1
|
Kester R, Zalani S, Ebbinghaus S, Rubin E. The Utilization of the Accelerated Approval Pathway in Oncology: A Case Study of Pembrolizumab. Drugs 2024; 84:1579-1601. [PMID: 39538109 PMCID: PMC11652590 DOI: 10.1007/s40265-024-02111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
The accelerated approval (AA) pathway was established by the United States Food and Drug Administration (FDA) to provide earlier access to therapies for patients with serious medical conditions and unmet medical needs. Since its inception, the AA pathway has been used for novel treatments across different therapeutic areas, but most prominently in oncology, including the immune checkpoint inhibitor class. This review article describes the history of regulatory approvals for pembrolizumab, an immunotherapy agent targeting programmed death receptor-1 (PD-1), and use of the AA pathway and the corresponding regulatory decisions made by the FDA. From its first AA in September 2014 to February 2024, pembrolizumab has used the accelerated pathway for roughly 40% of the approved indications listed in the US Prescribing Information and was the first oncology therapy to receive an AA for an alternate dosing regimen and a tissue-agnostic indication. As of February 2024, 14 of the 18 indication-specific AAs and 1 post-marketing requirement (PMR) for the alternate dosing regimen AA were converted to traditional approvals. Accelerated approvals for two indications were withdrawn, and the remaining ongoing PMRs are not due until later in 2024 or 2025. The median conversion time from AA to traditional approval was 2.6 years, which is roughly 6 months earlier than the median time reported for oncology AAs. While FDA was the first agency to establish an expedited approval pathway, regulators from other countries have established similar pathways. For pembrolizumab, approximately half of the datasets that supported US AAs also supported expedited approval, or sometimes full approval, in Canada, EU, Australia or Japan. Ultimately, the AA pathway balances the provision of earlier access to therapies with overcoming uncertainty about potential effectiveness, and therefore it is important to confirm treatment benefit and withdraw indications that do not confirm benefit in a timely manner. The regulatory strategy and use of this expedited program for pembrolizumab highlights the importance of the AA pathway in providing oncology patients with earlier access to life-saving medications.
Collapse
Affiliation(s)
- Robert Kester
- Global Regulatory Affairs, Merck & Co., Inc., 126 E Lincoln Ave, Rahway, NJ, 07065, USA.
| | - Sunita Zalani
- Global Regulatory Affairs, Merck & Co., Inc., 126 E Lincoln Ave, Rahway, NJ, 07065, USA
| | - Scot Ebbinghaus
- Global Clinical Development Late-Stage Oncology, Merck & Co., Inc., Rahway, NJ, USA
| | - Eric Rubin
- Global Clinical Development-Oncology, Merck & Co., Inc., Rahway, NJ, USA
| |
Collapse
|
2
|
Hampel H, Caruso G, Nisticò R, Piccioni G, Mercuri NB, Giorgi FS, Ferrarelli F, Lemercier P, Caraci F, Lista S, Vergallo A. Biological Mechanism-based Neurology and Psychiatry: A BACE1/2 and Downstream Pathway Model. Curr Neuropharmacol 2023; 21:31-53. [PMID: 34852743 PMCID: PMC10193755 DOI: 10.2174/1570159x19666211201095701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 02/04/2023] Open
Abstract
In oncology, comprehensive omics and functional enrichment studies have led to an extensive profiling of (epi)genetic and neurobiological alterations that can be mapped onto a single tumor's clinical phenotype and divergent clinical phenotypes expressing common pathophysiological pathways. Consequently, molecular pathway-based therapeutic interventions for different cancer typologies, namely tumor type- and site-agnostic treatments, have been developed, encouraging the real-world implementation of a paradigm shift in medicine. Given the breakthrough nature of the new-generation translational research and drug development in oncology, there is an increasing rationale to transfertilize this blueprint to other medical fields, including psychiatry and neurology. In order to illustrate the emerging paradigm shift in neuroscience, we provide a state-of-the-art review of translational studies on the β-site amyloid precursor protein cleaving enzyme (BACE) and its most studied downstream effector, neuregulin, which are molecular orchestrators of distinct biological pathways involved in several neurological and psychiatric diseases. This body of data aligns with the evidence of a shared genetic/biological architecture among Alzheimer's disease, schizoaffective disorder, and autism spectrum disorders. To facilitate a forward-looking discussion about a potential first step towards the adoption of biological pathway-based, clinical symptom-agnostic, categorization models in clinical neurology and psychiatry for precision medicine solutions, we engage in a speculative intellectual exercise gravitating around BACE-related science, which is used as a paradigmatic case here. We draw a perspective whereby pathway-based therapeutic strategies could be catalyzed by highthroughput techniques embedded in systems-scaled biology, neuroscience, and pharmacology approaches that will help overcome the constraints of traditional descriptive clinical symptom and syndrome-focused constructs in neurology and psychiatry.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | | | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- School of Pharmacy, University of Rome “Tor Vergata”, Rome, Italy
| | - Gaia Piccioni
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- Department of Physiology and Pharmacology “V.Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Nicola B. Mercuri
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Pablo Lemercier
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, Troina, Italy
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Simone Lista
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
- Memory Resources and Research Center (CMRR), Neurology Department, Gui de Chauliac University Hospital, Montpellier, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| |
Collapse
|
3
|
Klein C, Zeng Q, Arbaretaz F, Devêvre E, Calderaro J, Lomenie N, Maiuri MC. Artificial Intelligence for solid tumor diagnosis in digital pathology. Br J Pharmacol 2021; 178:4291-4315. [PMID: 34302297 DOI: 10.1111/bph.15633] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 11/30/2022] Open
Abstract
Tumor diagnosis relies on the visual examination of histological slides by pathologists through a microscope eyepiece. Digital pathology, the digitalization of histological slides at high magnification with slides scanners, has raised the opportunity to extract quantitative information thanks to image analysis. In the last decade, medical image analysis has made exceptional progress due to the development of artificial intelligence (AI) algorithms. AI has been successfully used in the field of medical imaging and more recently in digital pathology. The feasibility and usefulness of AI assisted pathology tasks have been demonstrated in the very last years and we can expect those developments to be applied on routine histopathology in the future. In this review, we will describe and illustrate this technique and present the most recent applications in the field of tumor histopathology.
Collapse
Affiliation(s)
- Christophe Klein
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Qinghe Zeng
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France.,Laboratoire d'informatique Paris Descartes (LIPADE), Université de Paris, Paris, France
| | - Floriane Arbaretaz
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Estelle Devêvre
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Julien Calderaro
- Département de pathologie, Hôpital Henri Mondor, Créteil, France
| | - Nicolas Lomenie
- Laboratoire d'informatique Paris Descartes (LIPADE), Université de Paris, Paris, France
| | - Maria Chiara Maiuri
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
4
|
The development of HPLC-DAD method for determination of active pharmaceutical ingredient in the potassium 2-((4-amino-5-(morpholinomethyl)-4H-1,2,4-triazol-3-yl)thio) acetate substance. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2019. [DOI: 10.2478/cipms-2019-0001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Derivatives of 1,2,4-triazole are actively researched by scientists and synthetic pharmacologists. The last studies have shown that potassium 2-((4-amino-5-(morpholinomethyl)-4H-1,2,4-triazol-3-yl)thio)acetate with low toxicity series exhibits antioxidant and hepatoprotective properties. Therefore, the purpose of this work was to develop a method for determining the API in the potassium 2-((4-amino-5-(morpholinomethyl)-4H-1,2,4-triazol-3-yl)thio)acetate substance using the method of high-performance liquid chromatography with diode array detection (HPLC-DAD). As a result of this work, it is shown that the developed method is specific and meets the requirements of linearity, accuracy and precision. The results of determining the contents of the API in real samples indicate that the method can be proposed to control the quality of the potassium 2-((4-amino-5-(morpholinomethyl)-4H-1,2,4-triazol-3-yl)thio)acetate substance.
Collapse
|
5
|
Ottaiano A. Real practice studies in oncology: A positive perspective. World J Gastrointest Oncol 2018; 10:228-230. [PMID: 30254718 PMCID: PMC6147770 DOI: 10.4251/wjgo.v10.i9.228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/12/2018] [Accepted: 08/04/2018] [Indexed: 02/05/2023] Open
Abstract
In the majority of phase III clinical trials, patients are generally excluded on the basis of specific comorbidities, performance status Eastern Cooperative Oncology Group ≥ 2, age ≥ 65 years, previous malignancies, brain metastases, active infections, psychiatric disorders, non-measurable disease, number and type of previous lines of chemotherapies or biologic therapies. A question is raised: Can results of phase IIIstudies be extended to the general population? There is increasing attention to and a resurgence of some terms as “real world” or “real practice” which are wrongly viewed as contrary to clinical trial protocols. In fact, the general perception is that a contraposition exists between “wrong” (retrospective and biased) and “right” (prospective, randomized, well statistically designed) clinical research. We have to change this perspective. Real practice studies, generally retrospective in their nature, deserve to be reevaluated; biases are physiologically present but their punctual and rigorous description and analysis can help the interpretation of and in some cases reinforce results and their hypothesis-generating power. The correct and balanced interaction between clinical trials and real practice reports can help the scientific community to improve the knowledge on anti-cancer drug efficacy.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- SSD-Innovative Therapies for Abdominal Metastases, Clinical and Experimental Abdominal Oncology, Istituto Nazionale Tumori di Napoli, Naples 80131, Italy
| |
Collapse
|
6
|
Serrano P, Hartmann M, Schmitt E, Franco P, Amexis G, Gross J, Mayer-Nicolai C. Clinical Development and Initial Approval of Novel Immune Checkpoint Inhibitors in Oncology: Insights From a Global Regulatory Perspective. Clin Pharmacol Ther 2018; 105:582-597. [PMID: 29923615 DOI: 10.1002/cpt.1123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/18/2018] [Indexed: 12/28/2022]
Abstract
Immune checkpoint inhibitors (ICI) have demonstrated meaningful patterns of clinical efficacy across various cancers. During their development, novel regulatory strategies and clinical design approaches were explored. This metrics-based narrative review examines submission strategies and clinical evidence expectations of the US, European, and Japanese drug agencies, as well as their impact on approval and overall development times. Also discussed is the role of emerging clinical science and biomarker evaluation to get the first six ICI initially approved.
Collapse
Affiliation(s)
- Philippe Serrano
- R&D Regulatory Oncology, EMD Serono Research & Development Institute, Billerica, Massachusetts, USA
| | | | - Elmar Schmitt
- R&D Regulatory Oncology, Merck KGaA, Darmstadt, Germany
| | - Pedro Franco
- Global Regulatory & Scientific Policy, Merck Serono Europe Ltd, London, UK
| | | | - Jan Gross
- R&D Regulatory Oncology, Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
7
|
Kuderer NM, Lyman GH. Evolving Landscape of US Food and Drug Administration Drug Approval in the Era of Precision Oncology: Finding the Right Balance Between Access and Safety. J Clin Oncol 2018; 36:1773-1776. [PMID: 29742010 DOI: 10.1200/jco.2018.78.5592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Nicole M Kuderer
- Nicole M. Kuderer, Advanced Cancer Research Group, Seattle, WA; and Gary H. Lyman, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Gary H Lyman
- Nicole M. Kuderer, Advanced Cancer Research Group, Seattle, WA; and Gary H. Lyman, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
8
|
Jørgensen JT, Hersom M. Clinical and Regulatory Aspects of Companion Diagnostic Development in Oncology. Clin Pharmacol Ther 2018; 103:999-1008. [PMID: 29197081 DOI: 10.1002/cpt.955] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/16/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
Nearly 20 years have passed since the US Food and Drug Administration (FDA) approved the first companion diagnostic and today this type of assay governs the use of 21 different anticancer drugs. The regulators deem these assays essential for the safe and effective use of a corresponding therapeutic product. The companion diagnostic assays are important both during the drug development process as well as essential treatment decision tools after the approval of the drugs.
Collapse
Affiliation(s)
| | - Maria Hersom
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Goldberg KB, Blumenthal GM, McKee AE, Pazdur R. The FDA Oncology Center of Excellence and precision medicine. Exp Biol Med (Maywood) 2017; 243:308-312. [PMID: 29105511 DOI: 10.1177/1535370217740861] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In January 2017, the U.S. Food and Drug Administration (FDA) formally established the Oncology Center of Excellence (OCE) to streamline the development of cancer therapies by uniting experts from FDA product centers to conduct expedited review of drugs, biologics, and devices. In May 2017, the FDA approved a cancer treatment based on a biomarker, without regard to the tumor's site, by granting accelerated approval to pembrolizumab for patients with solid tumors that have the microsatellite instability-high or mismatch repair deficient biomarker. We describe here the OCE's role in this first site-agnostic approval and OCE programs for further advancement of oncology-related regulatory science and policy. In addition, the FDA's four expedited review programs that enable transformative therapies to reach patients with life-threatening malignancies earlier in the development process are key to the continued rapid development of safe and effective therapies for patients with few or no other treatment options. These changes at FDA are taking place in the context of recent progress in the understanding of the genetic and immunologic foundations of cancer, resulting in the development of targeted therapies and immunotherapies. The traditional system of phased clinical trials has evolved as early trials of breakthrough therapies use expansion cohorts in a process known as seamless drug development. Increasingly, FDA approvals of targeted therapies are likely to have contemporaneous approvals of companion diagnostics to identify patients whose cancers harbor actionable abnormalities. Impact statement This publication describes the U.S. Food and Drug Administration's (FDA) first site-agnostic oncology drug approval, a landmark event in the history of cancer drug development. The role of the FDA's newly established Oncology Center of Excellence (OCE) in this approval is described, as are several OCE programs to advance excellence in regulatory science in the era of precision medicine. Also provided is an overview of FDA's expedited drug review programs, which are important to the continued acceleration of therapeutics development for patients with life-threatening diseases and few or no other treatment options.
Collapse
Affiliation(s)
- Kirsten B Goldberg
- 1 Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Gideon M Blumenthal
- 1 Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Amy E McKee
- 1 Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Richard Pazdur
- 1 Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA.,2 Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
10
|
Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, Barker AD, Bastani R, Bhatia S, Bluestone JA, Brawley O, Butte AJ, Coit DG, Davidson NE, Davis M, DePinho RA, Diasio RB, Draetta G, Frazier AL, Futreal A, Gambhir SS, Ganz PA, Garraway L, Gerson S, Gupta S, Heath J, Hoffman RI, Hudis C, Hughes-Halbert C, Ibrahim R, Jadvar H, Kavanagh B, Kittles R, Le QT, Lippman SM, Mankoff D, Mardis ER, Mayer DK, McMasters K, Meropol NJ, Mitchell B, Naredi P, Ornish D, Pawlik TM, Peppercorn J, Pomper MG, Raghavan D, Ritchie C, Schwarz SW, Sullivan R, Wahl R, Wolchok JD, Wong SL, Yung A. Future cancer research priorities in the USA: a Lancet Oncology Commission. Lancet Oncol 2017; 18:e653-e706. [PMID: 29208398 PMCID: PMC6178838 DOI: 10.1016/s1470-2045(17)30698-8] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022]
Abstract
We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US$2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.
Collapse
Affiliation(s)
| | - Chi Van Dang
- Ludwig Institute for Cancer Research New York, NY; Wistar Institute, Philadelphia, PA, USA.
| | - David B Agus
- University of Southern California, Beverly Hills, CA, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Alan Ashworth
- University of California San Francisco, San Francisco, CA, USA
| | | | - Roshan Bastani
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Sangeeta Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey A Bluestone
- University of California San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Atul J Butte
- University of California San Francisco, San Francisco, CA, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Mark Davis
- California Institute for Technology, Pasadena, CA, USA
| | | | | | - Giulio Draetta
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Lindsay Frazier
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Patricia A Ganz
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Levi Garraway
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA; Eli Lilly and Company, Boston, MA, USA
| | | | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Faculty of Medicine and IHPME, University of Toronto, Toronto, Canada
| | - James Heath
- California Institute for Technology, Pasadena, CA, USA
| | - Ruth I Hoffman
- American Childhood Cancer Organization, Beltsville, MD, USA
| | - Cliff Hudis
- Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Chanita Hughes-Halbert
- Medical University of South Carolina and the Hollings Cancer Center, Charleston, SC, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Hossein Jadvar
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Kavanagh
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| | - Rick Kittles
- College of Medicine, University of Arizona, Tucson, AZ, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | - Scott M Lippman
- University of California San Diego Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - David Mankoff
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Mardis
- The Institute for Genomic Medicine at Nationwide Children's Hospital Columbus, OH, USA; College of Medicine, Ohio State University, Columbus, OH, USA
| | - Deborah K Mayer
- University of North Carolina Lineberger Cancer Center, Chapel Hill, NC, USA
| | - Kelly McMasters
- The Hiram C Polk Jr MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dean Ornish
- University of California San Francisco, San Francisco, CA, USA
| | - Timothy M Pawlik
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | | | - Martin G Pomper
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Raghavan
- Levine Cancer Institute, Carolinas HealthCare, Charlotte, NC, USA
| | | | - Sally W Schwarz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Richard Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Sandra L Wong
- Department of Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alfred Yung
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Mayawala K, Tse A, Rubin EH, Jain L, de Alwis DP. Dose Finding Versus Speed in Seamless Immuno-Oncology Drug Development. J Clin Pharmacol 2017; 57 Suppl 10:S143-S145. [PMID: 28921649 DOI: 10.1002/jcph.1001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 02/01/2023]
Affiliation(s)
- Kapil Mayawala
- Quantitative Pharmacology and Pharmacometrics, PPDM, Kenilworth, NJ, USA
| | - Archie Tse
- Oncology Early Development, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Eric H Rubin
- Oncology Early Development, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Lokesh Jain
- Quantitative Pharmacology and Pharmacometrics, PPDM, Kenilworth, NJ, USA
| | - Dinesh P de Alwis
- Quantitative Pharmacology and Pharmacometrics, PPDM, Kenilworth, NJ, USA
| |
Collapse
|
12
|
Flexibility and innovation in the FDA's novel regulatory approval strategies for hematologic drugs. Blood 2017; 130:1285-1289. [PMID: 28774876 DOI: 10.1182/blood-2017-04-742726] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/13/2017] [Indexed: 02/04/2023] Open
|
13
|
Lee J, Blumenthal GM, Hohl RJ, Huang SM. Cancer Therapy: Shooting for the Moon. Clin Pharmacol Ther 2017; 101:552-558. [PMID: 28418166 PMCID: PMC5525193 DOI: 10.1002/cpt.655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/06/2017] [Indexed: 11/15/2022]
Affiliation(s)
- Jsh Lee
- Office of the Director, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - G M Blumenthal
- Office of Hematology & Oncology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - R J Hohl
- Penn State Cancer Institute, Pennsylvania State University, Hershey, Pennsylvania, USA
| | - S-M Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|