65 results on '"Yuan-Li Shen"'
Search Results
2. Designing Dose-Optimization Studies in Cancer Drug Development: Discussions with Regulators
- Author
-
Olga Marchenko, Rajeshwari Sridhara, Qi Jiang, Elizabeth Barksdale, Yuki Ando, Dinesh De Alwis, Katie Brown, Laura Fernandes, Mark T. J. van Bussel, Qiuyi Choo, Michael Coory, Elizabeth Garrett-Mayer, Thomas Gwise, Lorenzo Hess, Rong Liu, Sumithra Mandrekar, Daniele Ouellet, José Pinheiro, Martin Posch, Nam Atiqur Rahman, Khadija Rerhou Rantell, Andrew Raven, Sarem Sarem, Suman Sen, Mirat Shah, Yuan Li Shen, Richard Simon, Marc Theoret, Ying Yuan, and Richard Pazdur
- Subjects
Statistics and Probability ,Pharmaceutical Science - Published
- 2023
3. Rejoinder to Comments on 'Non-Proportional Hazards – An Evaluation of the MaxCombo Test in Cancer Clinical Trials'
- Author
-
Yuan-Li Shen, Sirisha Mushti, Flora Mulkey, Thomas Gwise, Xin Wang, Jiaxi Zhou, Xin Gao, Shenghui Tang, Marc R. Theoret, Richard Pazdur, and Rajeshwari Sridhara
- Subjects
Statistics and Probability ,Pharmaceutical Science - Published
- 2023
4. Evaluation of Treatment Effect in Underrepresented Population in Cancer Trials: Discussion with International Regulators
- Author
-
Rajeshwari Sridhara, Olga Marchenko, Qi Jiang, Elizabeth Barksdale, Jie Chen, Nancy Dreyer, Lola Fashoyin-Aje, Elizabeth Garrett-Mayer, Nicole Gormley, Thomas Gwise, Lorenzo Hess, Sumithra Mandrekar, Francesco Pignatti, Khadija Rantell, Andrew Raven, Yuan-Li Shen, Harpreet Singh, Craig L. Tendler, Marc Theoret, and Richard Pazdur
- Subjects
Statistics and Probability ,Pharmaceutical Science - Published
- 2022
5. Cancer Clinical Trials beyond Pandemic: Report of an American Statistical Association Biopharmaceutical Section Open Forum Discussion
- Author
-
Rajeshwari Sridhara, Elizabeth Barksdale, Olga Marchenko, Qi Jiang, Yuki Ando, Erick Bloomquist, Michael Coory, Melissa Crouse, Evgeny Degtyarev, Theodor Framke, Boris Freidlin, David E. Gerber, Thomas Gwise, Filip Josephson, Lorenzo Hess, Paul Kluetz, Daniel Li, Sumithra Mandrekar, Martin Posch, Khadija Rantell, Bohdana Ratitch, Andrew Raven, Kit Roes, Kaspar Rufibach, Sinan B. Sarac, Richard Simon, Harpreet Singh, Marc Theoret, Andrew Thomson, Emmanuel Zuber, Yuan Li Shen, and Richard Pazdur
- Subjects
Statistics and Probability ,Pharmaceutical Science - Published
- 2022
6. Supplementary Table S1. Use of Grouped Termsa for Summary Tabulations of Adverse Reactions from FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Richard Pazdur, Ann T. Farrell, Reena Philip, Albert Deisseroth, Rajeshwari Sridhara, Donna Roscoe, Yuan Li Shen, Donna Przepiorka, Aaron Schetter, Qing Xu, and Emily Y. Jen
- Abstract
Grouped terms used in safety analyses
- Published
- 2023
7. Supplementary Figure S1: Overall Survival Subpopulation Analysis from FDA Approval Summary: Glasdegib for Newly Diagnosed Acute Myeloid Leukemia
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Ruby Leong, Chao Liu, Christopher M. Sheth, Yuan-Li Shen, Donna Przepiorka, Pedro L. Del Valle, Luning Zhuang, Sriram Subramaniam, Kunthel By, and Kelly J. Norsworthy
- Abstract
Supplementary Figure 1
- Published
- 2023
8. Supplementary Figure S1 from FDA Approval Summary: Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Gene M. Williams, Rosane Charlab, Christopher M. Sheth, Yuan-Li Shen, Albert Deisseroth, Donna Przepiorka, Sarah E. Dorff, Ramadevi Gudi, Vicky Hsu, Lola Luo, and Kelly J. Norsworthy
- Abstract
Rate of CR+CRh by subgroup
- Published
- 2023
9. Supplementary Table S2: Laboratory Abnormalities from FDA Approval Summary: Glasdegib for Newly Diagnosed Acute Myeloid Leukemia
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Ruby Leong, Chao Liu, Christopher M. Sheth, Yuan-Li Shen, Donna Przepiorka, Pedro L. Del Valle, Luning Zhuang, Sriram Subramaniam, Kunthel By, and Kelly J. Norsworthy
- Abstract
Supplementary Table S2
- Published
- 2023
10. Data from FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Richard Pazdur, Ann T. Farrell, Reena Philip, Albert Deisseroth, Rajeshwari Sridhara, Donna Roscoe, Yuan Li Shen, Donna Przepiorka, Aaron Schetter, Qing Xu, and Emily Y. Jen
- Abstract
On March 29, 2018, the FDA granted accelerated approval for blinatumomab (Blincyto; Amgen, Inc.) for the treatment of adults and children with B-cell precursor acute lymphoblastic leukemia (BCP ALL) in first or second complete remission with minimal residual disease (MRD) greater than or equal to 0.1%. Blinatumomab is a CD3xCD19 bispecific antibody approved previously for the treatment of relapsed or refractory BCP ALL. The basis for this accelerated approval was a single-arm trial. For the 86 patients in first and second complete remission with MRD ≥ 0.1%, conversion to MRD < 0.01% was achieved after one cycle of blinatumomab by 85.2% [95% confidence interval (CI): 73.8%, 93.0%] and 72.0% (95% CI: 50.6%, 87.9%), respectively, and the estimated median hematologic relapse-free survivals (RFS) were 35.2 months (95% CI: 0.4–53.5) and 12.3 months (95% CI: 0.7–42.3), respectively. Hematologic RFS was considered substantial independent of whether patients underwent subsequent allogeneic stem cell transplantation. The safety profile for blinatumomab was established in prior studies, and no new safety signals were observed in the new population. Cytokine release syndrome and neurotoxicity remain significant risks. The FDA is requiring confirmation of clinical benefit in a randomized trial.
- Published
- 2023
11. Supplementary Figure S1. Study 20120148 - Hematological relapse-free survival for patients in CR1 by MRD level at baseline after at least 3 intensive chemotherapy blocks from FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Richard Pazdur, Ann T. Farrell, Reena Philip, Albert Deisseroth, Rajeshwari Sridhara, Donna Roscoe, Yuan Li Shen, Donna Przepiorka, Aaron Schetter, Qing Xu, and Emily Y. Jen
- Abstract
RFS by baseline MRD level from Study 20120148
- Published
- 2023
12. Supplementary Table S1 from FDA Approval Summary: Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Gene M. Williams, Rosane Charlab, Christopher M. Sheth, Yuan-Li Shen, Albert Deisseroth, Donna Przepiorka, Sarah E. Dorff, Ramadevi Gudi, Vicky Hsu, Lola Luo, and Kelly J. Norsworthy
- Abstract
Preferred Terms included in grouped terms
- Published
- 2023
13. Supplementary Table S1: Preferred Terms included in Grouped Terms from FDA Approval Summary: Glasdegib for Newly Diagnosed Acute Myeloid Leukemia
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Ruby Leong, Chao Liu, Christopher M. Sheth, Yuan-Li Shen, Donna Przepiorka, Pedro L. Del Valle, Luning Zhuang, Sriram Subramaniam, Kunthel By, and Kelly J. Norsworthy
- Abstract
Supplementary Table S1
- Published
- 2023
14. Supplementary Table S1: FDA Capillary Leak Syndrome Screening Criteria from FDA Approval Summary: Tagraxofusp-erzs For Treatment of Blastic Plasmacytoid Dendritic Cell Neoplasm
- Author
-
Richard Pazdur, Gideon M. Blumenthal, Ann T. Farrell, Kirsten B. Goldberg, Steven Bowen, Christopher M. Sheth, Chao Liu, Ruby Leong, Yuan Li Shen, Donna Przepiorka, Rong Wang, Baikuntha Aryal, Natalie E. Simpson, Luning Zhuang, Liang Li, Xin Gao, and Emily Y. Jen
- Abstract
Supplementary Table 1 contains FDA's screening algorithm for capillary leak syndrome (CLS).
- Published
- 2023
15. Data from FDA Approval Summary: Glasdegib for Newly Diagnosed Acute Myeloid Leukemia
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Ruby Leong, Chao Liu, Christopher M. Sheth, Yuan-Li Shen, Donna Przepiorka, Pedro L. Del Valle, Luning Zhuang, Sriram Subramaniam, Kunthel By, and Kelly J. Norsworthy
- Abstract
On November 21, 2018, the FDA approved glasdegib (Daurismo; Pfizer), a small-molecule Hedgehog inhibitor, in combination with low-dose cytarabine (LDAC) for treatment of newly diagnosed acute myeloid leukemia (AML) in adults ≥ 75 years or with comorbidities that preclude use of intensive induction chemotherapy. Evidence of clinical benefit came from Study BRIGHT AML 1003, a randomized trial comparing glasdegib+LDAC with LDAC alone for treatment of newly diagnosed AML in 115 patients either ≥ 75 years old or ≥ 55 years old with preexisting comorbidities. Efficacy was established by improved overall survival (OS) with the combination compared with LDAC alone (HR, 0.46; 95% confidence interval, 0.30–0.71; one-sided stratified log-rank P = 0.0002). Median OS was 8.3 months with the combination and 4.3 months with LDAC alone. Common adverse reactions included cytopenias, fatigue, hemorrhage, febrile neutropenia, musculoskeletal pain, nausea, edema, dyspnea, decreased appetite, dysgeusia, mucositis, constipation, and rash. The label includes a boxed warning for embryo-fetal toxicity and a warning for QT interval prolongation. There is a limitation of use for patients with moderate-to-severe hepatic and severe renal impairment; trials studying glasdegib in these patient populations are required as a condition of this approval.See related commentary by Fathi, p. 6015
- Published
- 2023
16. Data from FDA Approval Summary: Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation
- Author
-
Richard Pazdur, Ann T. Farrell, Kirsten B. Goldberg, Gene M. Williams, Rosane Charlab, Christopher M. Sheth, Yuan-Li Shen, Albert Deisseroth, Donna Przepiorka, Sarah E. Dorff, Ramadevi Gudi, Vicky Hsu, Lola Luo, and Kelly J. Norsworthy
- Abstract
The FDA approved ivosidenib (Tibsovo; Agios), a small-molecule inhibitor of isocitrate dehydrogenase (IDH)1 on July 20, 2018, for treatment of adults with relapsed or refractory acute myeloid leukemia (R/R AML) with susceptible IDH1 mutation as detected by an FDA-approved test. The efficacy of ivosidenib was established on the basis of complete remission (CR) + CR with partial hematologic recovery (CRh) rate, duration of CR + CRh, and conversion from transfusion dependence (TD) to transfusion independence (TI) in Study AG120-C-001, a single-arm trial. With median follow-up of 8.3 months for 174 adults with IDH1-mutated R/R AML treated with 500 mg ivosidenib daily, the CR + CRh rate was 33% [95% confidence interval (CI), 26–40], median duration of response was 8.2 (95% CI, 5.6–12) months, and conversion from TD to TI occurred in 37% of patients. These endpoints reflect short-term benefit in patients with an unmet medical need; long-term efficacy outcomes were not assessed. Serious adverse reactions (AR) in ≥5% of patients were differentiation syndrome (10%), leukocytosis (10%), and QT interval prolongation (7%). Common (≥20%) ARs of any grade were fatigue, leukocytosis, arthralgia, diarrhea, dyspnea, edema, nausea, mucositis, QT interval prolongation, rash, pyrexia, cough, and constipation. Assessment of long-term safety of ivosidenib is a condition of this approval.
- Published
- 2023
17. Supplementary Table S3. Safety Population - Grade {greater than or equal to} 3 Adverse Reactions from FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Richard Pazdur, Ann T. Farrell, Reena Philip, Albert Deisseroth, Rajeshwari Sridhara, Donna Roscoe, Yuan Li Shen, Donna Przepiorka, Aaron Schetter, Qing Xu, and Emily Y. Jen
- Abstract
Grade >/= 3 AR
- Published
- 2023
18. Data from FDA Approval Summary: (Daunorubicin and Cytarabine) Liposome for Injection for the Treatment of Adults with High-Risk Acute Myeloid Leukemia
- Author
-
Richard Pazdur, Gideon M. Blumenthal, Ann T. Farrell, Kirsten B. Goldberg, Jiang Liu, Anamitro Banerjee, Christopher M. Sheth, Stacy S. Shord, Yuan Li Shen, Donna Przepiorka, David A. Bateman, Gerlie Gieser, Kumar G. Janoria, Wentao Fu, Paresma Patel, Michael L. Manning, Liang Li, Xin Gao, and Aviva C. Krauss
- Abstract
On August 3, 2017, the FDA granted regular approval to Vyxeos (also known as CPX-351; Jazz Pharmaceuticals), a liposomal formulation of daunorubicin and cytarabine in a fixed combination, for the treatment of adults with newly diagnosed therapy-related acute myeloid leukemia (t-AML) or acute myeloid leukemia (AML) with myelodysplasia-related changes (AML-MRC). Approval was based on data from Study CLTR0310-301, a randomized, multicenter, open-label, active-controlled trial comparing Vyxeos with a standard combination of daunorubicin and cytarabine (“7+3”) in 309 patients 60–75 years of age with newly diagnosed t-AML or AML-MRC. Because of elemental copper concerns with the Vyxeos formulation, patients with Wilson disease were excluded from the study. Vyxeos demonstrated an improvement in overall survival (HR 0.69; 95% confidence interval, 0.52–0.90; P = 0.005) with an estimated median overall survival of 9.6 months compared with 5.9 months for the “7+3” control arm. The toxicity profile of Vyxeos was similar to that seen with standard “7+3” with the exception of more prolonged neutropenia and thrombocytopenia on the Vyxeos arm. Because the pharmacology of Vyxeos differs from that of other formulations of daunorubicin and cytarabine, labeling includes a warning against interchanging formulations during treatment. This is the first FDA-approved treatment specifically for patients with t-AML or AML-MRC.
- Published
- 2023
19. Supplemental Table S1 from FDA Approval Summary: (Daunorubicin and Cytarabine) Liposome for Injection for the Treatment of Adults with High-Risk Acute Myeloid Leukemia
- Author
-
Richard Pazdur, Gideon M. Blumenthal, Ann T. Farrell, Kirsten B. Goldberg, Jiang Liu, Anamitro Banerjee, Christopher M. Sheth, Stacy S. Shord, Yuan Li Shen, Donna Przepiorka, David A. Bateman, Gerlie Gieser, Kumar G. Janoria, Wentao Fu, Paresma Patel, Michael L. Manning, Liang Li, Xin Gao, and Aviva C. Krauss
- Abstract
Supplemental Table S1: Grouped Terms for Adverse Reactions on Study 301
- Published
- 2023
20. Supplementary Table S4. Literature Review - Efficacy Outcomes by MRD Log-Group for B-cell ALL CR1 using End-Of-Induction MRD Measurements from FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Richard Pazdur, Ann T. Farrell, Reena Philip, Albert Deisseroth, Rajeshwari Sridhara, Donna Roscoe, Yuan Li Shen, Donna Przepiorka, Aaron Schetter, Qing Xu, and Emily Y. Jen
- Abstract
Literature review - efficacy by log-group MRD
- Published
- 2023
21. Data from FDA Approval Summary: Tagraxofusp-erzs For Treatment of Blastic Plasmacytoid Dendritic Cell Neoplasm
- Author
-
Richard Pazdur, Gideon M. Blumenthal, Ann T. Farrell, Kirsten B. Goldberg, Steven Bowen, Christopher M. Sheth, Chao Liu, Ruby Leong, Yuan Li Shen, Donna Przepiorka, Rong Wang, Baikuntha Aryal, Natalie E. Simpson, Luning Zhuang, Liang Li, Xin Gao, and Emily Y. Jen
- Abstract
Tagraxofusp-erzs (Elzonris, Stemline) is a cytotoxin that targets CD123-expressing cells. On December 21, 2018, FDA approved tagraxofusp-erzs for the treatment of blastic plasmacytoid dendritic cell neoplasms (BPDCN) in adult and pediatric patients 2 years and older. Approval was based on the response rate in a single-arm trial, Study STML-401-0114; the pivotal cohort included 13 patients with treatment-naïve BPDCN who received tagraxofusp-erzs monotherapy. The complete response or clinical complete response (CR/CRc) rate in the pivotal cohort was 54% (95% CI: 25%–81%), and the median duration of CR/CRc was not reached with a median follow-up of 11.5 months (range: 0.2–12.7). In a separate exploratory cohort, a CR/CRc was achieved by 2 (13%) patients with R/R BPDCN. Safety was assessed in 94 patients with myeloid neoplasms treated with tagraxofusp-erzs at the approved dose and schedule. The major toxicity was capillary leak syndrome (CLS), which occurred in 55% of patients and was fatal in 2%. Hepatotoxicity and hypersensitivity reactions were reported in 88% and 46% of patients, respectively. Other common (≥30%) adverse reactions were nausea, fatigue, peripheral edema, pyrexia, and weight increase. A high proportion of patients (85%) developed neutralizing antidrug antibodies. Tagraxofusp-erzs is the first FDA-approved treatment for BPDCN.
- Published
- 2023
22. Supplementary Table S5. Literature Review - Efficacy Outcomes by Binary MRD Level for B-cell ALL from FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Richard Pazdur, Ann T. Farrell, Reena Philip, Albert Deisseroth, Rajeshwari Sridhara, Donna Roscoe, Yuan Li Shen, Donna Przepiorka, Aaron Schetter, Qing Xu, and Emily Y. Jen
- Abstract
Literature review - efficacy by binary MRD outcome
- Published
- 2023
23. Supplementary Table S1 from FDA Approval Summary: Osimertinib for Adjuvant Treatment of Surgically Resected Non–Small Cell Lung Cancer, a Collaborative Project Orbis Review
- Author
-
Harpreet Singh, Julia A. Beaver, Richard Pazdur, Dianne Spillman, Nataliya Fesenko, Ulrich-Peter Rohr, Arūnas Girčys, Qiuyi Choo, Aleksandr Gamarian, Elitza Palazov, Jeanne Fourie Zirkelbach, Jiang Liu, Yangbing Li, Huiming Xia, Yuan Li Shen, Pallavi S. Mishra-Kalyani, Xiaoxue Li, Nicole Drezner, Paz J. Vellanki, and Abigail L. Koch
- Abstract
Lung Cancer Staging per AJCC 7th and 8th Editions
- Published
- 2023
24. Response Rate, Event-Free Survival, and Overall Survival in Newly Diagnosed Acute Myeloid Leukemia: US Food and Drug Administration Trial-Level and Patient-Level Analyses
- Author
-
Kelly J. Norsworthy, Xin Gao, Chia-Wen Ko, E. Dianne Pulte, Jiaxi Zhou, Yutao Gong, Yuan Li Shen, Jonathon Vallejo, Thomas E. Gwise, Rajeshwari Sridhara, Albert B. Deisseroth, Ann T. Farrell, R. Angelo de Claro, Gideon M. Blumenthal, and Richard Pazdur
- Subjects
Cancer Research ,United States Food and Drug Administration ,Remission Induction ,Cytarabine ,ORIGINAL REPORTS ,Disease-Free Survival ,Progression-Free Survival ,United States ,Leukemia, Myeloid, Acute ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Biomarkers ,Randomized Controlled Trials as Topic - Abstract
PURPOSE To explore trial-level and patient-level associations between response (complete remission [CR] and CR + CR with incomplete hematologic [CRi] or platelet [CRp] recovery), event-free survival (EFS), and overall survival (OS) in newly diagnosed acute myeloid leukemia (AML) trials of intensive chemotherapy. METHODS We identified data from eight randomized, active-controlled trials of intensive chemotherapy submitted to the US Food and Drug Administration for treatment of newly diagnosed AML (N = 4,482). Associations between trial-level odds ratios (ORs) for CR and CR + CRi or CRp, and hazard ratios (HRs) for EFS and OS were analyzed using weighted linear regression models. We performed patient-level responder analyses to compare OS by response using pooled data from all studies. RESULTS In trial-level analyses, association between HR for OS and OR for CR was moderate (R2 = 0.49; 95% CI, 0.05 to 0.86), as was the association with OR for CR + CRi or CRp (R2 = 0.48; 95% CI, 0.05 to 0.99). For OS versus EFS, a strong association was observed (R2 = 0.87; 95% CI, 0.47 to 0.98) when EFS definitions were harmonized across trials using raw data. In the patient-level responder analyses, patients who achieved CR had better OS compared with CRi or CRp responders (0.73; 95% CI, 0.64 to 0.84) and nonresponders (HR, 0.33; 95% CI, 0.31 to 0.37). CONCLUSION On a trial level, there is a moderate association between OS and CR rate. A strong association between EFS and OS was observed. However, CIs were wide, and results became moderate using alternative definitions for EFS. Patient-level analyses showed CR responders have better OS compared with CRi or CRp responders and nonresponders. A therapy in newly diagnosed AML with benefit in EFS or substantial benefit in CR rate would be likely to have an OS effect.
- Published
- 2023
25. Analysis of racial and ethnic disparities in multiple myeloma US FDA drug approval trials
- Author
-
Bindu Kanapuru, Laura L. Fernandes, Lola A. Fashoyin-Aje, Andrea C. Baines, Vishal Bhatnagar, Rachel Ershler, Thomas Gwise, Paul Kluetz, Richard Pazdur, Elizabeth Pulte, Yuan-Li Shen, and Nicole Gormley
- Subjects
Black or African American ,Ethnicity ,Humans ,Hispanic or Latino ,Hematology ,Multiple Myeloma ,Drug Approval ,United States - Abstract
African Americans (AAs) have a higher incidence of multiple myeloma (MM) than White patients. Mortality is also higher in AAs compared with White patients. AAs more commonly have immunoglobulin H translocations t(11;14) and t(14;16) compared with White patients. We sought to characterize the demographic representation in MM clinical trials and evaluate outcomes based on race and ethnicity. We conducted a pooled analysis of all trials submitted to the US Food and Drug Administration (FDA) to support approval of a MM therapeutic between 2006 and 2019. Demographic characteristics were analyzed descriptively. An age-adjusted stratified Cox regression model was used to evaluate the relationship between time-to-event outcomes and race and ethnicity. Nineteen global trials comprising 10 157 patients were pooled. White, Asian, and Black patients comprised 84%, 7%, and 4% of the dataset, respectively; Hispanic patients comprised 4%. The age-adjusted overall survival hazard ratio (HR) for Black compared with White patients was 0.89 (95% confidence interval [CI], 0.75-1.05). The age-adjusted HR for US Black vs US White patients was 0.82 (95% CI, 0.66-1.02). For rest-of-world (RoW) Black vs RoW White patients, the HR was 1.31 (95% CI, 0.97-1.77). Black and Hispanic patients were underrepresented in the trials supporting FDA approval of MM drugs. Black patients were primarily enrolled in the United States. Outcomes in US patients were more favorable compared with those in patients in the RoW. Given the higher incidence of MM in AAs and the different disease characteristics, efforts should be made to improve representation of AAs in MM clinical trials.
- Published
- 2022
26. Nonproportional Hazards—An Evaluation of the MaxCombo Test in Cancer Clinical Trials
- Author
-
Yuan-Li Shen, Xin Wang, Mushti Sirisha, Flora Mulkey, Jiaxi Zhou, Xin Gao, Lijun Zhang, Thomas Gwise, Shenghui Tang, Marc Theoret, Richard Pazdur, and Rajeshwari Sridhara
- Subjects
Statistics and Probability ,Pharmaceutical Science - Published
- 2022
27. FDA Approval Summary: Osimertinib for Adjuvant Treatment of Surgically Resected Non–Small Cell Lung Cancer, a Collaborative Project Orbis Review
- Author
-
Elitza Palazov, Yangbing Li, Harpreet Singh, Jeanne Fourie Zirkelbach, Julia A. Beaver, Dianne Spillman, Arūnas Girčys, Jiang Liu, Aleksandr Gamarian, Huiming Xia, Qiuyi Choo, Paz J. Vellanki, Pallavi S. Mishra-Kalyani, Nataliya Fesenko, Nicole Drezner, Ulrich-Peter Rohr, Richard Pazdur, Yuan Li Shen, Xiaoxue Li, and Abigail L. Koch
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Population ,Placebo ,Targeted therapy ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Adjuvant therapy ,Humans ,Osimertinib ,Stage (cooking) ,education ,Lung cancer ,Protein Kinase Inhibitors ,Acrylamides ,education.field_of_study ,Aniline Compounds ,business.industry ,medicine.disease ,ErbB Receptors ,Mutation ,business ,Biomedical sciences - Abstract
On December 18, 2020, the FDA approved osimertinib as adjuvant therapy in patients with non–small cell lung cancer (NSCLC) whose tumors have EGFR exon 19 deletions or exon 21 (L858R) mutations, as detected by an FDA-approved test. The approval was based on the ADAURA study, in which 682 patients with NSCLC were randomized to receive osimertinib (n = 339) or placebo (n = 343). Disease-free survival (DFS) in the overall population (stage IB–IIIA) was improved for patients who received osimertinib, with an HR of 0.20; 95% confidence interval (CI), 0.15–0.27; P < 0.0001. Median DFS was not reached for the osimertinib arm compared with 27.5 months (95% CI, 22.0–35.0) for patients receiving placebo. Overall survival data were not mature at the time of the approval. This application was reviewed under FDA's Project Orbis, in collaboration with Australia Therapeutic Goods Administration, Brazil ANVISA, Health Canada, Singapore Health Sciences Authority, Switzerland Swissmedic, and the United Kingdom Medicines and Healthcare products Regulatory Agency. This is the first targeted therapy adjuvant approval for NSCLC and has practice-changing implications.
- Published
- 2021
28. FDA Approval Summary: Cabozantinib for Differentiated Thyroid Cancer
- Author
-
Elizabeth S. Duke, Amy K. Barone, Somak Chatterjee, Pallavi S. Mishra-Kalyani, Yuan-Li Shen, Emasenyie Isikwei, Hong Zhao, Youwei Bi, Jiang Liu, Nam Atiqur Rahman, Emily Wearne, John K. Leighton, Maritsa Stephenson, Idara Ojofeitimi, Barbara Scepura, Abhilasha Nair, Richard Pazdur, Julia A. Beaver, and Harpreet Singh
- Subjects
Adult ,Iodine Radioisotopes ,Cancer Research ,Oncology ,Pyridines ,Humans ,Angiogenesis Inhibitors ,Anilides ,Thyroid Neoplasms ,Adenocarcinoma ,Child ,Protein Kinase Inhibitors ,Article - Abstract
On September 17, 2021, the U.S. Food and Drug Administration (FDA) approved cabozantinib (Cabometyx™; Exelixis, Inc.) for the treatment of adult and pediatric patients 12 years of age and older with locally advanced or metastatic differentiated thyroid cancer (DTC) that has progressed following prior VEGFR-targeted therapy and who are radioactive iodine (RAI)-refractory or ineligible. This is the first approval for patients with RAI-refractory locally advanced or metastatic DTC who have progressed following prior therapy and the first approval in pediatric patients with DTC. The approval was based on data from COSMIC-311 (Study XL184-311, NCT03690388), an international, randomized, double-blind trial in which patients with locally advanced or metastatic RAI-refractory DTC that progressed during or following treatment with at least one VEGFR-targeting tyrosine kinase inhibitor (TKI) were treated with either cabozantinib 60 mg orally once daily (N=170) or placebo with best supportive care (N=88). The primary efficacy outcome measures were progression-free survival (PFS) and overall response rate (ORR) by blinded independent central review (BICR) per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria. The median PFS was 11.0 months (95% CI: 7.4, 13.8) in the cabozantinib arm compared to 1.9 months (95% CI 1.9, 3.7) in the control arm, with a hazard ratio (HR) of 0.22 (95% CI: 0.15, 0.31). The endpoint of ORR was not met. No new safety signals were identified with the exception of hypocalcemia which was added as a warning in the product labeling.
- Published
- 2022
29. Use of Non-concurrent Common Control in Master Protocols in Oncology Trials: Report of an American Statistical Association Biopharmaceutical Section Open Forum Discussion
- Author
-
Qi Jiang, Rajeshwari Sridhara, Thomas E. Gwise, Martin Posch, Cindy Lu, Richard Simon, Andrew Raven, Mary W. Redman, Lorenzo Hess, Yuan Li Shen, Kit C.B. Roes, R. Pazdur, Olga Marchenko, Yuan Ji, Scott Berry, Marc R. Theoret, and Khadija Rantell
- Subjects
Statistics and Probability ,FOS: Computer and information sciences ,medicine.medical_specialty ,Biopharmaceutical ,Political science ,62P10 ,Section (typography) ,medicine ,Pharmaceutical Science ,Medical physics ,Applications (stat.AP) ,Statistics - Applications - Abstract
This article summarizes the discussions from the American Statistical Association (ASA) Biopharmaceutical (BIOP) Section Open Forum that took place on December 10, 2020 and was organized by the ASA BIOP Statistical Methods in Oncology Scientific Working Group, in coordination with the U.S. FDA Oncology Center of Excellence. Diverse stakeholders including experts from international regulatory agencies, academicians, and representatives of the pharmaceutical industry engaged in a discussion on the use of nonconcurrent control in Master Protocols for oncology trials. While the use of nonconcurrent control with the concurrent control may increase the power of detecting the therapeutic difference between a treatment and the control, the panelists had diverse opinion on the statistical approaches for modeling nonconcurrent and concurrent controls. Some were more concerned about the temporality of the nonconcurrent control and bias introduced by different confounders related to time, for example, changes in standard of care, changes in patient population, changes in recruiting strategies, changes in assessment of endpoints. Nevertheless, in some situations such as when the recruitment is extremely challenging for a rare disease, the panelists concluded that the use of a nonconcurrent control can be justified.
- Published
- 2022
30. FDA Approval Summary: Mobocertinib for Metastatic Non-Small Cell Lung Cancer with EGFR Exon 20 Insertion Mutations
- Author
-
Elizabeth S. Duke, Liza Stapleford, Nicole Drezner, Anup K. Amatya, Pallavi S. Mishra-Kalyani, Yuan-Li Shen, Kimberly Maxfield, Jeanne Fourie Zirkelbach, Youwei Bi, Jiang Liu, Xinyuan Zhang, Hezhen Wang, Yuching Yang, Nan Zheng, Kelie Reece, Emily Wearne, Jacqueline J. Glen, Idara Ojofeitimi, Barbara Scepura, Abhilasha Nair, Rama Kamesh Bikkavilli, Soma Ghosh, Reena Philip, Richard Pazdur, Julia A. Beaver, Harpreet Singh, and Martha Donoghue
- Subjects
Cancer Research ,Oncology - Abstract
On September 15, 2021, the FDA granted accelerated approval to mobocertinib (Exkivity, Takeda Pharmaceuticals USA, Inc.) for the treatment of adult patients with locally advanced or metastatic non–small cell lung cancer (NSCLC) with EGFR exon 20 insertion mutations, as detected by an FDA-approved test, whose disease has progressed on or after platinum-based chemotherapy. The approval was based on data from Study AP32788–15–101 (NCT02716116), an international, non-randomized, multi-cohort clinical trial that included patients with locally advanced or metastatic NSCLC with EGFR exon 20 insertion mutations. The overall response rate in 114 patients whose disease had progressed on or after platinum-based chemotherapy was 28% [95% confidence interval (CI), 20%–37%] with a median duration of response of 17.5 months (95% CI, 7.4–20.3). The most common adverse reactions (>20%) were diarrhea, rash, nausea, stomatitis, vomiting, decreased appetite, paronychia, fatigue, dry skin, and musculoskeletal pain. Product labeling includes a Boxed Warning for QTc prolongation and torsades de pointes. This is the first approval of an oral targeted therapy for patients with advanced EGFR exon 20 insertion mutation–positive NSCLC.
- Published
- 2022
31. FDA Approval Summary: Ivosidenib for the treatment of patients with advanced unresectable or metastatic, chemotherapy refractory cholangiocarcinoma with an IDH1 mutation
- Author
-
Sandra J. Casak, Shan Pradhan, Lola A. Fashoyin-Aje, Yi Ren, Yuan-Li Shen, Yuan Xu, Edwin Chiu Yuen Chow, Ye Xiong, Jeanne Fourie Zirklelbach, Jiang Liu, Rosane Charlab, William F. Pierce, Nataliya Fesenko, Julia A. Beaver, Richard Pazdur, Paul G. Kluetz, and Steven J. Lemery
- Subjects
Adult ,Cancer Research ,Pyridines ,United States Food and Drug Administration ,Vomiting ,Glycine ,Nausea ,Article ,Isocitrate Dehydrogenase ,United States ,Abdominal Pain ,Cholangiocarcinoma ,Bile Ducts, Intrahepatic ,Oncology ,Bile Duct Neoplasms ,Double-Blind Method ,Asthenia ,Mutation ,Disease Progression ,Humans ,Drug Approval ,Fatigue - Abstract
On August 25, 2021, the FDA approved ivosidenib for the treatment of adult patients with unresectable locally advanced or metastatic hepatocellular isocitrate dehydrogenase 1 (IDH1) mutated cholangiocarcinoma (CCA) as detected by an FDA-approved test with disease progression after 1 to 2 prior lines of systemic therapy for advanced disease. The approval was based on data from Study AG120-C-005 (ClarIDHy), a double-blind placebo-controlled trial that randomly allocated (2:1) patients to receive either ivosidenib or placebo. Independently assessed progression-free survival (PFS) was the primary endpoint. With a median follow-up of 6.9 months, the HR for PFS was 0.37 [95% confidence interval (CI), 0.25–0.54; P < 0.0001). Overall survival (OS) was the key secondary endpoint. At the final analysis of OS, with 70.5% of patients in the placebo arm receiving ivosidenib post disease progression, a non–statistically significant improvement in the ivosidenib arm with an HR = 0.79 (95% CI, 0.56–1.12) and median OS of 10.3 months (95% CI, 7.8–12.4) and 7.5 months (95% CI, 4.8–11.1) in the ivosidenib and placebo arms, respectively, were reported. Adverse reactions occurring in >20% of patients receiving ivosidenib were fatigue/asthenia, nausea, diarrhea, abdominal pain, ascites, vomiting, cough, and decreased appetite. Adverse reactions occurring in >20% of patients receiving placebo were fatigue/asthenia, nausea, abdominal pain, and vomiting. This is the first approval for the subset of patients with CCA harboring an IDH1 mutation.
- Published
- 2022
32. FDA Approval Summary: Selumetinib for Plexiform Neurofibroma
- Author
-
Arup Kumar Sinha, Olen Stephens, Marc R. Theoret, Mei Ou, R. Pazdur, Gregory H. Reaman, Katherine Windsor, Yuching Yang, Suzanne Demko, Denise Casey, Kirsten B. Goldberg, Harpreet Singh, Abhilasha Nair, Jianghong Fan, Yuan Li Shen, Ruby Leong, Whitney S. Helms, M. Anwar Goheer, Byeongtaek Oh, Jiang Liu, Vishal Bhatnagar, Sachia Khasar, Selena R. Daniels, Stacy Shifflett Shord, Sharon Sickafuse, Lili Pan, Yuan Xu, and Pallavi Mishra-Kalyani
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Adolescent ,03 medical and health sciences ,0302 clinical medicine ,Plexiform neurofibroma ,Multicenter trial ,Internal medicine ,Humans ,Medicine ,Neurofibromatosis ,Child ,Schwannomatosis ,Drug Approval ,Neurofibroma, Plexiform ,business.industry ,Cancer ,medicine.disease ,United States ,Confidence interval ,Clinical trial ,030104 developmental biology ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Selumetinib ,Benzimidazoles ,Female ,business - Abstract
On April 10, 2020, the FDA approved selumetinib (KOSELUGO, AstraZeneca) for the treatment of pediatric patients 2 years of age and older with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas. Approval was based on demonstration of a durable overall response rate per Response Evaluation in Neurofibromatosis and Schwannomatosis criteria and supported by observed clinical improvements in plexiform neurofibroma–related symptoms and functional impairments in 50 pediatric patients with inoperable plexiform neurofibromas in a single-arm, multicenter trial. The overall reponse rate per NCI investigator assessment was 66% (95% confidence interval, 51–79) with at least 12 months of follow-up. The median duration of response was not reached, and 82% of responding patients experienced duration of response ≥12 months. Clinical outcome assessment endpoints provided supportive efficacy data. Risks of selumetinib are consistent with MAPK (MEK) inhibitor class effects, including ocular, cardiac, musculoskeletal, gastrointestinal, and dermatologic toxicities. Safety was assessed across a pooled database of 74 pediatric patients with plexiform neurofibromas and supported by adult and pediatric selumetinib clinical trial data in cancer indications. The benefit–risk assessment for selumetinib in patients with inoperable plexiform neurofibromas was considered favorable.
- Published
- 2021
33. Patient- and Clinician-Reported Performance Status in Patients with Multiple Myeloma Treated in the United States Using Real World Data
- Author
-
Vishal Bhatnagar, Bindu Kanapuru, Laura Fernandes, Ting-Yu Chen, Mallorie H. Fiero, Erica Horodniceanu, Nicole Gormley, Catherine Lerro, Fatima Rizvi, Yuan-Li Shen, Felice Yang, Anna Barcellos, Andrew J. Belli, Eric Hansen, Ching-Kun Wang, Donna Rivera, and Paul Kluetz
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
34. FDA Approval Summary: Ivosidenib for Relapsed or Refractory Acute Myeloid Leukemia with an Isocitrate Dehydrogenase-1 Mutation
- Author
-
Rosane Charlab, Vicky Hsu, Kelly J. Norsworthy, Ramadevi Gudi, Ann T. Farrell, Gene M. Williams, Kirsten B. Goldberg, Lola Luo, Albert Deisseroth, Sarah E. Dorff, Donna Przepiorka, Richard Pazdur, Yuan Li Shen, and Christopher M. Sheth
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Pyridines ,Glycine ,Antineoplastic Agents ,QT interval ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Mucositis ,Humans ,Leukocytosis ,Enzyme Inhibitors ,Drug Approval ,United States Food and Drug Administration ,business.industry ,Myeloid leukemia ,medicine.disease ,Rash ,Isocitrate Dehydrogenase ,United States ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Isocitrate dehydrogenase ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,medicine.symptom ,business - Abstract
The FDA approved ivosidenib (Tibsovo; Agios), a small-molecule inhibitor of isocitrate dehydrogenase (IDH)1 on July 20, 2018, for treatment of adults with relapsed or refractory acute myeloid leukemia (R/R AML) with susceptible IDH1 mutation as detected by an FDA-approved test. The efficacy of ivosidenib was established on the basis of complete remission (CR) + CR with partial hematologic recovery (CRh) rate, duration of CR + CRh, and conversion from transfusion dependence (TD) to transfusion independence (TI) in Study AG120-C-001, a single-arm trial. With median follow-up of 8.3 months for 174 adults with IDH1-mutated R/R AML treated with 500 mg ivosidenib daily, the CR + CRh rate was 33% [95% confidence interval (CI), 26–40], median duration of response was 8.2 (95% CI, 5.6–12) months, and conversion from TD to TI occurred in 37% of patients. These endpoints reflect short-term benefit in patients with an unmet medical need; long-term efficacy outcomes were not assessed. Serious adverse reactions (AR) in ≥5% of patients were differentiation syndrome (10%), leukocytosis (10%), and QT interval prolongation (7%). Common (≥20%) ARs of any grade were fatigue, leukocytosis, arthralgia, diarrhea, dyspnea, edema, nausea, mucositis, QT interval prolongation, rash, pyrexia, cough, and constipation. Assessment of long-term safety of ivosidenib is a condition of this approval.
- Published
- 2019
35. FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease
- Author
-
Rajeshwari Sridhara, Qing Xu, Reena Philip, Yuan Li Shen, Aaron J. Schetter, Albert Deisseroth, Donna Przepiorka, Richard Pazdur, Donna Roscoe, Emily Y. Jen, and Ann T. Farrell
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Adolescent ,Gastroenterology ,law.invention ,Young Adult ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Randomized controlled trial ,Refractory ,law ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Antibodies, Bispecific ,medicine ,Humans ,Drug Approval ,B cell ,Aged ,Aged, 80 and over ,business.industry ,Remission Induction ,Middle Aged ,Prognosis ,medicine.disease ,Combined Modality Therapy ,Minimal residual disease ,Confidence interval ,Transplantation ,Cytokine release syndrome ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Female ,Blinatumomab ,business ,medicine.drug - Abstract
On March 29, 2018, the FDA granted accelerated approval for blinatumomab (Blincyto; Amgen, Inc.) for the treatment of adults and children with B-cell precursor acute lymphoblastic leukemia (BCP ALL) in first or second complete remission with minimal residual disease (MRD) greater than or equal to 0.1%. Blinatumomab is a CD3xCD19 bispecific antibody approved previously for the treatment of relapsed or refractory BCP ALL. The basis for this accelerated approval was a single-arm trial. For the 86 patients in first and second complete remission with MRD ≥ 0.1%, conversion to MRD < 0.01% was achieved after one cycle of blinatumomab by 85.2% [95% confidence interval (CI): 73.8%, 93.0%] and 72.0% (95% CI: 50.6%, 87.9%), respectively, and the estimated median hematologic relapse-free survivals (RFS) were 35.2 months (95% CI: 0.4–53.5) and 12.3 months (95% CI: 0.7–42.3), respectively. Hematologic RFS was considered substantial independent of whether patients underwent subsequent allogeneic stem cell transplantation. The safety profile for blinatumomab was established in prior studies, and no new safety signals were observed in the new population. Cytokine release syndrome and neurotoxicity remain significant risks. The FDA is requiring confirmation of clinical benefit in a randomized trial.
- Published
- 2019
36. FDA Approval Summary: Nivolumab in Combination with Ipilimumab for the Treatment of Unresectable Malignant Pleural Mesothelioma
- Author
-
Somak Chatterjee, Martha Donoghue, Pallavi S. Mishra-Kalyani, Harpreet Singh, Erin Larkins, Hong Zhao, Missiratch Biable, Youwei Bi, Julia A. Beaver, Jiang Liu, Hisham Qosa, Richard Pazdur, Erica C Nakajima, Paz J. Vellanki, Lauren Tesh Hotaki, and Yuan Li Shen
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Pleural Neoplasms ,Ipilimumab ,Article ,law.invention ,chemistry.chemical_compound ,law ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Drug Approval ,Aged ,Randomized Controlled Trials as Topic ,Cisplatin ,Chemotherapy ,Clinical pharmacology ,Pleural mesothelioma ,business.industry ,Mesothelioma, Malignant ,Carboplatin ,Pemetrexed ,Nivolumab ,Treatment Outcome ,chemistry ,Female ,business ,medicine.drug - Abstract
On October 2, 2020, FDA approved nivolumab with ipilimumab as first-line treatment for adult patients with unresectable malignant pleural mesothelioma (MPM). The approval was based on results from Study CA209743 (CHECKMATE-743), an open-label trial of patients with MPM randomized to receive nivolumab and ipilimumab for up to 2 years (n = 303) or six cycles of chemotherapy with cisplatin or carboplatin plus pemetrexed (n = 302). Overall survival (OS) was improved for patients who received nivolumab and ipilimumab, with a median OS of 18.1 months [95% confidence interval (CI), 16.8–21.5] compared with 14.1 months (95% CI: 12.5–16.2; HR, 0.74; 95% CI, 0.61–0.89; P = 0.002), for patients who received chemotherapy. The magnitude of benefit was larger for patients with non-epithelioid versus epithelioid histology. Additional clinical pharmacology data support an alternative dosing regimen of nivolumab than evaluated in the trial, which will reduce the number of required treatment visits. This application was reviewed under FDA's Project Orbis, in collaboration with Australia's Therapeutic Goods Administration, Switzerland's Swissmedic, Health Canada, and Brazil's National Health Surveillance Agency or ANVISA (Agência Nacional de Vigilância Sanitária). Nivolumab and ipilimumab is the first drug regimen approved by FDA for MPM since 2004.
- Published
- 2021
37. FDA Approval Summary: Pembrolizumab for the First-line Treatment of Patients with MSI-H/dMMR Advanced Unresectable or Metastatic Colorectal Carcinoma
- Author
-
Kirsten B. Goldberg, Lola Fashoyin-Aje, William F. Pierce, Marc R. Theoret, Sirisha Mushti, Paul G. Kluetz, Richard Pazdur, Yuan Li Shen, Leah Her, Leigh Marcus, Sandra Casak, Steven Lemery, and Joyce Cheng
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Abdominal pain ,Constipation ,Colorectal cancer ,Nausea ,medicine.medical_treatment ,Pembrolizumab ,Neutropenia ,Antibodies, Monoclonal, Humanized ,DNA Mismatch Repair ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Internal medicine ,medicine ,Humans ,Drug Approval ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Chemotherapy ,business.industry ,Middle Aged ,medicine.disease ,United States ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Vomiting ,Female ,Microsatellite Instability ,medicine.symptom ,business ,Colorectal Neoplasms - Abstract
The FDA approved pembrolizumab on June 29, 2020, for the treatment of patients with unresectable or metastatic microsatellite instability-high (MSI-H) colorectal cancer with no prior systemic treatment for advanced disease. The approval was based on data from Study Keynote-177, which randomly allocated patients to receive either pembrolizumab or standard of care (SOC) with chemotherapy. Overall survival (OS) and independently assessed progression-free survival (PFS) were the primary endpoints. At the time of the final PFS analysis and second prespecified interim OS analysis, the estimated median PFS was 16.5 months (95% CI: 5.4–32.4) versus 8.2 months (95% CI: 6.1–10.2) in the pembrolizumab and SOC arms, respectively [HR: 0.60 (95% CI: 0.45–0.80); two-sided P = 0.0004]. FDA assessed unblinded OS data during the review of the application and identified no safety concerns that would preclude approval of this supplement. Adverse reactions occurring in >30% of patients receiving pembrolizumab were diarrhea, fatigue/asthenia, and nausea. Adverse reactions occurring in >30% of patients receiving SOC were diarrhea, nausea, fatigue/asthenia, neutropenia, decreased appetite, peripheral neuropathy (high-level term), vomiting, abdominal pain, constipation, and stomatitis. Duration of treatment in the pembrolizumab arm was almost double (median 11.1 months, range 0–30.6 months) than the duration of treatment in patients receiving SOC (median, 5.7 months). Approval of pembrolizumab is likely to change the treatment paradigm for first-line treatment with MSI-H advanced colorectal cancer given the study results and different safety profile.
- Published
- 2021
38. FDA Approval Summary: Atezolizumab Plus Bevacizumab for the Treatment of Patients with Advanced Unresectable or Metastatic Hepatocellular Carcinoma
- Author
-
Hong Zhao, Lisa Rodriguez, Kirsten B. Goldberg, Lola Fashoyin-Aje, Jiang Liu, Yuan Xu, Richard Pazdur, Yuan Li Shen, Sandra Casak, Martha Donoghue, Marc R. Theoret, Xiling Jiang, Shubhangi Mehta, Steven Lemery, Xiaoping Jiang, Paul G. Kluetz, and William F. Pierce
- Subjects
0301 basic medicine ,Sorafenib ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Bevacizumab ,Population ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Neoplasm Metastasis ,education ,neoplasms ,Drug Approval ,Aged ,Aged, 80 and over ,education.field_of_study ,Proteinuria ,business.industry ,Liver Neoplasms ,Middle Aged ,Confidence interval ,Diarrhea ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Drug and Narcotic Control ,Female ,medicine.symptom ,business ,Varices ,medicine.drug - Abstract
On May 29, 2020, the FDA approved atezolizumab for use in combination with bevacizumab, for the treatment of adult patients with unresectable locally advanced or metastatic hepatocellular carcinoma (HCC) with no prior systemic treatment. The approval was based on data from Study IMbrave150, which randomly allocated (2:1) patients to receive either atezolizumab plus bevacizumab (atezolizumab-bevacizumab) or sorafenib. Overall survival (OS) and independently assessed progression-free survival (PFS) in the intent-to-treat population were the primary endpoints. At the time of the primary analysis, the estimated median OS could not be estimated in the atezolizumab-bevacizumab arm and was 13.2 months in the sorafenib arm [HR, 0.58; 95% confidence interval (CI), 0.42–0.79]. The estimated median PFS was 6.8 months (95% CI, 5.8–8.3) and 4.3 months (95% CI, 4.0–5.6) in the atezolizumab-bevacizumab and sorafenib arms, respectively. Adverse reactions occurring in >20% of patients receiving atezolizumab-bevacizumab were hypertension, fatigue/asthenia, and proteinuria. Adverse reactions occurring in >20% of patients receiving sorafenib were palmar-plantar erythrodysesthesia, diarrhea, hypertension, and decreased appetite. Hemorrhage was reported more frequently in patients receiving atezolizumab-bevacizumab (25%) than in patients receiving sorafenib (17%). An evaluation for the presence of varices is recommended within 6 months of initiation of atezolizumab-bevacizumab in patients with HCC. Approval of atezolizumab-bevacizumab is likely to change the treatment paradigm for HCC, given that treatment with atezolizumab-bevacizumab resulted in improved OS and PFS compared with sorafenib, an accepted standard of care for first-line treatment of patients with unresectable HCC. See related commentary by Castet et al., p. 1827
- Published
- 2020
39. FDA Approval Summary: Vemurafenib for the Treatment of Patients with Erdheim‐Chester Disease with the BRAFV600 Mutation
- Author
-
Lola Luo, Yuan Li Shen, Kirsten B. Goldberg, Sriram Subramaniam, Amy E. McKee, Ann T. Farrell, Virginia E. Kwitkowski, Edvardas Kaminskas, Stacy S. Shord, Patricia A. Oneal, and Richard Pazdur
- Subjects
0301 basic medicine ,Adult ,Male ,Proto-Oncogene Proteins B-raf ,Cancer Research ,medicine.medical_specialty ,Erdheim-Chester Disease ,genetic structures ,Zelboraf ,Antineoplastic Agents ,Disease ,QT interval ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Vemurafenib ,health care economics and organizations ,Regulatory Issues: FDA ,Aged ,business.industry ,United States Food and Drug Administration ,Middle Aged ,medicine.disease ,Rash ,BRAFV600 mutations ,United States ,Histiocytosis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Erdheim–Chester disease ,Cohort ,Mutation ,Non‐Langerhans cell histocytosis ,Female ,medicine.symptom ,business ,medicine.drug ,Cohort study - Abstract
The FDA has granted regular approval to vemurafenib for the treatment of adult patients with Erdheim‐Chester Disease (ECD) with BRAFV600 mutation. This article describes the FDA review of the evidence and the clinical implications for this rare patient population., On November 6, 2017, the U.S. Food and Drug Administration (FDA) granted regular approval to vemurafenib for the treatment of adult patients with Erdheim‐Chester disease (ECD) with BRAFV600 mutation. ECD is a type of histiocytosis, a rare disorder characterized by an abnormal accumulation and behavior of cells of the mononuclear phagocytic system, which includes antigen‐processing cells, dendritic cells, monocytes, or macrophages. Recently published data confirm a frequency of 54% of BRAFV600E mutations in patients with ECD. Approval was based on a cohort of 22 patients who received 960 mg of vemurafenib twice daily within the VE Basket Trial (MO28072), a single‐arm, multicenter, multiple cohort study. Patients in the ECD cohort had histologically confirmed ECD with BRAFV600 mutations that were refractory to standard therapy. The ECD cohort achieved an overall response rate of 54.5% (95% confidence interval: 32.2–75.6), with a complete response rate of 4.5%. With a median duration of follow‐up of 26.6 months, the median duration of response has not been reached. The most frequently reported adverse reactions (>50%) in the ECD cohort were arthralgia, rash maculo‐papular, alopecia, fatigue, electrocardiogram QT interval prolonged, and skin papilloma. The median treatment duration for ECD patients in this study was 14.2 months. This article describes the FDA review of the vemurafenib efficacy supplement for patients with ECD with BRAFV600 mutations. Implications for Practice. Vemurafenib, an oral monotherapy targeting a mutation in BRAF, is the first U.S. Food and Drug Administration approval for the treatment of Erdheim‐Chester disease (ECD). ECD is an extremely rare hematopoietic neoplasm that represents clonal proliferation of myeloid progenitor cells. ECD may involve bone and one or more organ systems, primarily affecting adults in their 5th and 7th decades of life, with a slight male predominance. This approval provides an effective and reasonably safe therapy for patients with a serious and life‐threatening condition for which no approved therapy exists.
- Published
- 2018
40. FDA Approval Summary: Daratumumab for Treatment of Multiple Myeloma After One Prior Therapy
- Author
-
Lola Luo, Sriram Subramaniam, Nicole J. Gormley, Guoxiang Shen, Lian Ma, Rajeshwari Sridhara, Yuan Li Shen, Richard Pazdur, Stacy S. Shord, Kirsten B. Goldberg, Ann T. Farrell, Vishal Bhatnagar, and Amy E. McKee
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Peripheral edema ,Dexamethasone ,Bortezomib ,0302 clinical medicine ,Multiple myeloma ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Drug Approval ,Lenalidomide ,health care economics and organizations ,Regulatory Issues: FDA ,Aged, 80 and over ,Hazard ratio ,Antibodies, Monoclonal ,Middle Aged ,Thalidomide ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,medicine.symptom ,medicine.drug ,Adult ,medicine.medical_specialty ,Neutropenia ,Disease-Free Survival ,03 medical and health sciences ,Daratumumab ,Internal medicine ,medicine ,Humans ,Aged ,business.industry ,medicine.disease ,United States ,030104 developmental biology ,Drug Resistance, Neoplasm ,business - Abstract
Multiple myeloma is mostly an incurable disease. The U.S. Food and Drug Administration (FDA) granted daratumumab accelerated approval in November 2015 as monotherapy for patients with multiple myeloma who have received at least three prior lines of therapy, including a proteasome inhibitor and an immunomodulatory agent, or who are double refractory to a proteasome and an immunomodulatory agent. This article describes the FDA review of the strength of evidence for this application and its clinical implications for the multiple myeloma population., On November 21, 2016, the U.S. Food and Drug Administration granted regular approval to daratumumab in combination with lenalidomide and dexamethasone, or bortezomib and dexamethasone, for the treatment of patients with multiple myeloma who have received at least one prior therapy. Approval was based on two randomized, open‐label trials in which daratumumab was added to these backbone therapies. The MMY3003 trial demonstrated substantial improvement in progression‐free survival (PFS) when daratumumab was added to lenalidomide and dexamethasone compared with lenalidomide and dexamethasone alone. The estimated median PFS had not been reached in the daratumumab arm and was 18.4 months in the control arm (hazard ratio [HR] = 0.37; 95% confidence interval [CI]: 0.27–0.52; p
- Published
- 2017
41. FDA Approval Summary: Tagraxofusp-erzs For Treatment of Blastic Plasmacytoid Dendritic Cell Neoplasm
- Author
-
Steven Bowen, Natalie Simpson, Christopher M. Sheth, Xin Gao, Liang Li, Luning Zhuang, Ann T. Farrell, Ruby Leong, Donna Przepiorka, Baikuntha Aryal, Rong Wang, Emily Y. Jen, Kirsten B. Goldberg, Yuan Li Shen, Richard Pazdur, Gideon M. Blumenthal, and Chao Liu
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Nausea ,Recombinant Fusion Proteins ,Peripheral edema ,Interleukin-3 Receptor alpha Subunit ,Plasmacytoid dendritic cell ,Gastroenterology ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Drug Approval ,Aged ,Response rate (survey) ,Aged, 80 and over ,Clinical Trials as Topic ,biology ,business.industry ,Dendritic Cells ,Middle Aged ,United States ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Hematologic Neoplasms ,Toxicity ,Cohort ,biology.protein ,Female ,Patient Safety ,medicine.symptom ,Antibody ,business ,Plasmacytoma - Abstract
Tagraxofusp-erzs (Elzonris, Stemline) is a cytotoxin that targets CD123-expressing cells. On December 21, 2018, FDA approved tagraxofusp-erzs for the treatment of blastic plasmacytoid dendritic cell neoplasms (BPDCN) in adult and pediatric patients 2 years and older. Approval was based on the response rate in a single-arm trial, Study STML-401-0114; the pivotal cohort included 13 patients with treatment-naïve BPDCN who received tagraxofusp-erzs monotherapy. The complete response or clinical complete response (CR/CRc) rate in the pivotal cohort was 54% (95% CI: 25%–81%), and the median duration of CR/CRc was not reached with a median follow-up of 11.5 months (range: 0.2–12.7). In a separate exploratory cohort, a CR/CRc was achieved by 2 (13%) patients with R/R BPDCN. Safety was assessed in 94 patients with myeloid neoplasms treated with tagraxofusp-erzs at the approved dose and schedule. The major toxicity was capillary leak syndrome (CLS), which occurred in 55% of patients and was fatal in 2%. Hepatotoxicity and hypersensitivity reactions were reported in 88% and 46% of patients, respectively. Other common (≥30%) adverse reactions were nausea, fatigue, peripheral edema, pyrexia, and weight increase. A high proportion of patients (85%) developed neutralizing antidrug antibodies. Tagraxofusp-erzs is the first FDA-approved treatment for BPDCN.
- Published
- 2019
42. FDA Approval Summary: Glasdegib for Newly Diagnosed Acute Myeloid Leukemia
- Author
-
Christopher M. Sheth, Sriram Subramaniam, Ruby Leong, Kunthel By, Kirsten B. Goldberg, Yuan Li Shen, Kelly J. Norsworthy, Luning Zhuang, Donna Przepiorka, Ann T. Farrell, Pedro L. Del Valle, Chao Liu, and Richard Pazdur
- Subjects
0301 basic medicine ,Male ,Cancer Research ,medicine.medical_specialty ,Boxed warning ,Antineoplastic Agents ,Comorbidity ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Mucositis ,Humans ,Drug Approval ,Aged ,Drug Labeling ,Aged, 80 and over ,Dose-Response Relationship, Drug ,business.industry ,United States Food and Drug Administration ,Phenylurea Compounds ,Age Factors ,Cytarabine ,Induction chemotherapy ,Induction Chemotherapy ,Middle Aged ,medicine.disease ,Rash ,Smoothened Receptor ,Survival Analysis ,United States ,Dysgeusia ,Clinical trial ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Benzimidazoles ,Female ,medicine.symptom ,business ,Febrile neutropenia ,medicine.drug - Abstract
On November 21, 2018, the FDA approved glasdegib (Daurismo; Pfizer), a small-molecule Hedgehog inhibitor, in combination with low-dose cytarabine (LDAC) for treatment of newly diagnosed acute myeloid leukemia (AML) in adults ≥ 75 years or with comorbidities that preclude use of intensive induction chemotherapy. Evidence of clinical benefit came from Study BRIGHT AML 1003, a randomized trial comparing glasdegib+LDAC with LDAC alone for treatment of newly diagnosed AML in 115 patients either ≥ 75 years old or ≥ 55 years old with preexisting comorbidities. Efficacy was established by improved overall survival (OS) with the combination compared with LDAC alone (HR, 0.46; 95% confidence interval, 0.30–0.71; one-sided stratified log-rank P = 0.0002). Median OS was 8.3 months with the combination and 4.3 months with LDAC alone. Common adverse reactions included cytopenias, fatigue, hemorrhage, febrile neutropenia, musculoskeletal pain, nausea, edema, dyspnea, decreased appetite, dysgeusia, mucositis, constipation, and rash. The label includes a boxed warning for embryo-fetal toxicity and a warning for QT interval prolongation. There is a limitation of use for patients with moderate-to-severe hepatic and severe renal impairment; trials studying glasdegib in these patient populations are required as a condition of this approval. See related commentary by Fathi, p. 6015
- Published
- 2019
43. A pooled analysis of response to selective RET inhibitors among patients with medullary thyroid cancer with M918T versus non-M918T RET mutations
- Author
-
Janice Kim, Somak Chatterjee, Diana Bradford, Yuan-Li Shen, Julia A. Beaver, Pallavi S. Mishra-Kalyani, and Harpreet Singh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,business.industry ,Thyroid ,Medullary thyroid cancer ,medicine.disease ,Pooled analysis ,medicine.anatomical_structure ,Internal medicine ,medicine ,business - Abstract
6078 Background: Medullary thyroid cancer (MTC) accounts for 1 to 2% of thyroid cancers in the United States; RET alterations occur in >95% of hereditary and 50% of sporadic forms. Up to 80% of patients with sporadic MTC have somatic M918T RET mutations, which is associated with poor prognosis (1). The tyrosine kinase inhibitors (TKIs) cabozantinib and vandetanib are approved to treat patients with MTC regardless of RET status; however, retrospective analyses have suggested that there may be greater benefit in patients with M918T mutations (1,2). Newly approved therapies selpercatinib and pralsetinib, developed for patients with RET mutations, have demonstrated higher response rates than previous first line therapies. In this analysis, we examine the differences in overall response rate (ORR) between patients with MTC with RET M918T non- RET M918T mutations. Methods: An analysis of ORR in patients with MTC with RET M918T mutations with non-M918T mutations was conducted using the efficacy populations used to support the approvals of pralsetinib and selpercatinib using the following groups: Patients who received prior cabozantinib or vandetanib (referred to as “previously treated”). Patients with no prior cabozantinib or vandetanib (“TKI naïve”). All patients regardless of prior therapy. Results: Exploratory analysis of ORR of pooled population of Selpercatinib and Pralsetinib in patients with MTC with RET M918T mutations and non-M918T mutations. 1 Prior vandetanib or cabozantinib. 2 No prior vandetanib or cabozantinib. Two groups of patients were analyzed ( RET M918T mutation and RET non-M918T mutation), with subgroups with respect to prior treatment. Among all patients regardless of prior therapy, the ORR was similar between M918T non-M918T groups. Among previously treated patients, the ORR was lower in the M918T group vs. the non-M918T group, while in the TKI naïve group the ORR was higher in the M918T groups vs the non-M918T group although the 95% CIs overlap in both comparisons. Conclusions: There were no major differences in ORR among mutational subtypes in patients with MTC treated with RET inhibitors, regardless of prior therapy. ORR was similar between patients with M918T and non-M918T mutations. Additional experience in ongoing clinical studies may provide additional data regarding responses across specific mutation types. References: 1.Sherman SI et al “Correlative analyses of RET and RAS mutations in a phase 3 trial of cabozantinib...” Cancer. 2016;122(24):3856-3864. 2.Wells SA Jr et al “Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer...” J Clin Oncol. 2012;30(2):134-41.[Table: see text]
- Published
- 2021
44. An FDA analysis of the association of tumor growth rate and overall and progression-free survival in metastatic non-small cell lung cancer (NSCLC) patients
- Author
-
Dickran Kazandjian, Jeremy Mason, Elaine Chang, Marc R. Theoret, Richard Pazdur, Yutao Gong, Shenghui Tang, Yuan-Li Shen, Gideon M. Blumenthal, Harpreet Singh, and Julia A. Beaver
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,non-small cell lung cancer (NSCLC) ,medicine.disease ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Antigen ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Overall survival ,Tumor growth ,Progression-free survival ,business ,030215 immunology - Abstract
9541 Background: Previous studies have suggested that tumor growth rate (g), estimated using prostate-specific antigen values, is associated with overall survival (OS) in prostate cancer (Wilkerson, 2016). We performed a retrospective pooled analysis in non-small cell lung cancer (NSCLC) to investigate the extent to which g values estimated using radiological tumor measurements in clinical trials are associated with survival. Methods: We identified 24 randomized clinical trials submitted to FDA between 2013 and 2019 investigating either immune checkpoint inhibitor (ICI) or targeted therapy (TT) in pts with metastatic NSCLC. Of 9934 patients (pts) enrolled, 5532 pts (2401, 1189, and 1942 pts treated with ICI, TT, and chemotherapy respectively) had sufficient data to derive a valid g. The g was evaluated by both type and line of therapy. Pts were then grouped according to quartiles of g, with Q1 being the lowest. We calculated OS and progression-free survival (PFS) for each group via the Kaplan-Meier method, and used the Cox model for group comparison. Results: Median g was 9.7E-4, 1.4E-3, and 2.2E-3/day, and median OS was 34.2, 21.3, and 15.3 months (mo), in pts treated with TT, ICI, and chemotherapy, respectively, regardless of lines of therapy. When treated with the same type of therapy, pts receiving 2nd line therapy had a higher median g than those receiving 1st line. The median survival and log-rank hazard ratios for pts treated with 1st line ICI monotherapy are shown in the Table. Conclusions: TT is associated with the lowest median g, followed by ICI, and then chemotherapy, perhaps due to patient selection, better inherent biology/natural history, or favorable results of TT on selected tumors. Regardless, we found that g is inversely associated with survival, across treatment types. This relationship is also observed in pts treated with the same type and line of therapy (for example, 1st line ICI), where Q1 has the longest survival, followed by Q2, Q3, and then Q4. In summary, our exploratory analysis suggests that g derived from radiological tumor measurements in NSCLC may relate to survival. Prospective studies are needed to evaluate if g might be an earlier endpoint compared to classical response criteria. [Table: see text]
- Published
- 2020
45. Current Statistical Challenges in Oncology Clinical Trials in the Era of Targeted Therapy
- Author
-
Kun He, Rajeshwari Sridhara, Lei Nie, Yuan-Li Shen, and Shenghui Tang
- Subjects
Statistics and Probability ,Protocol (science) ,medicine.medical_specialty ,Pathology ,business.industry ,medicine.medical_treatment ,Alternative medicine ,Pharmaceutical Science ,Cancer ,Disease ,medicine.disease ,Targeted therapy ,Treatment and control groups ,Clinical trial ,Drug development ,medicine ,Intensive care medicine ,business - Abstract
The mapping of the human genome has led to rapid advances in our understanding of diseases at the molecular level. This new knowledge is shifting the drug development paradigm in cancer trials. For example, traditional Phase 1 to 3 trials of dose finding, evaluating activity, and confirming efficacy are becoming less common with all the three objectives accomplished within one protocol. Molecular marker-based clinical trials are also on the rise in place of the traditional disease-based clinical trials. In addition, patients receiving control treatment are routinely switched to the experimental treatment after disease progression, complicating the final comparison of survival rates between treatment groups. In this article, we describe these and other challenges in the regulatory review of cancer therapies as a result of this paradigm shift and also describe some innovative ideas that have the potential to improve evaluation of new agents to treat cancer in clinical trials.
- Published
- 2015
46. FDA Approval: Belinostat for the Treatment of Patients with Relapsed or Refractory Peripheral T-cell Lymphoma
- Author
-
Xiao Hong Chen, M. Stacey Ricci, Hyon-Zu Lee, Yuan Li Shen, Richard Pazdur, Julie Bullock, Sarah E. Dorff, Rosane Charlab, Haleh Saber, Bahru A. Habtemariam, Robert C. Kane, Robert Justice, Edvardas Kaminskas, Virginia E. Kwitkowski, Erik Bloomquist, Ann T. Farrell, Nitin Mehrotra, Pedro L. Del Valle, and Janice Brown
- Subjects
Cancer Research ,medicine.medical_specialty ,Anemia ,Nausea ,Antineoplastic Agents ,Neutropenia ,Hydroxamic Acids ,Gastroenterology ,chemistry.chemical_compound ,Refractory ,Internal medicine ,medicine ,Humans ,Drug Approval ,Sulfonamides ,United States Food and Drug Administration ,business.industry ,Lymphoma, T-Cell, Peripheral ,medicine.disease ,United States ,Confidence interval ,Lymphoma ,Surgery ,Histone Deacetylase Inhibitors ,Oncology ,chemistry ,Vomiting ,medicine.symptom ,business ,Belinostat - Abstract
On July 3, 2014, the FDA granted accelerated approval for belinostat (Beleodaq; Spectrum Pharmaceuticals, Inc.), a histone deacetylase inhibitor, for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma (PTCL). A single-arm, open-label, multicenter, international trial in the indicated patient population was submitted in support of the application. Belinostat was administered intravenously at a dose of 1000 mg/m2 over 30 minutes once daily on days 1 to 5 of a 21-day cycle. The primary efficacy endpoint was overall response rate (ORR) based on central radiology readings by an independent review committee. The ORR was 25.8% [95% confidence interval (CI), 18.3–34.6] in 120 patients that had confirmed diagnoses of PTCL by the Central Pathology Review Group. The complete and partial response rates were 10.8% (95% CI, 5.9–17.8) and 15.0% (95% CI, 9.1–22.7), respectively. The median duration of response, the key secondary efficacy endpoint, was 8.4 months (95% CI, 4.5–29.4). The most common adverse reactions (>25%) were nausea, fatigue, pyrexia, anemia, and vomiting. Grade 3/4 toxicities (≥5.0%) included anemia, thrombocytopenia, dyspnea, neutropenia, fatigue, and pneumonia. Belinostat is the third drug to receive accelerated approval for the treatment of relapsed or refractory PTCL. Clin Cancer Res; 21(12); 2666–70. ©2015 AACR.
- Published
- 2015
47. FDA Approval Summary: Nivolumab for the Treatment of Relapsed or Progressive Classical Hodgkin Lymphoma
- Author
-
Yuan Li Shen, Ann T. Farrell, Richard Pazdur, Yvette L. Kasamon, Yaping Wang, and R. Angelo de Claro
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Immunoconjugates ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,law.invention ,0302 clinical medicine ,Randomized controlled trial ,law ,Programmed death‐ligand 1 ,Brentuximab vedotin ,Drug Approval ,health care economics and organizations ,Regulatory Issues: FDA ,Brentuximab Vedotin ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,Antibodies, Monoclonal ,Programmed cell death 1 inhibitor ,Rash ,Combined Modality Therapy ,Hodgkin Disease ,Nivolumab ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,medicine.drug ,medicine.medical_specialty ,Breakthrough therapy ,Drug-Related Side Effects and Adverse Reactions ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Pneumonitis ,Hepatitis ,business.industry ,United States Food and Drug Administration ,medicine.disease ,United States ,030104 developmental biology ,business ,Hodgkin lymphoma - Abstract
On May 17, 2016, after an expedited priority review, the U.S. Food and Drug Administration granted accelerated approval for nivolumab for the treatment of patients with classical Hodgkin L that has relapsed or progressed after autologous HSCT and post‐transplantation brentuximab vedotin. This was the first FDA application for a PD‐1 inhibitor in hematologic malignancies. A summary of the FDA review and approval is presented here., On May 17, 2016, after an expedited priority review, the U.S. Food and Drug Administration granted accelerated approval to nivolumab for the treatment of patients with classical Hodgkin lymphoma (cHL) that has relapsed or progressed after autologous hematopoietic stem cell transplantation (HSCT) and post‐transplantation brentuximab vedotin (BV). Nivolumab in cHL had been granted breakthrough therapy designation. Accelerated approval was based on two single‐arm, multicenter trials in adults with cHL. In 95 patients with relapsed or progressive cHL after autologous HSCT and post‐transplantation BV, nivolumab, dosed at 3 mg/kg intravenously every 2 weeks, produced a 65% (95% confidence interval: 55%–75%) objective response rate (58% partial remission, 7% complete remission). The estimated median duration of response was 8.7 months, with 4.6‐month median follow‐up for response duration. The median time to response was 2.1 (range: 0.7–5.7) months. Among 263 patients with cHL treated with nivolumab, 21% reported serious adverse reactions (ARs). The most common all‐grade ARs (reported in ≥20%) were fatigue, upper respiratory tract infection, cough, pyrexia, diarrhea, elevated transaminases, and cytopenias. Infusion‐related reaction and hypothyroidism or thyroiditis occurred in >10% of patients; other immune‐mediated ARs, occurring in 1%–5%, included rash, pneumonitis, hepatitis, hyperthyroidism, and colitis. A new Warning and Precaution was issued for complications of allogeneic HSCT after nivolumab, including severe or hyperacute graft‐versus‐host disease, other immune‐mediated ARs, and transplant‐related mortality. Continued approval for the cHL indication may be contingent upon verification of clinical benefit in a randomized trial. The Oncologist 2017;22:585–591 Implications for Practice. Based on response rate and duration in single‐arm studies, nivolumab is a new treatment option for patients with classical Hodgkin lymphoma (cHL) that has relapsed or progressed despite autologous hematopoietic stem cell transplantation (HSCT) and brentuximab vedotin. This was the first U.S. Food and Drug Administration marketing application for a programmed cell death 1 inhibitor in hematologic malignancies. The use of immune checkpoint blockade in cHL represents a new treatment paradigm. The safety of allogeneic HSCT after nivolumab requires further evaluation, as does the safety of nivolumab after allogeneic HSCT.
- Published
- 2017
48. FDA Analysis of Survival Outcomes in Older Adults with Relapsed-Refractory Multiple Myeloma (RRMM) Treated with Novel Drug Regimens
- Author
-
Ann T. Farrell, Susan Jin, Theresa Carioti, Kunthel By, Bindu Kanapuru, Rajeshwari Sridhara, Nicole J. Gormley, Richard Pazdur, and Yuan-Li Shen
- Subjects
Oncology ,Drug ,medicine.medical_specialty ,business.industry ,media_common.quotation_subject ,Immunology ,Treatment outcome ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Transplantation ,Internal medicine ,Relapsed refractory ,medicine ,business ,Survival analysis ,Multiple myeloma ,media_common - Abstract
Background: Multiple novel therapies have been approved for the treatment of RRMM in recent years, resulting in improvements in progression free survival (PFS) and overall survival (OS). However, clinical trials in MM often enroll only a small proportion of older patients, particularly patients ≥75 years (Kanapuru 2017). Evaluating the impact of novel therapies, especially triplet therapies, in older adults with RRMM from individual clinical trials is challenging due to the small sample size. Furthermore, significant heterogeneity exists among the older adult population with regards to tolerability of anti-myeloma therapy. In newly diagnosed transplant-ineligible patients with MM, evidence from pooled analysis indicates that patients >80 years may be at increased risk for adverse clinical outcomes (Palumbo 2015). We evaluated the prognostic impact of age on survival outcomes in patients with RRMM receiving novel therapies. Methods: Data from 10 clinical trials submitted for approval between 2011-2015 were pooled for this analysis. Participants were grouped according to four age strata: 80 years. PFS and OS were calculated using the Kaplan-Meier method (K-M). Within each age stratum, we conducted a subgroup analysis comparing doublet versus triplet regimens. Cox's proportional hazards regression model was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs), adjusting for gender, race, ISS stage, ECOG status, regimen (only for primary age analysis) and prior transplant. Results: In total, 4766 patients were included in the analysis. Forty-seven percent were 80 years of age. The percentage of patients with baseline ISS stage III and ECOG 2 was higher in the 75-80 years (31.0% and 11.0%) and >80 years group (32.0%, 19.0%) compared to 65-74 years (24.0%, 8.0%) and Adjusted PFS HR (95% CI) for triplet versus doublet regimens was 0.69 (0.60, 0.79), 0.71 (0.61, 0.83), 0.61 (0.46, 0.81), and 0.62 (0.36, 1.05) for 80 years respectively. The HR (95% CI) for OS was 0.70 (0.59, 0.83), 0.86 (0.72, 1.02), 0.55 (0.40, 0.77) and 0.98 (0.56, 1.73). Conclusions: Improvement in PFS with novel therapies, including triplet regimens, appears to extend to older adults including patients >80 years of age. No trend in treatment effect for PFS was observed across the age groups. Overall survival was lower in adults ≥65 years of age compared to patients 80 years of age. Triplet regimens appear to improve survival over doublet regimens; however, a consistent trend across age groups was not observed. The OS results from this analysis must be interpreted with caution due to immature OS data at the time of submission, differential follow-up for individual trials, and small sample size, particularly in patients >80 years of age. Enrolling a representative population of older adults in MM clinical trials is needed to allow for an accurate assessment of outcomes in this population. Furthermore, considering biologic age rather than chronologic age to identify older patients who can benefit from these therapies would serve to further advance treatment in patients with MM. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
49. U.S. Food and Drug Administration Approval: Rituximab in Combination with Fludarabine and Cyclophosphamide for the Treatment of Patients with Chronic Lymphocytic Leukemia
- Author
-
Richard Pazdur, Hong Zhao, Steven Lemery, Vaishali Jarral, Sandra Casak, Yuan Li Shen, Patricia Keegan, Aakanksha Khandelwal, Gina Davis, and Mark D. Rothmann
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cyclophosphamide ,Chronic lymphocytic leukemia ,Food and drug administration ,Antibodies, Monoclonal, Murine-Derived ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Drug Approval ,health care economics and organizations ,Regulatory Issues: FDA ,Vidarabine ,Aged ,Randomized Controlled Trials as Topic ,United States Food and Drug Administration ,business.industry ,Middle Aged ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,United States ,Fludarabine ,Leukemia ,Immunology ,Monoclonal ,Female ,Rituximab ,business ,medicine.drug - Abstract
Learning Objectives After completing this course, the reader will be able to: Compare the survival benefits of rituximab in combination with fludarabine and cyclophosphamide to those of alemtuzumab, bendamustine, and ofatumumab in patients with CLL.Identify CLL patients for whom rituximab in combination with fludarabine and cyclophosphamide may be an appropriate first-line regimen. CME This article is available for continuing medical education credit at CME.TheOncologist.com Purpose. To describe the clinical studies that led to the FDA approval of rituximab in combination with fludarabine and cyclophosphamide (FC) for the treatment of patients with chronic lymphocytic leukemia (CLL). Materials and Methods. The results of two multinational, randomized trials in CLL patients comparing rituximab combined with fludarabine and cyclophosphamide versus FC were reviewed. The primary endpoint of both studies was progression-free survival (PFS). Results. The addition of rituximab to FC decreased the risk of a PFS event by 44% in 817 previously untreated patients and by 24% in 552 previously treated patients. Median survival times could not be estimated. Exploratory analysis in patients older than 70 suggested that there was no benefit to patients when rituximab was added to FC. The safety profile observed in both trials was consistent with the known toxicity profile of rituximab, FC, or CLL. Conclusions. On the basis of the demonstration of clinically meaningful prolongation of PFS, the FDA granted regular approval to rituximab in combination with FC for the treatment of patients with CLL. The magnitude of the treatment effect in patients 70 years and older is uncertain.
- Published
- 2011
50. Response Rate, Event-Free Survival (EFS), and Overall Survival (OS) in Newly-Diagnosed Acute Myeloid Leukemia (AML): U.S. Food and Drug Administration (FDA) Trial-Level and Patient-Level Analyses
- Author
-
Yutao Gong, Jiaxi Zhou, Chia-Wen Ko, Gideon M. Blumenthal, Yuan Li Shen, Rajeshwari Sridhara, Ann T. Farrell, Albert B. Deisseroth, Xin Gao, Kelly J. Norsworthy, and Richard Pazdur
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Randomization ,Gemtuzumab ozogamicin ,Proportional hazards model ,business.industry ,Immunology ,Hazard ratio ,Salvage therapy ,Cell Biology ,Hematology ,Biochemistry ,Chemotherapy regimen ,Confidence interval ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Internal medicine ,medicine ,Midostaurin ,business ,medicine.drug - Abstract
Background: Several novel therapeutics are being developed for AML with demonstrable effects on response rates (e.g. complete remission [CR], CR with incomplete hematologic recovery [CRi], CR with incomplete platelet recovery [CRp], overall response rate [ORR = CR+CRi/CRp]), and EFS. Improvement in EFS recently served as the basis of approval of gemtuzumab ozogamicin (GO) for the treatment of newly-diagnosed CD33-positive AML in adults (Jen et al. Clin Cancer Res 2018). The relationship between response rate, EFS, and OS in newly-diagnosed AML has not been conclusively established. Therefore, we conducted trial-level and patient-level meta-analyses of newly-diagnosed AML trials submitted to the FDA. Methods: We searched for trials submitted with Biologics License or New Drug Applications for treatment of newly-diagnosed AML between 2007 and 2017. Criteria for inclusion were randomized, active-controlled, multicenter trials of intensive AML induction and consolidation chemotherapy. The estimated odd ratios (OR - ratio of odds of response in treatment to odds of response in controls) of CR and ORR and hazard ratios (HR - ratio of hazard of treatment versus control) for EFS and OS were calculated for each study. EFS was defined as time from randomization to treatment failure (defined as date of randomization for patients who failed to achieve CR following induction), disease relapse following CR, or death. Associations between treatment effects at the trial-level were evaluated using weighted least square (WLS) regression analyses on the log-scale (weighted by sample size of each randomized comparison). Coefficient of determination (R2) and 95% confidence interval (CI) were calculated to measure the strength of associations. Individual patient data for OS were plotted against EFS to explore their relationship. An exploratory patient-level responder analysis was performed to compare OS between responders and non-responders, regardless of treatment assignment in the pooled dataset. We estimated HRs of OS from Cox proportional hazards models. OS estimates by response were obtained based on the Kaplan-Meier method. Results: We identified 8 trials with a total of 4482 patients and 3 experimental agents (GO, n=5; [daunorubicin and cytarabine] liposome injection, n=2; midostaurin, n=1) for treatment of newly-diagnosed AML. Two trials tested therapy in defined patient populations (FLT3 mutant = 1 and secondary AML = 1). The association between OS HR and CR OR at the trial-level was moderate (R2 = 0.67, 95% CI: 0.16 - 0.94; Figure 1), whereas the association between OS HR and ORR OR was weak (R2 = 0.43, 95% CI: 0.03 - 0.98). For the OS vs. EFS HR analysis, a weaker than expected association was observed (data to be presented at the meeting). Individual patient level data scatter plots suggested that one possible reason for the lack of a strong association between EFS and OS was that early failures and relapses did not always result in worse OS (Figure 2). In the patient-level responder analysis, patients who achieved a CR response had better OS compared with CRi+CRp response and no response (Figure 3). Conclusions: On a trial level, the meta-analysis of randomized, active-controlled trials in newly-diagnosed AML suggests a moderate association between OS and CR rate, but not ORR. A strong association between EFS and OS is not established and may be confounded by improvements in salvage therapy, supportive care, hematopoietic stem cell transplantation, and/or differing censoring across trials. A patient-level responder analysis showed that CR responders have better OS compared with CRi+CRp responders and nonresponders. While our results are limited by the number of trials, they suggest that CR remains the response endpoint associated with greatest long-term benefit and that EFS, while clinically meaningful, is not a surrogate for OS. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.