1
|
Hu H, Li Y, Piasecki J, Hosseyni D, Yan Z, Liu X, Ogasawara K, Zhou S, Cheng Y. Population Pharmacokinetics of Orvacabtagene Autoleucel, an Autologous BCMA-Directed Chimeric Antigen Receptor T-cell Product, in Patients with Relapsed/Refractory Multiple Myeloma. Clin Cancer Res 2025; 31:1163-1171. [PMID: 39836430 DOI: 10.1158/1078-0432.ccr-24-2753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE Orvacabtagene autoleucel (orva-cel; JCARH125), a chimeric antigen receptor T-cell therapy targeting B-cell maturation antigen, was evaluated in patients with relapsed/refractory multiple myeloma in the EVOLVE phase I/II study (NCT03430011). We applied a modified piecewise model to characterize orva-cel transgene kinetics and assessed the impact of various covariates on its pharmacokinetics (PK). EXPERIMENTAL DESIGN The population PK analysis included 159 patients from the EVOLVE study. Traditional piecewise models, employing a first-order expansion rate with or without lag time followed by a biexponential contraction phase, were compared with a modified model incorporating a cell number-dependent expansion phase aligned with cellular physiology. Covariates assessed encompassed baseline demographics, dose levels (50-600 × 106 CD3+ chimeric antigen receptor+ T cells), prior/concomitant medications, baseline disease burden, and antitherapeutic antibody status. RESULTS Traditional piecewise models failed to accurately describe maximum orva-cel transgene level (Cmax) and underestimated the time to Cmax (Tmax). Our modified model incorporating a cell number-dependent expansion rate outperformed traditional models by (i) more accurately capturing the cellular expansion phase and (ii) yielding a Tmax that closely matches observed values. Additionally, dose level, percentage of plasma cells in bone marrow, and treatment-induced antitherapeutic antibody were identified as statistically significant covariates and associated with orva-cel expansion and/or persistence. CONCLUSIONS Orva-cel PK was adequately described by the modified piecewise model incorporating a cell number-dependent expansion phase, which aligns closely with T-cell biology.
Collapse
Affiliation(s)
- Hongxiang Hu
- Translational Medicine and Clinical Pharmacology, Bristol Myers Squibb, Summit, New Jersey
| | - Yan Li
- Translational Medicine and Clinical Pharmacology, Bristol Myers Squibb, Summit, New Jersey
| | - Julia Piasecki
- Cancer Immunology and Cell Therapy TRC, Bristol Myers Squibb, Seattle, Washington
| | - Daniela Hosseyni
- Late Clinical Development Cellular Therapy, Bristol Myers Squibb, Boudry, Switzerland
| | - Zhicheng Yan
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Lawrenceville, New Jersey
| | - Xianghong Liu
- Translational Medicine and Clinical Pharmacology, Bristol Myers Squibb, Summit, New Jersey
| | - Ken Ogasawara
- Translational Medicine and Clinical Pharmacology, Bristol Myers Squibb, Summit, New Jersey
| | - Simon Zhou
- Research and Pharmacology, Aurinia Pharmaceuticals, Rockville, Maryland
| | - Yiming Cheng
- Translational Medicine and Clinical Pharmacology, Bristol Myers Squibb, Summit, New Jersey
| |
Collapse
|
2
|
Li R, Grosskopf AK, Joslyn LR, Stefanich EG, Shivva V. Cellular Kinetics and Biodistribution of Adoptive T Cell Therapies: from Biological Principles to Effects on Patient Outcomes. AAPS J 2025; 27:55. [PMID: 40032717 DOI: 10.1208/s12248-025-01017-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 03/05/2025] Open
Abstract
Cell-based immunotherapy has revolutionized cancer treatment in recent years and is rapidly expanding as one of the major therapeutic options in immuno-oncology. So far ten adoptive T cell therapies (TCTs) have been approved by the health authorities for cancer treatment, and they have shown remarkable anti-tumor efficacy with potent and durable responses. While adoptive T cell therapies have shown success in treating hematological malignancies, they are lagging behind in establishing promising efficacy in treating solid tumors, partially due to our incomplete understanding of the cellular kinetics (CK) and biodistribution (including tumoral penetration) of cell therapy products. Indeed, recent clinical studies have provided ample evidence that CK of TCTs can influence clinical outcomes in both hematological malignancies and solid tumors. In this review, we will discuss the current knowledge on the CK and biodistribution of anti-tumor TCTs. We will first describe the typical CK and biodistribution characteristics of these "living" drugs, and the biological factors that influence these characteristics. We will then review the relationships between CK and pharmacological responses of TCT, and potential strategies in enhancing the persistence and tumoral penetration of TCTs in the clinic. Finally, we will also summarize bioanalytical methods, preclinical in vitro and in vivo tools, and in silico modeling approaches used to assess the CK and biodistribution of TCTs.
Collapse
Affiliation(s)
- Ran Li
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Abigail K Grosskopf
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Louis R Joslyn
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Eric Gary Stefanich
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Vittal Shivva
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
3
|
Zhang N, Li Y, Cui W, Yu X, Huang Y. Application of quantitative pharmacology analysis to support early clinical development of oncology drugs: dose selection. Xenobiotica 2024; 54:420-423. [PMID: 38994684 DOI: 10.1080/00498254.2024.2377577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024]
Abstract
The selection of appropriate starting dose and suitable method to predict an efficacious dose for novel oncology drug in the early clinical development stage poses significant challenges. The traditional methods of using body surface area transformation from toxicology studies to predict the first-in human (FIH) starting dose, or simply selecting the maximum tolerated dose (MTD) or maximum administered dose (MAD) as efficacious dose or recommended phase 2 dose (RP2D), are usually inadequate and risky for novel oncology drugs.Due to the regulatory efforts aimed at improving dose optimisation in oncology drug development, clinical dose selection is now shifting away from these traditional methods towards a comprehensive benefit/risk assessment-based approach. Quantitative pharmacology analysis (QPA) plays a crucial role in this new paradigm. This mini-review summarises the use of QPA in selecting the starting dose for oncology FIH studies and potential efficacious doses for expansion or phase 2 trials. QPA allows for a more rational and scientifically based approach to dose selection by integrating information across studies and development phases.In conclusion, the application of QPA in oncology drug development has the potential to significantly enhance the success rates of clinical trials and ultimately support clinical decision-making, particularly in dose selection.
Collapse
Affiliation(s)
| | - Yu Li
- Hansoh Pharmaceutical Group, Shanghai, China
| | - Wenbin Cui
- Hansoh Pharmaceutical Group, Shanghai, China
| | | | - Ying Huang
- Hansoh Pharmaceutical Group, Shanghai, China
| |
Collapse
|
4
|
Mody H, Sutaria DS, Miles D. Clinical Pharmacology Considerations for the "Off-the-Shelf" Allogeneic Cell Therapies. Clin Pharmacol Ther 2024; 115:1233-1250. [PMID: 38501153 DOI: 10.1002/cpt.3241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Autologous chimeric antigen receptor T-cell (CAR-T) therapies have garnered unprecedented clinical success with multiple regulatory approvals for the treatment of various hematological malignancies. However, there are still several clinical challenges that limit their broad utilization for aggressive disease conditions. To address some of these challenges, allogeneic cell therapies are evaluated as an alternative approach. As compared with autologous products, they offer several advantages, such as a more standardized "off the shelf" product, reduced manufacturing complexity, and no requirement of bridging therapy. As with autologous CAR-T therapies, allogeneic cell therapies also present clinical pharmacology challenges due to their in vivo living nature, unique pharmacokinetics or cellular kinetics (CKs), and complex dose-exposure-response relationships that are impacted by various patient- and product-related factors. On top of that, allogeneic cell therapies present additional unique challenges, including attenuated in vivo persistence and graft-vs.-host disease risk as compared with autologous counterparts. This review draws comparison between autologous and allogeneic cell therapies, summarizing key engineering aspects unique to allogeneic cell therapy. Clinical pharmacology learnings from emerging clinical data of allogeneic cell therapy programs are also highlighted, with particular emphasis on CK, dose-exposure-response relationship, lymphodepletion regimen, repeat dosing, and patient- and product-related factors that can impact CK and patient outcomes. There are specific unique challenges and opportunities arising from the development of allogeneic cell therapies, especially in optimizing lymphodepletion and establishing a regimen for repeat dosing. This review highlights how clinical pharmacologists are well positioned to help address these challenges by leveraging novel clinical pharmacology and modeling and simulation approaches.
Collapse
Affiliation(s)
- Hardik Mody
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | - Dale Miles
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| |
Collapse
|
5
|
Zhu X, Zhong Z, Meng F, Zou P, You Y, Li Q, Zhu X. CD26 CAR-T cells have attenuated mitochondrial and glycolytic metabolic profiling. Immunopharmacol Immunotoxicol 2023; 45:709-718. [PMID: 37382435 DOI: 10.1080/08923973.2023.2231632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/23/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Multiple targets of chimeric antigen receptor T cells (CAR-T cells) are shared expressed by tumor cells and T cells, these self-antigens may stimulate CAR-T cells continuously during the expansion. Persistent exposure to antigens is considered to cause metabolic reprogramming of T cells and the metabolic profiling is critical in determining the cell fate and effector function of CAR-T cells. However, whether the stimulation of self-antigens during CAR-T cell generation could remodel the metabolic profiling is unclear. In this study, we aim to investigate the metabolic characteristics of CD26 CAR-T cells, which expressed CD26 antigens themselves. METHODS The mitochondrial biogenesis of CD26 and CD19 CAR-T cells during expansion was evaluated by the mitochondrial content, mitochondrial DNA copy numbers and genes involved in mitochondrial regulation. The metabolic profiling was investigated by the ATP production, mitochondrial quality and the expression of metabolism-related genes. Furthermore, we assessed the phenotypes of CAR-T cells through memory-related markers. RESULTS We reported that CD26 CAR-T cells had elevated mitochondrial biogenesis, ATP production and oxidative phosphorylation at early expansion stage. However, the mitochondrial biogenesis, mitochondrial quality, oxidative phosphorylation and glycolytic activity were all weakened at later expansion stage. On the contrary, CD19 CAR-T cells did not exhibit such characteristics. CONCLUSION CD26 CAR-T cells showed distinctive metabolic profiling during expansion that was extremely unfavorable to cell persistence and function. These findings may provide new insights for the optimization of CD26 CAR-T cells in terms of metabolism.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhaodong Zhong
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fankai Meng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Zou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong You
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Li
- Department of Hematology, Wuhan No.1 Hospital, Wuhan, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Connarn JN, Witjes H, van Zutphen‐van Geffen M, de Greef R, Campbell TB, Hege K, Zhou S, Lamba M. Characterizing the exposure-response relationship of idecabtagene vicleucel in patients with relapsed/refractory multiple myeloma. CPT Pharmacometrics Syst Pharmacol 2023; 12:1687-1697. [PMID: 36794354 PMCID: PMC10681434 DOI: 10.1002/psp4.12922] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 02/17/2023] Open
Abstract
Idecabtagene vicleucel (ide-cel; bb2121) is a B-cell maturation antigen-directed chimeric antigen receptor (CAR) T cell therapy approved for treatment of patients with heavily pretreated relapsed and refractory multiple myeloma. This analysis evaluated exposure-response (ER) relationships of ide-cel with key efficacy end points and safety events. Ide-cel exposure data were available from 127 patients treated at target doses of 150, 300, or 450 × 106 CAR+ T cells from the phase II KarMMa study (NCT03361748). Key exposure metrics, including area under the curve of the transgene level from 0 to 28 days and maximum transgene level, were calculated using noncompartmental methods. Logistic regression models, using both linear and maximum response function of exposure on the logit scale, were evaluated to quantify observed ER trends, and modified by including statistically significant individual covariates in a stepwise regression analysis. There was wide overlap of exposures across the target doses. ER relationships were observed for the overall and complete response rates, with higher response rates associated with higher exposures. Model-based evaluations identified female sex and baseline serum monoclonal protein less than or equal to 10 g/L as predictive of a higher objective response rate and a higher complete response rate, respectively. ER relationships were observed for safety events of cytokine release syndrome requiring tocilizumab or corticosteroids. The established ER models were used to quantify the ide-cel dose-response, which showed a positive benefit-risk assessment for the range of ide-cel exposures associated with the target dose range of 150-450 × 106 CAR+ T cells.
Collapse
Affiliation(s)
| | - Han Witjes
- Certara Strategic ConsultingOssThe Netherlands
| | | | | | | | | | | | | |
Collapse
|
7
|
Mc Laughlin AM, Milligan PA, Yee C, Bergstrand M. Model-informed drug development of autologous CAR-T cell therapy: Strategies to optimize CAR-T cell exposure leveraging cell kinetic/dynamic modeling. CPT Pharmacometrics Syst Pharmacol 2023; 12:1577-1590. [PMID: 37448343 PMCID: PMC10681459 DOI: 10.1002/psp4.13011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023] Open
Abstract
Autologous Chimeric antigen receptor (CAR-T) cell therapy has been highly successful in the treatment of aggressive hematological malignancies and is also being evaluated for the treatment of solid tumors as well as other therapeutic areas. A challenge, however, is that up to 60% of patients do not sustain a long-term response. Low CAR-T cell exposure has been suggested as an underlying factor for a poor prognosis. CAR-T cell therapy is a novel therapeutic modality with unique kinetic and dynamic properties. Importantly, "clear" dose-exposure relationships do not seem to exist for any of the currently approved CAR-T cell products. In other words, dose increases have not led to a commensurate increase in the measurable in vivo frequency of transferred CAR-T cells. Therefore, alternative approaches beyond dose titration are needed to optimize CAR-T cell exposure. In this paper, we provide examples of actionable variables - design elements in CAR-T cell discovery, development, and clinical practice, which can be modified to optimize autologous CAR-T cell exposure. Most of these actionable variables can be assessed throughout the various stages of discovery and development as part of a well-informed research and development program. Model-informed drug development approaches can enable such study and program design choices from discovery through to clinical practice and can be an important contributor to cell therapy effectiveness and efficiency.
Collapse
Affiliation(s)
| | | | - Cassian Yee
- Department of Melanoma Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
- Department of ImmunologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | |
Collapse
|
8
|
Mody H, Ogasawara K, Zhu X, Miles D, Shastri PN, Gokemeijer J, Liao MZ, Kasichayanula S, Yang TY, Chemuturi N, Gupta S, Jawa V, Upreti VV. Best Practices and Considerations for Clinical Pharmacology and Pharmacometric Aspects for Optimal Development of CAR-T and TCR-T Cell Therapies: An Industry Perspective. Clin Pharmacol Ther 2023; 114:530-557. [PMID: 37393588 DOI: 10.1002/cpt.2986] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
With the promise of a potentially "single dose curative" paradigm, CAR-T cell therapies have brought a paradigm shift in the treatment and management of hematological malignancies. Both CAR-T and TCR-T cell therapies have also made great progress toward the successful treatment of solid tumor indications. The field is rapidly evolving with recent advancements including the clinical development of "off-the-shelf" allogeneic CAR-T therapies that can overcome the long and difficult "vein-to-vein" wait time seen with autologous CAR-T therapies. There are unique clinical pharmacology, pharmacometric, bioanalytical, and immunogenicity considerations and challenges in the development of these CAR-T and TCR-T cell therapies. Hence, to help accelerate the development of these life-saving therapies for the patients with cancer, experts in this field came together under the umbrella of International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) to form a joint working group between the Clinical Pharmacology Leadership Group (CPLG) and the Translational and ADME Sciences Leadership Group (TALG). In this white paper, we present the IQ consortium perspective on the best practices and considerations for clinical pharmacology and pharmacometric aspects toward the optimal development of CAR-T and TCR-T cell therapies.
Collapse
Affiliation(s)
- Hardik Mody
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Ken Ogasawara
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Lawrence Township, New Jersey, USA
| | - Xu Zhu
- Quantitative Clinical Pharmacology, AstraZeneca, Boston, Massachusetts, USA
| | - Dale Miles
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | - Jochem Gokemeijer
- Discovery Biotherapeutics, Bristol Myers Squibb, Cambridge, Massachusetts, USA
| | - Michael Z Liao
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | - Tong-Yuan Yang
- Bioanalytical Discovery and Development Sciences, Janssen R&D, LLC, Spring House, Pennsylvania, USA
| | - Nagendra Chemuturi
- Clinical Pharmacology, DMPK, Pharmacometrics, Moderna, Inc., Cambridge, Massachusetts, USA
| | - Swati Gupta
- Development Biological Sciences, Immunology, AbbVie, Irvine, California, USA
| | - Vibha Jawa
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Lawrence Township, New Jersey, USA
| | - Vijay V Upreti
- Clinical Pharmacology, Modeling & Simulation, Amgen, South San Francisco, California, USA
| |
Collapse
|
9
|
Mc Laughlin AM, Yee C. Letter to the editor: Quality criteria for computational models predicting individual outcomes in CAR-T cell therapy. J Immunother Cancer 2023; 11:e006990. [PMID: 37094987 PMCID: PMC10152039 DOI: 10.1136/jitc-2023-006990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 04/26/2023] Open
Affiliation(s)
| | - Cassian Yee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Paixão EA, Barros LRC, Fassoni AC, Almeida RC. Modeling Patient-Specific CAR-T Cell Dynamics: Multiphasic Kinetics via Phenotypic Differentiation. Cancers (Basel) 2022; 14:cancers14225576. [PMID: 36428671 PMCID: PMC9688514 DOI: 10.3390/cancers14225576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell immunotherapy revolutionized cancer treatment and consists of the genetic modification of T lymphocytes with a CAR gene, aiming to increase their ability to recognize and kill antigen-specific tumor cells. The dynamics of CAR-T cell responses in patients present multiphasic kinetics with distribution, expansion, contraction, and persistence phases. The characteristics and duration of each phase depend on the tumor type, the infused product, and patient-specific characteristics. We present a mathematical model that describes the multiphasic CAR-T cell dynamics resulting from the interplay between CAR-T and tumor cells, considering patient and product heterogeneities. The CAR-T cell population is divided into functional (distributed and effector), memory, and exhausted CAR-T cell phenotypes. The model is able to describe the diversity of CAR-T cell dynamical behaviors in different patients and hematological cancers as well as their therapy outcomes. Our results indicate that the joint assessment of the area under the concentration-time curve in the first 28 days and the corresponding fraction of non-exhausted CAR-T cells may be considered a potential marker to classify therapy responses. Overall, the analysis of different CAR-T cell phenotypes can be a key aspect for a better understanding of the whole CAR-T cell dynamics.
Collapse
Affiliation(s)
- Emanuelle A. Paixão
- Graduate Program, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil
- Correspondence:
| | - Luciana R. C. Barros
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Artur C. Fassoni
- Institute for Mathematics and Computer Science, Universidade Federal de Itajubá, Itajubá 37500-903, Brazil
| | - Regina C. Almeida
- Computational Modeling Department, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil
| |
Collapse
|
11
|
Venkatakrishnan K, van der Graaf PH. Toward Project Optimus for Oncology Precision Medicine: Multi-Dimensional Dose Optimization Enabled by Quantitative Clinical Pharmacology. Clin Pharmacol Ther 2022; 112:927-932. [PMID: 36264968 DOI: 10.1002/cpt.2742] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | - Piet H van der Graaf
- Certara QSP, Certara UK Ltd, Sheffield, UK.,Leiden University, Leiden, The Netherlands
| |
Collapse
|
12
|
He JZ, Wang H, Lim K, Ren S, Rollins F, Vallaster M, Wong R, Stebbings R, Standifer N, Keefe R, Phipps A, Gibbs M. A Consideration of Fixed Dosing Versus Body Size-Based Dosing Strategies for Chimeric Antigen Receptor T-Cell Therapies. Clin Pharmacol Drug Dev 2022; 11:1130-1135. [PMID: 36094760 PMCID: PMC9826131 DOI: 10.1002/cpdd.1171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/19/2022] [Indexed: 01/27/2023]
Affiliation(s)
- Jimmy Zhijian He
- Clinical Pharmacology and Quantitative PharmacologyBiopharmaceuticals R&D, AstraZenecaGaithersburgMarylandUSA
| | - Hechuan Wang
- Clinical Pharmacology and Quantitative PharmacologyBiopharmaceuticals R&D, AstraZenecaGaithersburgMarylandUSA
| | - KyoungSoo Lim
- Clinical Pharmacology and Quantitative PharmacologyBiopharmaceuticals R&D, AstraZenecaGaithersburgMarylandUSA
| | - Song Ren
- Clinical Pharmacology and Quantitative PharmacologyBiopharmaceuticals R&D, AstraZenecaGaithersburgMarylandUSA
| | - Fred Rollins
- Competitive Intelligence and AnalysisOncology R&D, AstraZenecaGaithersburgMarylandUSA
| | - Markus Vallaster
- Clinical DevelopmentCell Therapies and Immuno‐Oncology, AstraZenecaWalthamMassachusettsUSA
| | - Ryan Wong
- Clinical Pharmacology and Safety SciencesBiopharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Richard Stebbings
- Clinical Pharmacology and Safety SciencesBiopharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Nathan Standifer
- Integrated BioanalysisClinical Pharmacology and Safety SciencesBiopharmaceuticals R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Robert Keefe
- CMC DevelopmentCell Therapy, Oncology R&D, AstraZenecaGaithersburgMarylandUSA
| | - Alex Phipps
- Clinical Pharmacology and Quantitative PharmacologyBiopharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Megan Gibbs
- Clinical Pharmacology and Quantitative PharmacologyBiopharmaceuticals R&D, AstraZenecaGaithersburgMarylandUSA
| |
Collapse
|
13
|
Callréus T. The Randomised Controlled Trial at the Intersection of Research Ethics and Innovation. Pharmaceut Med 2022; 36:287-293. [PMID: 35877037 PMCID: PMC9309994 DOI: 10.1007/s40290-022-00438-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 11/12/2022]
Abstract
The randomised controlled trial (RCT) has been considered for a long time as the gold standard for evidence generation to support regulatory decision making for medicines. The randomisation procedure involves an ethical dilemma since it means leaving the treatment choice to chance. Although currently contested, the ethical justification for the RCT that has gained widespread acceptance is the notion of 'clinical equipoise'. This state exists when "there is no consensus within the expert clinical community about the comparative merits of the alternatives to be tested"; it is argued that this confers the ethical grounds for the conduct of an RCT. The prominent position of the RCT is being challenged by new therapeutic modalities for which this study design may be unsuitable. Moreover, alternative approaches to evidence generation represent another area where innovation may have implications for the relevance of the RCT. Against the backdrop of the debate around the equipoise principle and some recent therapeutic and data analytical innovations, the aim of this article is to explore the current standing of the RCT from a regulatory perspective.
Collapse
Affiliation(s)
- Torbjörn Callréus
- Malta Medicines Authority, Life Science Park, Sir Temi Żammit, San Gwann, 3000, Malta.
| |
Collapse
|
14
|
Mayawala K, de Alwis D. Dose Finding in Oncology: What is Impeding Coming of Age? Pharm Res 2022; 39:1817-1822. [PMID: 35474158 PMCID: PMC9314272 DOI: 10.1007/s11095-022-03263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Abstract
After a drug molecule enters clinical trials, there are primarily three levers to enhance probability of success: patient selection, dose selection and choice of combination agents. Of these, dose selection remains an under-appreciated aspect in oncology drug development despite numerous peer-reviewed publications. Here, we share practical challenges faced by the biopharmaceutical industry that reduce the willingness to invest in dose finding for oncology drugs. First, randomized dose finding admittedly slows down clinical development. To reduce the size of dose finding study, trend in exposure vs. tumor-size analysis can be assessed, instead of a statistical test for non-inferiority between multiple doses. Second, investment in testing a lower dose when benefit-risk at the higher dose is sufficient for regulatory approval (i.e., efficacy at the higher dose is better than standard of care and safety is acceptable) is perceived as low priority. Changing regulatory landscape must be considered to optimize dose in pre-marketing setting as post-marketing changes in dose can be commercially costly. Third, the risk of exposing patients to subtherapeutic exposures with a lower dose should be assessed scientifically instead of assuming a monotonic relationship between dose and efficacy. Only the doses which are expected to be at the plateau of dose/exposure-response curve should be investigated in Phase 1b/2. Overall, changing the perceptions that have been impeding investment in dose finding in oncology requires pragmatic discourse among biopharmaceutical industry, regulatory agencies and academia. These perceptions should also not deter dose finding for recently emerging modalities, including BITEs and CART cell therapies.
Collapse
Affiliation(s)
- Kapil Mayawala
- Oncology Early Development, Clinical Research, Merck and Co., Inc., NJ, Kenilworth, USA.
| | - Dinesh de Alwis
- Oncology Early Development, Clinical Research, Merck and Co., Inc., NJ, Kenilworth, USA
| |
Collapse
|