung Choi, Devalingam Devalingam, Angela Alistar, Anthony El-Khoueiry, Alain Mita, Hwankyu Kang, Jinho Choi, Hyunji Ahn, Jeongjun Kim, Seung-Joo Lee, Yeong-In Yang, Jiye Ahn, Borami Jeon, Jaeseung Kim, and Kiyean Nam
- Subjects
Cancer Research ,Oncology - Abstract
Background: Axl, Mer and CSF1 receptor tyrosine kinases play vital roles in promoting the immunosuppressive tumor microenvironment (TME) by affecting myeloid functions (e.g., tumor associated macrophage [TAM], myeloid derived suppression cell [MDSC]) and promoting epithelial-to-mesenchymal transition (EMT). Thus, simultaneous inhibition of Axl, Mer and CSF1R may be an effective strategy for TME modification. Q702 is a novel Axl/Mer/CSF1R kinase inhibitor that affects the immune components (modulating TAM and MDSC populations, inducing CD8+ T cell infiltration and increasing IFN-ɣ in CD8+ T cell) as well as changes in malignant cells such as increasing MHC I on the tumor cells of syngeneic mouse models. These nonclinical results suggest that Q702 monotherapy or Q702 combination with conventional therapies may have considerable potential as a novel treatment strategy for patients with advanced solid tumors. Methods: This is a Phase 1, Multicenter, Open-label, Dose-Escalation, Safety, Pharmacodynamic, Pharmacokinetic Study of Q702 with a Cohort Expansion at the recommended phase 2 dose (RP2D) in Patients with Advanced Solid Tumors (NCT04648254). Q702 was administered orally for seven days every other week. Peripheral blood samples were obtained on days 1,8,15, and 21. Axl, Mer and CSF1R target engagement is assessed by the quantifications of soluble Axl, Mer and M-CSF in plasma by Luminex xMAP® technology or ELISA. The pharmacodynamic biomarker changes are measured by flow cytometry for immune cell population shifts and IFN-ɣ levels in specific immune cells. Results: PK and PD biomarker samples from 22 patients with various tumor types (e.g. colon, pancreas, esophageal) from the dose escalation phase (4 mg to 240 mg) have been analyzed. Pharmacokinetic studies demonstrated dose proportional increase in Cmax and AUClast of Q702 and its two active metabolites which have activity against Axl and/or CSF1R. Axl and CSF1R target engagement by Q702 treatment is observed in a dose dependent manner. From the 60 mg cohort, target engagement for Axl and CSF1R reached a inhibitory level that was observed in nonclinical models. In the pharmacodynamic biomarker analysis, IFN-ɣ in CD8+ T cells and non-T cell populations is increased. Monocytes and M-MDSC population are decreased in peripheral blood. Conclusion: Up to 240 mg, Q702 has demonstrated the intended pharmacologic activity with acceptable safety profile. In biomarker analysis, immune modulation activity is exerted by Axl/Mer/CSF1R inhibition. Further assessment of pharmacokinetics, pharmacodynamics, safety and antitumor activity will be performed at the expansion phase at the RP2D in patients with selected advanced tumors. Citation Format: Bae Jung Choi, Devalingam Devalingam, Angela Alistar, Anthony El-Khoueiry, Alain Mita, Hwankyu Kang, Jinho Choi, Hyunji Ahn, Jeongjun Kim, Seung-Joo Lee, Yeong-In Yang, Jiye Ahn, Borami Jeon, Jaeseung Kim, Kiyean Nam. Patient pharmacodynamic biomarker and pk evaluation results from an ongoing phase I dose-escalation study of q702, an axl, mer and csf1r kinase inhibitor in patients with advanced solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2255.
- Published
- 2023
17. The role of concomitant chemoradiotherapy in AJCC 7
- Author
-
Diana J, Lu, Michael, Luu, Anthony T, Nguyen, Stephen L, Shiao, Kevin, Scher, Alain, Mita, Eric, Anderson, Jon Mallen-St, Clair, Allen S, Ho, and Zachary S, Zumsteg
- Subjects
Aged, 80 and over ,Male ,Databases, Factual ,Papillomavirus Infections ,Standard of Care ,Chemoradiotherapy ,Comorbidity ,Alphapapillomavirus ,Middle Aged ,Combined Modality Therapy ,Oropharyngeal Neoplasms ,Treatment Outcome ,Practice Guidelines as Topic ,Humans ,Female ,Disease Susceptibility ,Neoplasm Metastasis ,Aged ,Neoplasm Staging ,Proportional Hazards Models - Abstract
Radiotherapy (RT) without chemotherapy is considered a standard of care for the management of American Joint Committee on Cancer (AJCC) 7th edition (7E) T1-2N1 oropharyngeal squamous cell carcinoma (OPSCC). Recent data suggests concurrent chemoradiation (CCRT) may benefit these patients but did not include human papillomavirus (HPV) status. Given the radiosensitivity differences between HPV-positive versus HPV-negative OPSCC, the effect of chemotherapy may differ in these patients.We analyzed patients in the National Cancer Database diagnosed between 2010 and 2015 with AJCC 7E stage cT1-2N1M0 OPSCC and known HPV status undergoing definitive RT or CCRT.Overall, 1964 patients were included, including 1297 (66%) HPV-positive and 667 (34%) HPV-negative patients. 66% received CCRT and 34% received RT alone. In multivariate analysis, CCRT was associated with improved survival compared with RT alone (hazard ratio [HR], 0.70; 95% confidence interval [CI], 0.57-0.87; P = 0.001). In propensity score-matched cohorts, 4-year overall survival was 87.4% vs 78.4% in HPV-positive patients receiving CCRT and RT alone, respectively (P = 0.002), and 65.5% vs 58.9% in HPV-negative patients, respectively (P = 0.2). There was no evidence that HPV-positivity diminished the association between CCRT and longer survival (HR, 0.57; 95% CI, 0.42-0.81) versus what was observed in HPV-negative patients (HR, 0.86; 95% CI, 0.64-1.16) (interaction P = 0.06).CCRT is associated with improved survival in AJCC 7E T1-2N1 OPSCC. Despite the radiosensitivity of HPV-positive OPSCC, the association of CCRT with improved survival for T1-2N1 HPV-positive OPSCC was at least as strong, if not stronger, than what was observed in HPV-negative patients.
- Published
- 2020
18. Perspectives in Head and Neck Medical Oncology
- Author
-
Idoroenyi, Amanam, Rohan, Gupta, Alain, Mita, Kevin, Scher, and Erminia, Massarelli
- Subjects
Head and Neck Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Carcinoma, Squamous Cell ,Humans ,Medical Oncology - Abstract
The modern treatment of locoregionally advanced disease often requires a multimodality combination approach. A number of chemotherapeutic agents can be combined with radiation, but the platinum agent cisplatin, a potent radiation sensitizer, is best studied in head and neck cancer. Newer agents such as cetuximab can be used in combination with radiation therapy for those patients who cannot tolerate cisplatin. For chemotherapy-naïve patients with metastatic head and neck cancer who demonstrate a good performance status, platinum doublet regimens are commonly used. Doublet regimens generally improve response rates compared to single-agent chemotherapies, although they have not demonstrated a survival benefit over single agents and they have added toxicity. Immunotherapies, alternative cytotoxic chemotherapies, and targeted therapies are second-line options for patients with disease that has progressed on platinum-based therapy. Immunotherapy, in particular, has gained focus by enhancing the ability of the immune system to recognize and destroy malignant cells. When multimodal approaches are used, as in combined chemotherapy and radiation therapy, toxicities are increased. It is imperative that patients are followed closely in order to maximize treatment benefit while minimizing complications.
- Published
- 2018
19. MTOR Inhibition for Cancer Therapy: Past, Present and Future
- Author
-
Monica Mita, Alain Mita, Eric K. Rowinsky, Monica Mita, Alain Mita, and Eric K. Rowinsky
- Subjects
- Molecular biology, Hematology, Cancer--Treatment, Medicine, Oncology
- Abstract
This book describes the challenges involved in developing mTOR inhibitors for cancer treatment, starting with an in-depth examination of their molecular mechanism of action, with emphasis on the class side-effects, efficacy and mechanisms of resistance, as well as on promising novel directions for their development, including novel compounds and rational combinations with other anti-neoplastic drugs. Over the last 10 years, inhibitors of mTOR have emerged as a major class of anticancer drugs. Two rapamycin analogs are currently approved for the treatment of renal cell carcinoma, and it is estimated that a variety of other tumor types could benefit from mTOR inhibition, with numerous clinical trials (including pivotal registration trials) already underway. Second-generation small-molecule inhibitors of the pathway have also shown promise in terms of their superior tolerability and efficacy and are undergoing extensive clinical evaluation, with an estimated 30+ compoundscurrently under evaluation.
- Published
- 2016
20. Impact of concomitant chemoradiation on survival for patients with T1-2N1 head and neck cancer
- Author
-
Zachary S, Zumsteg, Sungjin, Kim, John M, David, Emi J, Yoshida, Mourad, Tighiouart, Stephen L, Shiao, Kevin, Scher, Alain, Mita, Eric J, Sherman, Nancy Y, Lee, and Allen S, Ho
- Subjects
Male ,Hypopharyngeal Neoplasms ,Databases, Factual ,Radiotherapy ,Squamous Cell Carcinoma of Head and Neck ,Chemoradiotherapy ,Middle Aged ,Survival Rate ,Oropharyngeal Neoplasms ,Head and Neck Neoplasms ,Multivariate Analysis ,Carcinoma, Squamous Cell ,Humans ,Female ,Propensity Score ,Laryngeal Neoplasms ,Aged ,Neoplasm Staging ,Retrospective Studies - Abstract
Single-modality radiotherapy is considered a standard-of-care option for certain stage III, T1-2N1 head and neck squamous cell carcinomas (HNSCCs). The role of concomitant chemoradiation is not well established because there have been no studies comparing chemoradiation with radiation alone in this population.This study analyzed patients in the National Cancer Data Base with cT1-2N1M0 invasive squamous cell carcinomas of the oropharynx, larynx, and hypopharynx who were diagnosed between 2004 and 2012 and were undergoing definitive radiation. Patients who were undergoing surgery before radiation with unknown follow-up or for whom either the receipt or timing of chemotherapy was unknown were excluded.In all, 5030 patients with T1-2N1 oropharyngeal, laryngeal, or hypopharyngeal cancer were included. The median follow-up was 56.8 months (95% confidence interval [CI], 55.7-58.6 months). Overall, 68% of the patients received concomitant chemoradiation (CCRT). The use of CCRT significantly increased during the time period of this study from 53% in 2004 to 78% in 2012 (P.001). CCRT was associated with improved overall survival (OS) in comparison with radiation alone in a multivariate analysis (hazard ratio [HR], 0.80; 95% CI, 0.72-0.88; P.001). In propensity score-adjusted analyses, CCRT remained significantly associated with improved OS, with 5-year OS rates of 63.5% (95% CI, 60.7%-66.2%) and 55.6% (95% CI, 52.7%-58.4%; P.001) with CCRT and radiation alone, respectively. Subgroup analyses showed a benefit across the majority of subgroups, including patients with oropharyngeal cancer (HR, 0.74; 95% CI, 0.65-0.85; P.001).Concomitant chemoradiation is associated with improved survival for patients with T1-2N1 HNSCC. Prospective trials in this population should be pursued. Cancer 2017;123:1555-1565. © 2017 American Cancer Society.
- Published
- 2016
21. Phase I and pharmacokinetic study of YM155, a small-molecule inhibitor of survivin
- Author
-
Anne Keating, Pamela Bartels, E. Till, Chris Takimoto, Scott Antonia, Amita Patnaik, Adil Daud, Lionel D. Lewis, Alain Mita, Anthony W. Tolcher, Kyri Papadopoulos, and Christopher R. Garrett
- Subjects
Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Maximum Tolerated Dose ,Nausea ,Survivin ,Antineoplastic Agents ,Apoptosis ,Pilot Projects ,Pharmacology ,Drug Administration Schedule ,Inhibitor of Apoptosis Proteins ,Excretion ,chemistry.chemical_compound ,Pharmacokinetics ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Infusions, Intravenous ,Stomatitis ,Acute tubular necrosis ,Aged ,Volume of distribution ,Creatinine ,Dose-Response Relationship, Drug ,business.industry ,Imidazoles ,Middle Aged ,medicine.disease ,Phase I and Clinical Pharmacology ,Neoplasm Proteins ,Treatment Outcome ,Oncology ,chemistry ,Female ,medicine.symptom ,business ,Microtubule-Associated Proteins ,Signal Transduction ,Naphthoquinones - Abstract
Purpose To determine the maximum-tolerated dose (MTD) and assess the safety, pharmacokinetics, and preliminary evidence of antitumor activity of YM155, a small-molecule inhibitor of survivin. Patients and Methods Patients with advanced solid malignancies or lymphoma were treated with escalating doses of YM155 administered by 168-hour continuous intravenous infusion (CIVI). Plasma and urine samples were assayed to determine pharmacokinetic parameters and excretion. Results Forty-one patients received 127 cycles of YM155 at doses ranging from 1.8 to 6.0 mg/m2/d by 168-hour CIVI every 3 weeks. Overall, the most common grade 1 to 2 toxicities were stomatitis, pyrexia, and nausea, whereas grade 3 and 4 toxicities were rare. Reversible elevation in serum creatinine in two patients, with one developing acute tubular necrosis, was dose-limiting at 6.0 mg/m2. The MTD was 4.8 mg/m2. At the MTD, the mean steady-state concentration, clearance, volume of distribution at steady-state, and terminal elimination half-life were 7.7 ng/mL, 47.7 L/h, 1,763 L, and 26 hours, respectively. One complete and two partial responses lasting 8, 24+ and 48+ months occurred in three patients with non-Hodgkin's lymphoma, two patients with hormone- and docetaxel-refractory prostate cancer had prostate-specific antigen responses, and one patient with non–small-cell lung cancer had a minor response. Conclusion YM155 can be administered safely at 4.8 mg/m2/d 168 hours CIVI every 3 weeks. The absence of severe toxicities, attainment of plasma concentrations active in preclinical models, and compelling antitumor activity warrant further disease-directed studies of this agent alone and in combination with chemotherapy in a broad array of tumors.
- Published
- 2008
22. The molecular target of rapamycin (mTOR) as a therapeutic target against cancer
- Author
-
Monica M, Mita, Alain, Mita, and Eric K, Rowinsky
- Subjects
Sirolimus ,Antibiotics, Antineoplastic ,TOR Serine-Threonine Kinases ,Cell Cycle ,G1 Phase ,Antineoplastic Agents ,Cell Cycle Proteins ,Tacrolimus Binding Protein 1A ,Phosphoproteins ,Models, Biological ,Retinoblastoma Protein ,S Phase ,Phosphatidylinositol 3-Kinases ,Neoplasms ,Animals ,Humans ,RNA, Messenger ,Phosphorylation ,Carrier Proteins ,Protein Kinases ,Adaptor Proteins, Signal Transducing ,Protein Binding ,Signal Transduction - Abstract
The molecular target of rapamycin (mTOR), which is a member of the phosphoinositide 3-kinase related kinase (PIKK) family and a central modulator of cell growth, is a prime strategic target for anti-cancer therapeutic development. mTOR plays a critical role in transducing proliferative signals mediated through the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) signaling pathway, principally by activating downstream protein kinases that are required for both ribosomal biosynthesis and translation of key mRNAs of proteins required for G(1) to S phase traverse. By targeting mTOR, the immunsuppressant and antiproliferative agent rapamycin (RAP) inhibits signals required for cell cycle progression, cell growth, and proliferation. RAP, a complex macrolide and highly potent fungicide, immunosuppressant, and anti-cancer agent, is a highly specific inhibitor of mTOR. In essence, RAP gains function by binding to the immunophilin FK506 binding protein 12 (FKBP12) and the resultant complex inhibits the activity of mTOR. Since mTOR activates both the 40S ribosomal protein S6 kinase ((p)70(s6k)) and the eukaryotic initiation factor 4E-binding protein-1 (4E-BP1), RAP blocks activation of these downstream signaling elements, which results in cell cycle arrest in the G1 arrest. RAP also prevents cyclin-dependent kinase (cdk) activation, inhibits retinoblastoma protein ((p)Rb) phosphorylation, and accelerates the turnover of cyclin D1 that leads to a deficienciy of active cdk4/cyclin D1 complexes, all of which potentially contribute to the prominent inhibitory effects of RAP at the G(1)/S phase transition. Both RAP and several RAP analogs with more favorable pharmaceutical properties have demonstrated prominent growth inhibitory effects against a broad range of human cancers in both preclinical and early clinical evaluations. This review will summarize the principal mechanisms of action of RAP and RAP derivatives and their potential utility of these agents as anti-cancer therapeutics. The preliminary results of early clinical evaluations with RAP analogs and the unique developmental challenges that lie ahead will also be discussed.
- Published
- 2003
23. The absence of functional glucosylceramide synthase does not sensitize melanoma cells for anticancer drugs
- Author
-
Stéphane Carpentier, John D. Campbell, Jan Willem Kok, Virginie Garcia, Karin Klappe, Robert Jan Veldman, Alain Mita, Jeffrey A. Medin, Olivier Cuvillier, and Thierry Levade
- Subjects
EXPRESSION ,SPHINGOLIPIDS ,Ceramide ,Cell Survival ,Melanoma, Experimental ,PROTEIN ,Antineoplastic Agents ,DAUNORUBICIN-INDUCED APOPTOSIS ,Biology ,GM95 melanoma cells ,Ceramides ,Transfection ,Biochemistry ,Models, Biological ,ACTIVATION ,chemistry.chemical_compound ,Mice ,multidrug resistance ,GLYCOLIPID BIOSYNTHESIS ,ADRIAMYCIN RESISTANCE ,Genetics ,Tumor Cells, Cultured ,polycyclic compounds ,Animals ,ceramide ,MULTIDRUG-RESISTANCE ,CANCER-CELLS ,Cytotoxicity ,Molecular Biology ,apoptosis ,Lipid signaling ,Sphingolipid ,chemistry ,Cell culture ,Apoptosis ,Doxorubicin ,Drug Resistance, Neoplasm ,Glucosyltransferases ,Cancer cell ,Cancer research ,ATP-Binding Cassette Transporters ,CERAMIDE GLYCOSYLATION ,Gene Deletion ,Biotechnology - Abstract
Conversion of ceramide, a putative mediator of anticancer drug-induced apoptosis, into glucosylceramide, by the action of glucosylceramide synthase (GCS), has been implicated in drug resistance. Herein, we compared GM95 mouse melanoma cells deficient in GCS activity, with cells stably transfected with a vector encoding GCS (GM95/GCS). Enzymatic and metabolic analysis demonstrated that GM95/GCS cells expressed a fully functional enzyme, resulting in normal ceramide glycosylation. However, cytotoxicity assays, as well as caspase activation and cytochrome c release studies, did not reveal any difference between the two cell lines with respect to their sensitivity toward doxorubicin, vinblastine, paclitaxel, cytosine arabinoside, or short-chain ceramide analogs. Administration of doxorubicin resulted in ceramide accumulation in both cell lines, with similar kinetics and amplitude. Although glucosylceramide formation was detected in doxorubicin-treated GM95/GCS cells, metabolism of drug-induced ceramide did not appear to be instrumental in cell survival. Furthermore, N-(n-butyl)deoxynojirimycin, a potent and non-toxic GCS inhibitor, had no chemosensitizing effect on wild-type melanoma cells. Altogether, both genetic and pharmacological alterations of the cellular ceramide glycosylation capacity failed to sensitize melanoma cells to anticancer drugs, therefore moderating the importance of ceramide glucosylation in drug-resistance mechanisms.
24. AKT Inhibition in Solid Tumors With AKT1 Mutations.
- Author
-
Hyman DM, Smyth LM, Donoghue MTA, Westin SN, Bedard PL, Dean EJ, Bando H, El-Khoueiry AB, Pérez-Fidalgo JA, Mita A, Schellens JHM, Chang MT, Reichel JB, Bouvier N, Selcuklu SD, Soumerai TE, Torrisi J, Erinjeri JP, Ambrose H, Barrett JC, Dougherty B, Foxley A, Lindemann JPO, McEwen R, Pass M, Schiavon G, Berger MF, Chandarlapaty S, Solit DB, Banerji U, Baselga J, and Taylor BS
- Subjects
- Adult, Aged, Alleles, Antineoplastic Agents therapeutic use, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Diarrhea chemically induced, Disease-Free Survival, Drug Eruptions etiology, Exanthema chemically induced, Female, Humans, Hyperglycemia chemically induced, Loss of Heterozygosity, Male, Middle Aged, Neoplasms blood, Phosphatidylinositol 3-Kinases metabolism, Protein Kinase Inhibitors therapeutic use, Pyrimidines therapeutic use, Pyrroles therapeutic use, Response Evaluation Criteria in Solid Tumors, Signal Transduction genetics, Antineoplastic Agents adverse effects, DNA, Neoplasm blood, Mutation, Neoplasms drug therapy, Neoplasms genetics, Protein Kinase Inhibitors adverse effects, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt genetics, Pyrimidines adverse effects, Pyrroles adverse effects
- Abstract
Purpose AKT1 E17K mutations are oncogenic and occur in many cancers at a low prevalence. We performed a multihistology basket study of AZD5363, an ATP-competitive pan-AKT kinase inhibitor, to determine the preliminary activity of AKT inhibition in AKT-mutant cancers. Patients and Methods Fifty-eight patients with advanced solid tumors were treated. The primary end point was safety; secondary end points were progression-free survival (PFS) and response according to Response Evaluation Criteria in Solid Tumors (RECIST). Tumor biopsies and plasma cell-free DNA (cfDNA) were collected in the majority of patients to identify predictive biomarkers of response. Results In patients with AKT1 E17K-mutant tumors (n = 52) and a median of five lines of prior therapy, the median PFS was 5.5 months (95% CI, 2.9 to 6.9 months), 6.6 months (95% CI, 1.5 to 8.3 months), and 4.2 months (95% CI, 2.1 to 12.8 months) in patients with estrogen receptor-positive breast, gynecologic, and other solid tumors, respectively. In an exploratory biomarker analysis, imbalance of the AKT1 E17K-mutant allele, most frequently caused by copy-neutral loss-of-heterozygosity targeting the wild-type allele, was associated with longer PFS (hazard ratio [HR], 0.41; P = .04), as was the presence of coincident PI3K pathway hotspot mutations (HR, 0.21; P = .045). Persistent declines in AKT1 E17K in cfDNA were associated with improved PFS (HR, 0.18; P = .004) and response ( P = .025). Responses were not restricted to patients with detectable AKT1 E17K in pretreatment cfDNA. The most common grade ≥ 3 adverse events were hyperglycemia (24%), diarrhea (17%), and rash (15.5%). Conclusion This study provides the first clinical data that AKT1 E17K is a therapeutic target in human cancer. The genomic context of the AKT1 E17K mutation further conditioned response to AZD5363.
- Published
- 2017
- Full Text
- View/download PDF
25. Phase I Dose-Escalation Study of JNJ-42756493, an Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients With Advanced Solid Tumors.
- Author
-
Tabernero J, Bahleda R, Dienstmann R, Infante JR, Mita A, Italiano A, Calvo E, Moreno V, Adamo B, Gazzah A, Zhong B, Platero SJ, Smit JW, Stuyckens K, Chatterjee-Kishore M, Rodon J, Peddareddigari V, Luo FR, and Soria JC
- Subjects
- Administration, Oral, Adult, Aged, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Dose-Response Relationship, Drug, Drug Administration Schedule, Humans, Middle Aged, Neoplasms blood, Neoplasms metabolism, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Pyrazoles adverse effects, Pyrazoles pharmacokinetics, Quinoxalines adverse effects, Quinoxalines pharmacokinetics, Antineoplastic Agents administration & dosage, Neoplasms drug therapy, Protein Kinase Inhibitors administration & dosage, Pyrazoles administration & dosage, Quinoxalines administration & dosage, Receptors, Fibroblast Growth Factor antagonists & inhibitors
- Abstract
Purpose: JNJ-42756493 is an orally administered pan-fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor. This first-in-human study evaluates the safety, pharmacokinetics, and pharmacodynamics and defines the recommended phase II dose (RP2D) of JNJ-42756493., Patients and Methods: Eligible patients with advanced solid tumors received escalating doses of JNJ-42756493 from 0.5 to 12 mg administered continuously daily or JNJ-42756493 10 or 12 mg administered intermittently (7 days on/7 days off)., Results: Sixty-five patients were enrolled. The most common treatment-emergent adverse events included hyperphosphatemia (65%), asthenia (55%), dry mouth (45%), nail toxicity (35%), constipation (34%), decreased appetite (32%), and dysgeusia (31%). Twenty-seven patients (42%) experienced grade ≥ 3 treatment-emergent adverse events, and one dose-limiting toxicity of grade 3 ALT elevation was observed at 12 mg daily. Maximum-tolerated dose was not defined. Nine milligrams daily was considered as the initial RP2D; however, tolerability was improved with intermittent schedules, and 10 mg administered on a 7-days-on/7-days-off schedule was considered the final RP2D. Pharmacokinetics were linear, dose proportional, and predictable, with a half-life of 50 to 60 hours. Dose-dependent elevations in serum phosphate, a manifestation of pharmacodynamic effect, occurred in all patients starting at 4 mg daily. Among 23 response-evaluable patients with tumor FGFR pathway alterations, four confirmed responses and one unconfirmed partial response were observed in patients with glioblastoma and urothelial and endometrial cancer (all with FGFR2 or FGFR3 translocations); 16 patients had stable disease., Conclusion: JNJ-42756493 administered at 10 mg on a 7-days-on/7-days-off schedule achieved exposures at which clinical responses were observed, demonstrated pharmacodynamic biomarker activity, and had a manageable safety profile., (© 2015 by American Society of Clinical Oncology.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.