1
|
Cascone T, Bonanno L, Guisier F, Insa A, Liberman M, Bylicki O, Livi L, Egenod T, Corre R, Kim DW, Garcia Campelo MR, Provencio Pulla M, Shim BY, Metro G, Bennouna J, Bielska AA, Yohannes AR, He Y, Dowson A, Kar G, McGrath L, Kumar R, Grenga I, Spicer J, Forde PM. Perioperative durvalumab plus chemotherapy plus new agents for resectable non-small-cell lung cancer: the platform phase II NeoCOAST-2 trial. Nat Med 2025:10.1038/s41591-025-03746-z. [PMID: 40450142 DOI: 10.1038/s41591-025-03746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 04/30/2025] [Indexed: 06/03/2025]
Abstract
In the phase II NeoCOAST-2 platform study, 202 patients with untreated, resectable stage IIA-IIIB non-small-cell lung cancer (NSCLC) were randomized to receive neoadjuvant durvalumab plus platinum-doublet chemotherapy with oleclumab, a CD73 inhibitor (Arm 1), or with monalizumab, a NKG2A inhibitor (Arm 2), or neoadjuvant durvalumab plus single-agent platinum chemotherapy with the TROP-2 antibody-drug conjugate (ADC) datopotamab deruxtecan (Arm 4), followed by surgical resection and adjuvant durvalumab with oleclumab or monalizumab (Arms 1 and 2) or durvalumab alone (Arm 4). Primary endpoints were pathological complete response (pCR) rate and safety; secondary endpoints included feasibility of surgery and major pathological response (mPR) rate. In the modified intention-to-treat population (n = 198; Arm 1, n = 74; Arm 2, n = 70; Arm 4, n = 54), pCR rates were 20.3% (15/74; 95% CI, 11.8-31.2), 25.7% (18/70; 95% CI, 16.0-37.6) and 35.2% (19/54; 95% CI, 22.7-49.4), and mPR rates were 41.9% (31/74; 95% CI, 30.5-53.9), 50.0% (35/70; 95% CI, 37.8-62.2) and 63.0% (34/54; 95% CI, 48.7-75.7) in arms 1, 2, and 4, respectively. In the safety population, 69/74 (93.2%), 66/71 (93.0%), and 51/54 (94.4%) patients underwent surgery, respectively. Overall, grade ≥3 treatment-related adverse events occurred in 27/74 (36.5%), 29/71 (40.8%) and 11/54 (20.4%) patients, respectively. In NeoCOAST-2, the first neoadjuvant trial examining an ADC plus chemo-immunotherapy in resectable NSCLC, pCR rates were highest in the datopotamab-deruxtecan-containing arm, warranting further investigation in larger trials of ADCs and checkpoint inhibition in the neoadjuvant setting. ClinicalTrials.gov identifier: NCT05061550 .
Collapse
Affiliation(s)
- Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Laura Bonanno
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Florian Guisier
- Univ Rouen Normandie, LITIS Lab QuantIF team EA4108, CHU Rouen, Department of Pneumology and Inserm CIC-CRB 1404, Rouen, France
| | - Amelia Insa
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Moishe Liberman
- Division of Thoracic Surgery, University of Montréal, Montréal, Quebec, Canada
- CETOC - CHUM Endoscopic Tracheobronchial and Oesophageal Center, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Olivier Bylicki
- Pneumology Department, Hôpital d'Instruction des Armées Sainte-Anne, Toulon, France
| | - Lorenzo Livi
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Thomas Egenod
- Department of Thoracic Oncology, Dupuytren University Hospital, Limoges, France
| | - Romain Corre
- Department of Medical Oncology, CH de Cornouaille, Quimper, France
| | - Dong-Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National Hospital, Seoul, South Korea
| | | | | | - Byoung Yong Shim
- Department of Medical Oncology, The Catholic University of Korea, St. Vincent's Hospital, Seoul, South Korea
| | - Giulio Metro
- Santa Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Jaafar Bennouna
- Department of Medical Oncology, Hôpital Foch, Suresnes, France
| | | | | | - Yun He
- AstraZeneca, Waltham, MA, USA
| | | | | | | | | | | | - Jonathan Spicer
- Department of Thoracic Surgery, McGill University, Montreal, Quebec, Canada
| | - Patrick M Forde
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
2
|
Dunn A, Gobburu JVS. The trajectory of pharmacometrics to support drug licensing and labelling. Br J Clin Pharmacol 2025; 91:932-937. [PMID: 37005339 DOI: 10.1111/bcp.15728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
The field of pharmacometrics has been responsible for countless advancements within the drug development space. In recent years, we have witnessed the implementation of both new and revived analytical methods to increase clinical trial success and even supplement the need for clinical trials all together. Throughout this article, we will explore the path of pharmacometrics from its inception to the present day. At this point in time, the target of drug development has been the average patient, and population approaches have primarily been utilized to support just that. The challenge we are now facing involves the translation from treating the typical patient to treating the real-world patient. For this reason, it is our opinion that future development efforts should account more for the individual. With advanced pharmacometric methods and growing technological infrastructure, precision medicine can become a development priority rather than a clinician's burden.
Collapse
Affiliation(s)
- Allison Dunn
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Jogarao V S Gobburu
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Grieve AP. Pre-Posterior Distributions in Drug Development and Their Properties. Pharm Stat 2025; 24:e2450. [PMID: 39587429 DOI: 10.1002/pst.2450] [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: 04/15/2024] [Revised: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024]
Abstract
The topic of this article is pre-posterior distributions of success or failure. These distributions, determined before a study is run and based on all our assumptions, are what we should believe about the treatment effect if we are told only that the study has been successful, or unsuccessful. I show how the pre-posterior distributions of success and failure can be used during the planning phase of a study to investigate whether the study is able to discriminate between effective and ineffective treatments. I show how these distributions are linked to the probability of success (PoS), or failure, and how they can be determined from simulations if standard asymptotic normality assumptions are inappropriate. I show the link to the concept of the conditionalP o S $$ P o S $$ introduced by Temple and Robertson in the context of the planning of multiple studies. Finally, I show that they can also be constructed regardless of whether the analysis of the study is frequentist or fully Bayesian.
Collapse
|
4
|
Starling MS, Kehoe L, Burnett BK, Green P, Venkatakrishnan K, Madabushi R. The Potential of Disease Progression Modeling to Advance Clinical Development and Decision Making. Clin Pharmacol Ther 2025; 117:343-352. [PMID: 39410710 PMCID: PMC11739755 DOI: 10.1002/cpt.3467] [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: 05/16/2024] [Accepted: 09/25/2024] [Indexed: 01/19/2025]
Abstract
While some model-informed drug development frameworks are well recognized as enabling clinical trials, the value of disease progression modeling (DPM) in impacting medical product development has yet to be fully realized. The Clinical Trials Transformation Initiative assembled a diverse project team from across the patient, academic, regulatory, and industry sectors of practice to advance the use of DPM for decision making in clinical trials and medical product development. This team conducted a scoping review to explore current applications of DPM and convened a multi-stakeholder expert meeting to discuss its value in medical product development. In this article, we present the scoping review and expert meeting output and propose key questions that medical product developers and regulators may use to inform clinical development strategy, appreciate the therapeutic context and endpoint selection, and optimize trial design with disease progression models. By expanding awareness of the unique value of DPM, this article does not aim to be technical in nature but rather aims to highlight the potential of DPM to improve the quality and efficiency of medical product development.
Collapse
Affiliation(s)
- Mary Summer Starling
- The Clinical Trials Transformation InitiativeDuke Clinical Research InstituteDurhamNorth CarolinaUSA
- Department of Population Health SciencesDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Lindsay Kehoe
- The Clinical Trials Transformation InitiativeDuke Clinical Research InstituteDurhamNorth CarolinaUSA
| | - Bruce K. Burnett
- Division of Allergy, Immunology and TransplantationNational Institutes of HealthBethesdaMarylandUSA
| | | | | | | |
Collapse
|
5
|
Xiong Y, Samtani MN, Ouellet D. Applications of pharmacometrics in drug development. Adv Drug Deliv Rev 2025; 217:115503. [PMID: 39701388 DOI: 10.1016/j.addr.2024.115503] [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: 02/23/2024] [Revised: 11/17/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
The last two decades have witnessed profound changes in how advanced computational tools can help leverage tons of data to improve our knowledge, and ultimately reduce cost and increase productivity in drug development. Pharmacometrics has demonstrated its impact through model-informed drug development (MIDD) approaches. It is now an indispensable component throughout the whole continuum of drug discovery, development, regulatory review, and approval. Today, applications of pharmacometrics are common in designing better trials and accelerating evidence-based decisions. Newly emerging technologies, especially those from data and computer sciences, are being integrated with existing computational tools used in the pharmaceutical industry at a remarkably fast pace. The new challenges faced by the pharmacometrics community are not what or how to contribute, but which optimal MIDD strategy should be adopted to maximize its value in the decision-making process. While we are embracing new innovative approaches and tools, this article discusses how a variety of existing modeling tools, with differentiated advantages and focus, can work in concert to inform drug development.
Collapse
|
6
|
Sasaki T, Katsube T, Hayato S, Yamaguchi S, Tanaka J, Yoshimatsu H, Nakanishi Y, Kitamura A, Watase H, Suganami H, Matsuoka N, Hasegawa C. Application of model-informed drug development (MIDD) for dose selection in regulatory submissions for drug approval in Japan. J Pharmacokinet Pharmacodyn 2025; 52:10. [PMID: 39762622 DOI: 10.1007/s10928-024-09954-3] [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: 06/21/2024] [Accepted: 10/14/2024] [Indexed: 02/17/2025]
Abstract
Model-informed drug development (MIDD) is an approach to improve the efficiency of drug development. To promote awareness and application of MIDD in Japan, the Data Science Expert Committee of the Drug Evaluation Committee in the Japan Pharmaceutical Manufacturers Association established a task force, which surveyed MIDD applications for approved products in Japan. This study aimed to reveal the trends and challenges in the use of MIDD by analyzing the survey results. A total of 322 cases approved in Japan between January 2020 and March 2022 as medical products were included in the survey. Modeling analysis was performed in approximately half of the cases (47.8% [154/322]) and formed a major basis for the selection or justification of dosage and administration in approximately one-fourth of the cases [24.2% (78/322)]. Modeling analysis/model-based dose selection was frequently conducted in cases involving monoclonal antibodies, first indication, orphan drugs, and multi-regional trials. Moreover, the survey results indicated that modeling analyses contributed to dose optimization throughout the developmental phases, including changing dose levels from phase II to phase III and dose adjustment in special populations. Japanese data were included in most cases in which modeling analysis was used for dosage selection. Thus, modelling analysis may also address ethnic factors introduced in the ICH E5 and/or E17 guidelines. In summary, this survey is useful for understanding the current status of MIDD use in Japan and for future drug development.
Collapse
Affiliation(s)
- Tomohiro Sasaki
- Clinical Pharmacology, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| | - Takayuki Katsube
- Clinical Pharmacology and Pharmacokinetics, Shionogi & Co., Ltd., Osaka, Japan
| | - Seiichi Hayato
- Clinical Pharmacology Science, Eisai Co., Ltd., Tokyo, Japan
| | | | - Jun Tanaka
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb K.K., Tokyo, Japan
| | | | - Yushi Nakanishi
- Clinical Data Science Department, Kowa Company, Ltd., Tokyo, Japan
| | | | - Hirotaka Watase
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine and Early Development, R&D, Sanofi K.K., Tokyo, Japan
| | - Hideki Suganami
- Clinical Data Science Department, Kowa Company, Ltd., Tokyo, Japan
| | | | | |
Collapse
|
7
|
Yin A, de Groot FA, Guchelaar HJ, Nijland M, Doorduijn JK, Touw DJ, Munnink TO, de Winter BCM, Friberg LE, Vermaat JSP, Moes DJAR. Population Pharmacokinetic and Toxicity Analysis of High-Dose Methotrexate in Patients with Central Nervous System Lymphoma. Clin Pharmacokinet 2025; 64:79-91. [PMID: 39625585 DOI: 10.1007/s40262-024-01452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 01/26/2025]
Abstract
BACKGROUND High-dose methotrexate (HD-MTX)-based polychemotherapy is widely used for patients with central nervous system (CNS) lymphoma. The pharmacokinetic (PK) variability and unpredictable occurrence of toxicity remain major concerns in HD-MTX treatment. OBJECTIVES This study aimed to characterize the population PK of HD-MTX in patients with CNS lymphoma and to identify baseline predictors and exposure thresholds that predict a high risk of nephro- and hepatotoxicity. METHODS Routinely monitored serum MTX concentrations after intravenous infusion of HD-MTX and MTX dosing information were collected retrospectively. Acute event of toxicity (≥ grade 1) was defined according to the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 on the basis of serum creatinine and alanine aminotransferase. A population PK model was developed in NONMEM. Toxicity data were analyzed using a logistic regression model, and potential baseline and exposure-related predictors were investigated. RESULTS In total, 1584 MTX concentrations from 110 patients were available for analysis. A two-compartment population PK model adequately described the data. Estimated glomerular filtration rate (eGFR), treatment regimen, albumin, alkaline phosphatase, and body weight were identified as significant covariates that explain the PK variability of HD-MTX. Baseline eGFR and sex were identified as significant predictors for renal toxicity, and MTX dose (mg/m2) was the strongest predictor for hepatotoxicity. The MTX area under the concentration-time curve (AUC24-∞) and concentration at 24 h (C24h) were shown to correlate with renal toxicity only, and 49,800 μg/L × h (109.6 μmol/L × h) and C24h > 3930 μg/L (8.65 μmol/L) were potential exposure thresholds predicting high risk (proportion > 60%). CONCLUSIONS A population PK model was developed for HD-MTX in patients with CNS lymphoma. The toxicity analysis showed that lower baseline eGFR and male sex, and higher MTX dose are associated with increased risk of acute nephro- and hepatotoxicity, respectively. The proposed exposure thresholds that predict high risk of renal toxicity and the developed models hold the potential to guide HD-MTX dosage individualization and better prevent acute toxicity.
Collapse
Affiliation(s)
- Anyue Yin
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Fleur A de Groot
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Marcel Nijland
- Department of Hematology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jeanette K Doorduijn
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Thijs Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Brenda C M de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
8
|
Liu H, Ibrahim EIK, Centanni M, Sarr C, Venkatakrishnan K, Friberg LE. Integrated modeling of biomarkers, survival and safety in clinical oncology drug development. Adv Drug Deliv Rev 2025; 216:115476. [PMID: 39577694 DOI: 10.1016/j.addr.2024.115476] [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: 05/31/2024] [Revised: 09/12/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Model-based approaches, including population pharmacokinetic-pharmacodynamic modeling, have become an essential component in the clinical phases of oncology drug development. Over the past two decades, models have evolved to describe the temporal dynamics of biomarkers and tumor size, treatment-related adverse events, and their links to survival. Integrated models, defined here as models that incorporate at least two pharmacodynamic/ outcome variables, are applied to answer drug development questions through simulations, e.g., to support the exploration of alternative dosing strategies and study designs in subgroups of patients or other tumor indications. It is expected that these pharmacometric approaches will be expanded as regulatory authorities place further emphasis on early and individualized dosage optimization and inclusive patient-focused development strategies. This review provides an overview of integrated models in the literature, examples of the considerations that need to be made when applying these advanced pharmacometric approaches, and an outlook on the expected further expansion of model-informed drug development of anticancer drugs.
Collapse
Affiliation(s)
- Han Liu
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Eman I K Ibrahim
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Maddalena Centanni
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Céline Sarr
- Pharmetheus AB, Dragarbrunnsgatan 77, 753 19, Uppsala, Sweden
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden.
| |
Collapse
|
9
|
Azeredo FJ, Schmidt S. Editorial: Pharmacometrics and systems pharmacology: Principles and applications. Eur J Pharm Sci 2024; 203:106941. [PMID: 39426553 DOI: 10.1016/j.ejps.2024.106941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Affiliation(s)
- Francine Johansson Azeredo
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA.
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
10
|
Sharma VD, Bhattaram VA, Krudys K, Li Z, Marathe A, Mehrotra N, Wang X, Liu J, Stier E, Florian J, Madabushi R, Zhu H. Driving Efficiency: Leveraging Model-Informed Approaches in 505(b)(2) Regulatory Actions. J Clin Pharmacol 2024; 64:1484-1490. [PMID: 39120874 DOI: 10.1002/jcph.6109] [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: 03/22/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024]
Abstract
Introduced by the Hatch-Waxman Amendments of 1984, 505(b)(2) applications permit the US Food and Drug Administration to rely, for approval of a new drug application, on information from studies not conducted by or for the applicant and for which the applicant has not obtained a right of reference. This pathway is designed to circumvent the unnecessary duplication of studies already conducted on a previously approved drug. It can lead to a considerably more efficient and expedited route to approval compared to a traditional development path. Model-informed drug development refers to the utilization of a diverse array of quantitative models in drug development to streamline the decision-making process. In this approach, diverse quantitative models that integrate knowledge of physiology, disease processes, and drug pharmacology are employed to address drug development challenges and guide regulatory decisions. Integration of these model-informed approaches into 505(b)(2) regulatory submissions and decision-making can further expedite the approval of new drugs. This article discusses some applications of model-informed approaches that were used to support 505(b)(2) drug development and regulatory actions. Specifically, various quantitative models such as population pharmacokinetic and exposure-response models have been employed to provide evidence of effectiveness, guide dosing in subgroups such as subjects with hepatic or renal impairment, and inform policies. These case study examples collectively underscore the significance of model-informed approaches in drug development and regulatory decisions associated with 505(b)(2) submissions.
Collapse
Affiliation(s)
- Vishnu Dutt Sharma
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Venkatesh Atul Bhattaram
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Kevin Krudys
- Office of Neuroscience, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Zhihua Li
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Anshu Marathe
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Nitin Mehrotra
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Xiaofeng Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Jiang Liu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Ethan Stier
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Jeffry Florian
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Raj Madabushi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
11
|
Li Z, Roepcke S, Franke R, Yel L. Dose, exposure, and treatment regimen of intravenous immunoglobulin G in multifocal motor neuropathy. Front Neurol 2024; 15:1478419. [PMID: 39574508 PMCID: PMC11580011 DOI: 10.3389/fneur.2024.1478419] [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: 08/09/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction Intravenous immunoglobulin (IVIG) is the only approved treatment for multifocal motor neuropathy (MMN), a rare, chronic, immune-mediated demyelinating neuropathy. There is a significant gap in understanding of the role of serum immunoglobulin G (IgG) levels in the efficacy of IVIG in affected patients. We aimed to characterize the interplay between dose and exposure of IVIG and the effects of patient factors on individual variabilities. Methods Serum IgG trough concentration data from a phase 3, randomized, double-blind, placebo-controlled, crossover trial of IVIG 10% in 44 patients with MMN (NCT00666263) were analyzed using fit-for-purpose population PK modeling. Patient factors were tested as covariates, and IgG PK profiles following various dosing regimens were simulated. Results Serum IgG levels, with significant inter-patient variability, correlated with dose and treatment interruptions at the individual patient level. Simulated data for various dosing regimens (0.4-2 g/kg once every 1-4 weeks [Q1-4W]) revealed that more frequent dosing provided more stable IgG levels than less frequent dosing, and dose splitting over multiple days had no significant effects on PK. Discussion In patients with MMN, stable dosing and consistent serum IgG levels are crucial to avoid negative responses owing to treatment interruptions. Dosing intervals more frequent than Q4W may alleviate periodic symptom deterioration. Dose splitting potentially offers flexibility for patients requiring large volumes of IVIG without negatively affecting serum IgG PK, while maintaining treatment efficacy. Variability in serum IgG levels between patients suggests that individualizing IVIG treatment regimens and target IgG levels may play a key role in managing MMN.
Collapse
Affiliation(s)
- Zhaoyang Li
- Global Clinical and Translational Sciences, Plasma-Derived Therapies, Takeda Development Center Americas, Inc., Cambridge, MA, United States
| | - Stefan Roepcke
- Pharmacometrics, Simulations Plus, Inc., Buffalo, NY, United States
| | - Ryan Franke
- Clinical Pharmacology, Cognigen Division of Simulations Plus, Inc., Buffalo, NY, United States
| | - Leman Yel
- Global Clinical Sciences, Takeda Development Center Americas, Inc., Cambridge, MA, United States
| |
Collapse
|
12
|
Cao M, Wu X, Li J. Steady-State Drug Exposure of Repeated IV Bolus Administration for a One Compartment Pharmacokinetic Model with Sigmoidal Hill Elimination. Bull Math Biol 2024; 86:143. [PMID: 39487871 DOI: 10.1007/s11538-024-01375-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Drugs exhibiting nonlinear pharmacokinetics hold significant importance in drug research and development. However, evaluating drug exposure accurately is challenging with the current formulae established for linear pharmacokinetics. This article aims to investigate the steady-state drug exposure for a one-compartment pharmacokinetic (PK) model with sigmoidal Hill elimination, focusing on three key topics: the comparison between steady-state drug exposure of repeated intravenous (IV) bolus ( AUC ss ) and total drug exposure after a single IV bolus ( AUC 0 - ∞ ); the evolution of steady-state drug concentration with varying dosing frequencies; and the control of drug pharmacokinetics in multiple-dose therapeutic scenarios. For the first topic, we established conditions for the existence of AUC ss , derived an explicit formula for its calculation, and compared it with AUC 0 - ∞ . For the second, we identified the trending properties of steady-state average and trough concentrations concerning dosing frequency. For the third, we developed formulae to compute dose and dosing time for both regular and irregular dosing scenarios. As an example, our findings were applied to a real drug model of progesterone used in lactating dairy cows. In conclusion, these results provide a theoretical foundation for designing rational dosage regimens and conducting therapeutic trials.
Collapse
Affiliation(s)
- Meizhu Cao
- School of Science, Shanghai Maritime University, Shanghai, 201306, People's Republic of China
| | - Xiaotian Wu
- School of Science, Shanghai Maritime University, Shanghai, 201306, People's Republic of China.
| | - Jun Li
- Faculté de Pharmacie, Université de Montréal, Montréal, QC, H3C 3J7, Canada.
- Centre de Recherches Mathématiques, Université de Montréal, Montréal, QC, H3C 3J7, Canada.
| |
Collapse
|
13
|
Wang Y, Ji J, Yao Y, Nie J, Xie F, Xie Y, Li G. Current status and challenges of model-informed drug discovery and development in China. Adv Drug Deliv Rev 2024; 214:115459. [PMID: 39389423 DOI: 10.1016/j.addr.2024.115459] [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: 05/28/2024] [Revised: 08/18/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
In the past decade, biopharmaceutical research and development in China has been notably boosted by government policies, regulatory initiatives and increasing investments in life sciences. With regulatory agency acting as a strong driver, model-informed drug development (MIDD) is transitioning rapidly from an academic pursuit to a critical component of innovative drug discovery and development within the country. In this article, we provided a cross-sectional summary on the current status of MIDD implementations across early and late-stage drug development in China, illustrated by case examples. We also shared insights into regulatory policy development and decision-making. Various modeling and simulation approaches were presented across a range of applications. Furthermore, the challenges and opportunities of MIDD in China were discussed and compared with other regions where these practices have a more established history. Through this analysis, we highlighted the potential of MIDD to enhance drug development efficiency and effectiveness in China's evolving pharmaceutical landscape.
Collapse
Affiliation(s)
- Yuzhu Wang
- Center for Drug Evaluation, National Medicine Products Administration, China
| | - Jia Ji
- Johnson & Johnson Innovative Medicine, Beijing, China
| | - Ye Yao
- Certara (Shanghai) Pharmaceutical Consulting Co., Ltd, Shanghai, China
| | - Jing Nie
- Abbisko Therapeutics Co., Ltd, Shanghai, China
| | - Fengbo Xie
- School of Data Science and Technology, North University of China, Taiyuan, China
| | - Yehua Xie
- Certara (Shanghai) Pharmaceutical Consulting Co., Ltd, Shanghai, China
| | - Gailing Li
- Certara (Shanghai) Pharmaceutical Consulting Co., Ltd, Shanghai, China.
| |
Collapse
|
14
|
Lu J, Zhao J, Xie D, Ding J, Yu Q, Wang T. Use of a PK/PD Model to Select Cetagliptin Dosages for Patients with Type 2 Diabetes in Phase 3 Trials. Clin Pharmacokinet 2024; 63:1463-1476. [PMID: 39367290 DOI: 10.1007/s40262-024-01427-7] [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: 09/15/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Cetagliptin is a novel dipeptidyl peptidase-4 (DPP-4) inhibitor developed for the treatment of patients with type 2 diabetes (T2D). Several phase 1 studies have been conducted in China. Modelling and simulation were used to obtain cetagliptin dose for phase 3 trials in T2D patients. METHODS A pharmacokinetic (PK)/pharmacodynamic (PD) model and model-based analysis of the relationship between hemoglobin A1c (HbA1c) and dosage was explored to guide dose selection of cetagliptin for phase 3 trials. The PK/PD data were derived from four phase 1 clinical studies, and sitagliptin 100 mg was employed as a positive control in studies 1, 3, and 4. RESULTS The PK profiles of cetagliptin were well described by a two-compartment model with first-order absorption, saturated efflux, and first-order elimination. The final PD model was a sigmoid maximum inhibitory efficacy (Emax) model with the Hill coefficient. The final model accurately captured cetagliptin PK/PD, demonstrated by goodness-of-fit plots. Based on weighted average inhibition (WAI), the relationship between HbA1c and dose was well displayed. Cetagliptin 50 mg once daily or above as monotherapy or as add-on therapy appeared more effective in HbA1c reduction than sitagliptin 100 mg. Cetagliptin 50 mg or 100 mg once daily was selected as the dose for phase 3 trials of cetagliptin in T2D patients. CONCLUSIONS The PK/PD model supports dose selection of cetagliptin for phase 3 trials. A model‑informed approach can be used to replace a dose-finding trial and accelerate cetagliptin's development.
Collapse
Affiliation(s)
- Jinmiao Lu
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China.
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China.
| | - Jiahong Zhao
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China
| | - Daosheng Xie
- Beijing Noahpharm Medical Technology Co., Ltd., Beijing, China
| | - Juping Ding
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China
| | - Qiang Yu
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China
| | - Tong Wang
- CGeneTech (Suzhou, China) Co., Ltd., 218 Xinghu Street, Suzhou, 215123, China.
| |
Collapse
|
15
|
Li X, Liu S, Liu D, Yu M, Wu X, Wang H. Application of Virtual Drug Study to New Drug Research and Development: Challenges and Opportunity. Clin Pharmacokinet 2024; 63:1239-1249. [PMID: 39225885 DOI: 10.1007/s40262-024-01416-w] [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: 08/15/2024] [Indexed: 09/04/2024]
Abstract
In recent years, virtual drug study, as an emerging research strategy, has become increasingly important in guiding and promoting new drug research and development. Researchers can integrate a variety of technical methods to improve the efficiency of all phases of new drug research and development, including the use of artificial intelligence, modeling and simulation for target identification, compound screening and pharmacokinetic characteristics evaluation, and the application of clinical trial simulation to carry out clinical research. This paper aims to elaborate on the application of virtual drug study in the key stages of new drug research and development and discuss the opportunities and challenges it faces in supporting new drug research and development.
Collapse
Affiliation(s)
- Xiuqi Li
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Shupeng Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Mengyang Yu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaofei Wu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongyun Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
16
|
Wojciechowski J, Mukherjee A, Banfield C, Nicholas T. Model-Informed Assessment of Probability of Phase 3 Success for Ritlecitinib in Patients with Moderate-to-Severe Ulcerative Colitis. Clin Pharmacol Ther 2024; 116:724-735. [PMID: 38627914 DOI: 10.1002/cpt.3251] [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: 04/21/2023] [Accepted: 03/02/2024] [Indexed: 08/22/2024]
Abstract
Ritlecitinib, an oral Janus kinase 3/tyrosine kinase expressed in hepatocellular carcinoma family inhibitor, was evaluated in patients with ulcerative colitis (UC) in a phase 2b trial. Model-informed drug development strategies were applied to bridge observations from phase 2b to predictions for a proposed phase 3 study design to assess the probability of achieving the target efficacy outcome. A longitudinal exposure-response model of the time course of the 4 Mayo subscores (rectal bleeding, stool frequency, physician's global assessment, and endoscopic subscore) in patients with UC receiving placebo or ritlecitinib was developed using population modeling approaches and an item response theory framework. The quantitative relationships between the 4 Mayo subscores accommodated the prediction of composite endpoints such as total Mayo score and partial Mayo score (key endpoints from phase 2b), and modified clinical remission and endoscopic remission (proposed phase 3 endpoints). Clinical trial simulations using the final model assessed the probability of candidate ritlecitinib dosing regimens (including those tested in phase 2b and alternative) and phase 3 study designs for achieving target efficacy outcomes benchmarked against an approved treatment for moderate-to-severe UC. The probabilities of achieving target modified clinical remission and endoscopic improvement outcomes at both weeks 8 and 52 for ritlecitinib 100 mg once daily was 74.8%. Model-based assessment mitigated some of the risk associated with proceeding to pivotal phase 3 trials with dosing regimens of which there was limited clinical experience.
Collapse
|
17
|
Madabushi R, Benjamin J, Zhu H, Zineh I. The US Food and Drug Administration's Model-Informed Drug Development Meeting Program: From Pilot to Pathway. Clin Pharmacol Ther 2024; 116:278-281. [PMID: 38445751 DOI: 10.1002/cpt.3228] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/07/2024] [Indexed: 03/07/2024]
Affiliation(s)
- Rajanikanth Madabushi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jessica Benjamin
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Issam Zineh
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
18
|
Yoshioka H, Jin R, Hisaka A, Suzuki H. Disease progression modeling with temporal realignment: An emerging approach to deepen knowledge on chronic diseases. Pharmacol Ther 2024; 259:108655. [PMID: 38710372 DOI: 10.1016/j.pharmthera.2024.108655] [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: 01/31/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
The recent development of the first disease-modifying drug for Alzheimer's disease represents a major advancement in dementia treatment. Behind this breakthrough is a quarter century of research efforts to understand the disease not by a particular symptom at a given moment, but by long-term sequential changes in multiple biomarkers. Disease progression modeling with temporal realignment (DPM-TR) is an emerging computational approach proposed with this biomarker-based disease concept. By integrating short-term clinical observations of multiple disease biomarkers in a data-driven manner, DPM-TR provides a way to understand the progression of chronic diseases over decades and predict individual disease stages more accurately. DPM-TR has been developed primarily in the area of neurodegenerative diseases but has recently been extended to non-neurodegenerative diseases, including chronic obstructive pulmonary, autoimmune, and ophthalmologic diseases. This review focuses on opportunities for DPM-TR in clinical practice and drug development and discusses its current status and challenges.
Collapse
Affiliation(s)
- Hideki Yoshioka
- Office of Regulatory Science Research, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan; Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ryota Jin
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan.
| | - Hiroshi Suzuki
- Executive Director, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan; Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Kowthavarapu VK, Charbe NB, Gupta C, Iakovleva T, Stillhart C, Parrott NJ, Schmidt S, Cristofoletti R. Mechanistic Modeling of In Vitro Biopharmaceutic Data for a Weak Acid Drug: A Pathway Towards Deriving Fundamental Parameters for Physiologically Based Biopharmaceutic Modeling. AAPS J 2024; 26:44. [PMID: 38575716 DOI: 10.1208/s12248-024-00912-y] [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: 12/14/2023] [Accepted: 03/17/2024] [Indexed: 04/06/2024] Open
Abstract
Mechanistic modeling of in vitro experiments using metabolic enzyme systems enables the extrapolation of metabolic clearance for in vitro-in vivo predictions. This is particularly important for successful clearance predictions using physiologically based pharmacokinetic (PBPK) modeling. The concept of mechanistic modeling can also be extended to biopharmaceutics, where in vitro data is used to predict the in vivo pharmacokinetic profile of the drug. This approach further allows for the identification of parameters that are critical for oral drug absorption in vivo. However, the routine use of this analysis approach has been hindered by the lack of an integrated analysis workflow. The objective of this tutorial is to (1) review processes and parameters contributing to oral drug absorption in increasing levels of complexity, (2) outline a general physiologically based biopharmaceutic modeling workflow for weak acids, and (3) illustrate the outlined concepts via an ibuprofen (i.e., a weak, poorly soluble acid) case example in order to provide practical guidance on how to integrate biopharmaceutic and physiological data to better understand oral drug absorption. In the future, we plan to explore the usefulness of this tutorial/roadmap to inform the development of PBPK models for BCS 2 weak bases, by expanding the stepwise modeling approach to accommodate more intricate scenarios, including the presence of diprotic basic compounds and acidifying agents within the formulation.
Collapse
Affiliation(s)
- Venkata Krishna Kowthavarapu
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Nitin Bharat Charbe
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Churni Gupta
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Tatiana Iakovleva
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Cordula Stillhart
- Pharmaceutical Research & Development, Formulation & Process Development, F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - Neil John Parrott
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA.
| |
Collapse
|
20
|
Kirouac DC, Zmurchok C, Morris D. Making drugs from T cells: The quantitative pharmacology of engineered T cell therapeutics. NPJ Syst Biol Appl 2024; 10:31. [PMID: 38499572 PMCID: PMC10948391 DOI: 10.1038/s41540-024-00355-3] [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: 11/17/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Engineered T cells have emerged as highly effective treatments for hematological cancers. Hundreds of clinical programs are underway in efforts to expand the efficacy, safety, and applications of this immuno-therapeutic modality. A primary challenge in developing these "living drugs" is the complexity of their pharmacology, as the drug product proliferates, differentiates, traffics between tissues, and evolves through interactions with patient immune systems. Using publicly available clinical data from Chimeric Antigen Receptor (CAR) T cells, we demonstrate how mathematical models can be used to quantify the relationships between product characteristics, patient physiology, pharmacokinetics and clinical outcomes. As scientists work to develop next-generation cell therapy products, mathematical models will be integral for contextualizing data and facilitating the translation of product designs to clinical strategy.
Collapse
Affiliation(s)
- Daniel C Kirouac
- Notch Therapeutics, Vancouver, BC, Canada.
- The University of British Columbia, School of Biomedical Engineering, Vancouver, BC, Canada.
- Metrum Research Group, Tariffville, CT, USA.
| | | | | |
Collapse
|
21
|
Kasturi M, Mathur V, Gadre M, Srinivasan V, Vasanthan KS. Three Dimensional Bioprinting for Hepatic Tissue Engineering: From In Vitro Models to Clinical Applications. Tissue Eng Regen Med 2024; 21:21-52. [PMID: 37882981 PMCID: PMC10764711 DOI: 10.1007/s13770-023-00576-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 10/27/2023] Open
Abstract
Fabrication of functional organs is the holy grail of tissue engineering and the possibilities of repairing a partial or complete liver to treat chronic liver disorders are discussed in this review. Liver is the largest gland in the human body and plays a responsible role in majority of metabolic function and processes. Chronic liver disease is one of the leading causes of death globally and the current treatment strategy of organ transplantation holds its own demerits. Hence there is a need to develop an in vitro liver model that mimics the native microenvironment. The developed model should be a reliable to understand the pathogenesis, screen drugs and assist to repair and replace the damaged liver. The three-dimensional bioprinting is a promising technology that recreates in vivo alike in vitro model for transplantation, which is the goal of tissue engineers. The technology has great potential due to its precise control and its ability to homogeneously distribute cells on all layers in a complex structure. This review gives an overview of liver tissue engineering with a special focus on 3D bioprinting and bioinks for liver disease modelling and drug screening.
Collapse
Affiliation(s)
- Meghana Kasturi
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mrunmayi Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varadharajan Srinivasan
- Department of Civil Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
22
|
Lemaire V, Hu C, van der Graaf PH, Chang S, Wang W. No Recipe for Quantitative Systems Pharmacology Model Validation, but a Balancing Act Between Risk and Cost. Clin Pharmacol Ther 2024; 115:25-28. [PMID: 37943003 DOI: 10.1002/cpt.3082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023]
Affiliation(s)
- Vincent Lemaire
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Chuanpu Hu
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Steve Chang
- Immunetrics, Inc., Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
23
|
Di Stefano F, Rodrigues C, Galtier S, Guilleminot S, Robert V, Gasparini M, Saint-Hilary G. Incorporation of healthy volunteers data on receptor occupancy into a phase II proof-of-concept trial using a Bayesian dynamic borrowing design. Biom J 2023; 65:e2200305. [PMID: 37888795 DOI: 10.1002/bimj.202200305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 07/09/2023] [Accepted: 07/23/2023] [Indexed: 10/28/2023]
Abstract
Receptor occupancy in targeted tissues measures the proportion of receptors occupied by a drug at equilibrium and is sometimes used as a surrogate of drug efficacy to inform dose selection in clinical trials. We propose to incorporate data on receptor occupancy from a phase I study in healthy volunteers into a phase II proof-of-concept study in patients, with the objective of using all the available evidence to make informed decisions. A minimal physiologically based pharmacokinetic modeling is used to model receptor occupancy in healthy volunteers and to predict it in the patients of a phase II proof-of-concept study, taking into account the variability of the population parameters and the specific differences arising from the pathological condition compared to healthy volunteers. Then, given an estimated relationship between receptor occupancy and the clinical endpoint, an informative prior distribution is derived for the clinical endpoint in both the treatment and control arms of the phase II study. These distributions are incorporated into a Bayesian dynamic borrowing design to supplement concurrent phase II trial data. A simulation study in immuno-inflammation demonstrates that the proposed design increases the power of the study while maintaining a type I error at acceptable levels for realistic values of the clinical endpoint.
Collapse
Affiliation(s)
- Fulvio Di Stefano
- Dipartimento di Scienze Matematiche (DISMA) "Giuseppe Luigi Lagrange,", Politecnico di Torino, Torino, Italy
| | - Christelle Rodrigues
- Department of Quantitative Pharmacology, Institut de Recherches Internationales Servier, Suresnes, France
| | - Stephanie Galtier
- Department of Clinical Statistics, Institut de Recherches Internationales Servier, Suresnes, France
| | - Sandrine Guilleminot
- Department of Quantitative Pharmacology, Institut de Recherches Internationales Servier, Suresnes, France
| | - Veronique Robert
- Department of Clinical Statistics, Institut de Recherches Internationales Servier, Suresnes, France
| | - Mauro Gasparini
- Dipartimento di Scienze Matematiche (DISMA) "Giuseppe Luigi Lagrange,", Politecnico di Torino, Torino, Italy
| | - Gaelle Saint-Hilary
- Dipartimento di Scienze Matematiche (DISMA) "Giuseppe Luigi Lagrange,", Politecnico di Torino, Torino, Italy
- Department of Statistical Methodology, Saryga, Tournus, France
| |
Collapse
|
24
|
Zhang N, Chan ML, Li J, Brohawn PZ, Sun B, Vainshtein I, Roskos LK, Faggioni R, Savic RM. Combining pharmacometric models with predictive and prognostic biomarkers for precision therapy in Crohn's disease: A case study of brazikumab. CPT Pharmacometrics Syst Pharmacol 2023; 12:1945-1959. [PMID: 37691451 PMCID: PMC10725267 DOI: 10.1002/psp4.13044] [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: 04/08/2023] [Revised: 06/11/2023] [Accepted: 07/11/2023] [Indexed: 09/12/2023] Open
Abstract
Pharmacometric models were used to investigate the utility of biomarkers in predicting the efficacy (Crohn's Disease Activity Index [CDAI]) of brazikumab and provide a data-driven framework for precision therapy for Crohn's disease (CD). In a phase IIa trial in patients with moderate to severe CD, treatment with brazikumab, an anti-interleukin 23 monoclonal antibody, was associated with clinical improvement. Brazikumab treatment effect was determined to be dependent on the baseline IL-22 (BIL22) or baseline C-reactive protein (BCRP; predictive biomarkers), and placebo effect was found to be correlated with the baseline CDAI (a prognostic biomarker). A maximal total inhibition on CDAI input function of 50.6% and 42.4% was predicted for patients with extremely high BIL22 or BCRP, compared to a maximal total inhibition of 20.9% and 17.8% for patients with extremely low BIL22 or BCRP, respectively, which were mainly due to the placebo effect. We demonstrated that model-derived baseline biomarker levels that achieve 50% of maximum unbound systemic concentration of 22.8 pg/mL and 8.03 mg/L for BIL22 and BCRP as the cutoffs to select subpopulations can effectively identify high-response subgroup patients with improved separation of responders when compared to using the median values as the cutoff. This work exemplifies the utility of pharmacometrics to quantify biomarker-driven responses in biologic therapies and distinguish between predictive and prognostic biomarkers, complementing clinical efforts of identifying subpopulations with higher likelihood of response to brazikumab.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ming Liang Chan
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jing Li
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Philip Z. Brohawn
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceutical R&D, AstraZenecaGaithersburgMarylandUSA
| | - Bo Sun
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Inna Vainshtein
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Lorin K. Roskos
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Raffaella Faggioni
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Rada M. Savic
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
25
|
Ji L, Lu J, Gao L, Ying C, Sun J, Han J, Zhao W, Gao Y, Wang K, Zheng X, Xie D, Ding J, Zhao J, Yu Q, Wang T. Efficacy and safety of cetagliptin as monotherapy in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled phase 3 trial. Diabetes Obes Metab 2023; 25:3671-3681. [PMID: 37661308 DOI: 10.1111/dom.15261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023]
Abstract
AIM To assess the efficacy and safety of the dipeptidyl peptidase-4 inhibitor, cetagliptin, as monotherapy in Chinese patients with type 2 diabetes (T2D) and inadequate glycaemic control. MATERIALS AND METHODS In total, 504 eligible patients with T2D were enrolled and randomized to cetagliptin 50 mg once daily, cetagliptin 100 mg once daily or placebo at a ratio of 2:2:1 for 24 weeks of double-blind treatment, then all patients received cetagliptin 100 mg once daily for 28 weeks of open-label treatment. The primary efficacy endpoint was the change in HbA1c level from baseline at week 24. RESULTS After 24 weeks, HbA1c from baseline was significantly reduced with cetagliptin 50 mg (-1.08%) and cetagliptin 100 mg (-1.07%) compared with placebo (-0.35%). The placebo-subtracted HbA1c reduction was -0.72% with cetagliptin 50 mg and 100 mg. Patients with a baseline HbA1c of 8.5% or higher had a greater HbA1c reduction with cetagliptin than those patients with a baseline HbA1c of less than 8.5%. Both doses studied led to a significantly higher proportion of patients (42.3% with 100 mg and 45.0% with 50 mg) achieving an HbA1c of less than 7.0% compared with placebo (12.9%). Cetagliptin also significantly lowered fasting plasma glucose and 2-hour postmeal plasma glucose relative to placebo. The incidence of adverse experiences was similar between cetagliptin and placebo. No drug-related hypoglycaemia was reported. CONCLUSIONS Cetagliptin monotherapy was effective and well tolerated in Chinese patients with T2D who had inadequate glycaemic control on exercise and diet.
Collapse
Affiliation(s)
- Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Jinmiao Lu
- CGeneTech Co., Ltd, Suzhou, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Leili Gao
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Changjiang Ying
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jiao Sun
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jie Han
- Department of Endocrinology, Hebei Petro China Central Hospital, Langfang, China
| | - Wenhua Zhao
- Department of Endocrinology, Pepole's Hospital of Changzhi City, Changzhi, China
| | - Yunming Gao
- Department of Endocrinology, The Second Pepole's Hospital of Lianyungang, Lianyungang, China
| | - Kun Wang
- Department of Endocrinology, Nanjing Jiangning Hospital, Nanjing, China
| | - Xin Zheng
- Department of Endocrinology, Beijing Boai Hospital, Beijing, China
| | - Daosheng Xie
- Beijing Noahpharm Medical Technology Co., Ltd, Beijing, China
| | | | | | - Qiang Yu
- CGeneTech Co., Ltd, Suzhou, China
| | | |
Collapse
|
26
|
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
|
27
|
He W, Demas DM, Shajahan-Haq AN, Baumann WT. Modeling breast cancer proliferation, drug synergies, and alternating therapies. iScience 2023; 26:106714. [PMID: 37234088 PMCID: PMC10206440 DOI: 10.1016/j.isci.2023.106714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/12/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
Estrogen receptor positive (ER+) breast cancer is responsive to a number of targeted therapies used clinically. Unfortunately, the continuous application of targeted therapy often results in resistance, driving the consideration of combination and alternating therapies. Toward this end, we developed a mathematical model that can simulate various mono, combination, and alternating therapies for ER + breast cancer cells at different doses over long time scales. The model is used to look for optimal drug combinations and predicts a significant synergism between Cdk4/6 inhibitors in combination with the anti-estrogen fulvestrant, which may help explain the clinical success of adding Cdk4/6 inhibitors to anti-estrogen therapy. Furthermore, the model is used to optimize an alternating treatment protocol so it works as well as monotherapy while using less total drug dose.
Collapse
Affiliation(s)
- Wei He
- Program in Genetics, Bioinformatics, and Computational Biology, VT BIOTRANS, Virginia Tech, Blacksburg, VA 24061, USA
| | - Diane M. Demas
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Ayesha N. Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - William T. Baumann
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
28
|
Yamamoto H, Shanker R, Sugano K. Application of Population Balance Model to Simulate Precipitation of Weak Base and Zwitterionic Drugs in Gastrointestinal pH Environment. Mol Pharm 2023; 20:2266-2275. [PMID: 36929729 DOI: 10.1021/acs.molpharmaceut.3c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
The purpose of the present study was to evaluate whether the population balance model (PBM) could be a suitable model for the precipitation of weak base and zwitterionic drugs in the gastrointestinal pH environment. Five poorly soluble drugs were used as model drugs (dipyridamole, haloperidol, papaverine, phenazopyridine, and tosufloxacin). PBM consists of the equations for primary nucleation, secondary nucleation, and particle growth. Each equation has two empirical parameters. The pH shift (pH-dumping) precipitation test (pH 3.0 to 6.5) was used to determine the model parameters for each drug. It was difficult to determine all six parameters by simultaneously fitting them to the precipitation profiles. Therefore, the number of model parameters was reduced from six to three by neglecting the secondary nucleation process and applying a common exponent number for the particle growth equation. Despite reducing the parameter number, PBM appropriately described the precipitation profiles in the pH shift tests. The constructed PBM model was then used to predict the precipitation profiles in an artificial stomach-intestine transfer (ASIT) test. PBM appropriately predicted the precipitation profiles in the ASIT test. These results suggested that PBM can be a suitable model to represent the precipitation of weak base and zwitterionic drugs in the gastrointestinal pH environment for biopharmaceutics modeling and simulation.
Collapse
Affiliation(s)
- Hibiki Yamamoto
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Ravi Shanker
- Pfizer Worldwide Research, Development, and Medical, 280 Shennecossett Road, Groton, Connecticut 06340, United States
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
29
|
Zhao Y, Li D, Liu R, Yuan Y. Bayesian optimal phase II designs with dual-criterion decision making. Pharm Stat 2023. [PMID: 36871961 DOI: 10.1002/pst.2296] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 11/29/2022] [Accepted: 02/06/2023] [Indexed: 03/07/2023]
Abstract
The conventional phase II trial design paradigm is to make the go/no-go decision based on the hypothesis testing framework. Statistical significance itself alone, however, may not be sufficient to establish that the drug is clinically effective enough to warrant confirmatory phase III trials. We propose the Bayesian optimal phase II trial design with dual-criterion decision making (BOP2-DC), which incorporates both statistical significance and clinical relevance into decision making. Based on the posterior probability that the treatment effect reaches the lower reference value (statistical significance) and the clinically meaningful value (clinical significance), BOP2-DC allows for go/consider/no-go decisions, rather than a binary go/no-go decision. BOP2-DC is highly flexible and accommodates various types of endpoints, including binary, continuous, time-to-event, multiple, and coprimary endpoints, in single-arm and randomized trials. The decision rule of BOP2-DC is optimized to maximize the probability of a go decision when the treatment is effective or minimize the expected sample size when the treatment is futile. Simulation studies show that the BOP2-DC design yields desirable operating characteristics. The software to implement BOP2-DC is freely available at www.trialdesign.org.
Collapse
Affiliation(s)
- Yujie Zhao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel Li
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Berkeley Heights, New Jersey, USA
| | - Rong Liu
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Berkeley Heights, New Jersey, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
30
|
Soltantabar P, Lon HK, Parivar K, Wang DD, Elmeliegy M. Optimizing benefit/risk in oncology: Review of post-marketing dose optimization and reflections on the road ahead. Crit Rev Oncol Hematol 2023; 182:103913. [PMID: 36681205 DOI: 10.1016/j.critrevonc.2023.103913] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Oncology therapies shifted from chemotherapy to molecularly targeted agents and finally to the era of immune-oncology agents. In contrast to cytotoxic agents, molecularly targeted agents are more selective, exhibit a wider therapeutic window, and may maximally modulate tumor growth at doses lower than the maximum tolerated dose (MTD). However, first-in-patient oncology studies for molecularly targeted agents continued to evaluate escalating doses using limited number of patients per dose cohort assessing dose-limiting toxicities to identify the MTD which is commonly selected for further development adopting a 'more is better' approach that led to several post-marketing requirement (PMR) studies to evaluate alternative, typically lower, doses or dosing frequencies to optimize the benefit-risk profile. In this review, post-marketing dose optimization efforts were reviewed including those required by a regulatory pathway or voluntarily conducted by the sponsor to improve efficacy, safety, or method of administration. Lessons learned and future implications from this deep dive review are discussed considering the evolving regulatory landscape on dose optimization for oncology compounds.
Collapse
Affiliation(s)
| | - Hoi-Kei Lon
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | | - Diane D Wang
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | |
Collapse
|
31
|
Liu Z, Liu J, Xia M. A Bayesian three-tier quantitative decision-making framework for single arm studies in early phase oncology. J Biopharm Stat 2023; 33:60-76. [PMID: 35723946 DOI: 10.1080/10543406.2022.2089155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In early phase oncology drug development, single arm proof-of-concept (POC) studies are increasingly being used to drive the early decisions for future development of the drug. Decision-makings based on such studies, typically involving small sample size and early surrogate efficacy endpoints, are extremely challenging. In particular, given the tremendous competition in the development of immunotherapies, expedition of the most promising programs is desired. To this end, we have proposed a Bayesian three-tier approach to facilitate the decision-making process, inheriting all the benefits of Bayesian decision-making approaches and formally allowing the option of acceleration. With pre-specified Bayesian decision criteria, three types of decisions regarding the future development of the drug can be made: (1) terminating the program, (2) further investigation, considering totality of evidence or additional POC studies, and (3) accelerating the program. We further proposed a Bayesian adaptive three-tier (BAT) design, extending the decision-making approach to incorporate adaptive thresholds and allow for continuous monitoring of the study. We compare the performance of the proposed methods with some other existing methods through simulations.
Collapse
Affiliation(s)
- Zhuqing Liu
- Global Statistical Sciences and Advanced Analytics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jingyi Liu
- Global Statistical Sciences and Advanced Analytics, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Meng Xia
- Global Statistical Sciences and Advanced Analytics, Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
32
|
He W, Shajahan-Haq AN, Baumann WT. Mathematically Modeling the Effect of Endocrine and Cdk4/6 Inhibitor Therapies on Breast Cancer Cells. Methods Mol Biol 2023; 2634:337-355. [PMID: 37074587 PMCID: PMC11986823 DOI: 10.1007/978-1-0716-3008-2_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Mathematical modeling of cancer systems is beginning to be used to design better treatment regimens, especially in chemotherapy and radiotherapy. The effectiveness of mathematical modeling to inform treatment decisions and identify therapy protocols, some of which are highly nonintuitive, is because it enables the exploration of a huge number of therapeutic possibilities. Considering the immense cost of laboratory research and clinical trials, these nonintuitive therapy protocols would likely never be found by experimental approaches. While much of the work to date in this area has involved high-level models, which look simply at overall tumor growth or the interaction of resistant and sensitive cell types, mechanistic models that integrate molecular biology and pharmacology can contribute greatly to the discovery of better cancer treatment regimens. These mechanistic models are better able to account for the effect of drug interactions and the dynamics of therapy. The aim of this chapter is to demonstrate the use of ordinary differential equation-based mechanistic models to describe the dynamic interactions between the molecular signaling of breast cancer cells and two key clinical drugs. In particular, we illustrate the procedure for building a model of the response of MCF-7 cells to standard therapies used in the clinic. Such mathematical models can be used to explore the vast number of potential protocols to suggest better treatment approaches.
Collapse
Affiliation(s)
- Wei He
- Program in Genetics, Bioinformatics, and Computational Biology, VT BIOTRANS, Virginia Tech, Blacksburg, VA, USA.
| | - Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - William T Baumann
- Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
33
|
Benest J, Rhodes S, Evans TG, White RG. The Correlated Beta Dose Optimisation Approach: Optimal Vaccine Dosing Using Mathematical Modelling and Adaptive Trial Design. Vaccines (Basel) 2022; 10:1838. [PMID: 36366347 PMCID: PMC9693615 DOI: 10.3390/vaccines10111838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/16/2022] [Accepted: 10/28/2022] [Indexed: 12/02/2022] Open
Abstract
Mathematical modelling methods and adaptive trial design are likely to be effective for optimising vaccine dose but are not yet commonly used. This may be due to uncertainty with regard to the correct choice of parametric model for dose-efficacy or dose-toxicity. Non-parametric models have previously been suggested to be potentially useful in this situation. We propose a novel approach for locating optimal vaccine dose based on the non-parametric Continuous Correlated Beta Process model and adaptive trial design. We call this the 'Correlated Beta' or 'CoBe' dose optimisation approach. We evaluated the CoBe dose optimisation approach compared to other vaccine dose optimisation approaches using a simulation study. Despite using simpler assumptions than other modelling-based methods, we found that the CoBe dose optimisation approach was able to effectively locate the maximum efficacy dose for both single and prime/boost administration vaccines. The CoBe dose optimisation approach was also effective in finding a dose that maximises vaccine efficacy and minimises vaccine-related toxicity. Further, we found that these modelling methods can benefit from the inclusion of expert knowledge, which has been difficult for previous parametric modelling methods. This work further shows that using mathematical modelling and adaptive trial design is likely to be beneficial to locating optimal vaccine dose, ensuring maximum vaccine benefit and disease burden reduction, ultimately saving lives.
Collapse
Affiliation(s)
- John Benest
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Sophie Rhodes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Thomas G. Evans
- Vaccitech Ltd., The Schrodinger Building, Heatley Road, The Oxford Science Park, Oxford OX4 4GE, UK
| | - Richard G. White
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
34
|
Barrett JS, Nicholas T, Azer K, Corrigan BW. Role of Disease Progression Models in Drug Development. Pharm Res 2022; 39:1803-1815. [PMID: 35411507 PMCID: PMC9000925 DOI: 10.1007/s11095-022-03257-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022]
Abstract
The use of Disease progression models (DPMs) in Drug Development has been widely adopted across therapeutic areas as a method for integrating previously obtained disease knowledge to elucidate the impact of novel therapeutics or vaccines on disease course, thus quantifying the potential clinical benefit at different stages of drug development programs. This paper provides a brief overview of DPMs and the evolution in data types, analytic methods, and applications that have occurred in their use by Quantitive Clinical Pharmacologists. It also provides examples of how these models have informed decisions and clinical trial design across several therapeutic areas and at various stages of development. It briefly describes potential new applications of DPMs utilizing emerging data sources, and utilizing new analytic techniques, and discuss new challenges faced such as requiring description of multiple endpoints, rapid model development, application of machine learning-based analytics, and use of high dimensional and real-world data. Considerations for the continued evolution future of DPMs to serve as community-maintained expert systems are also provided.
Collapse
Affiliation(s)
- Jeffrey S. Barrett
- Rare Disease Cures Accelerator Data Analytics Platform, Critical Path Institute, Tuscon, AZ 85718 USA
| | - Tim Nicholas
- Global Product Development, Pfizer Inc, 445 Eastern Point Rd, Groton, CT 06340 USA
| | - Karim Azer
- Axcella Therapeutics, 840 Memorial Drive, Cambridge, MA 02139 USA
| | - Brian W. Corrigan
- Global Product Development, Pfizer Inc, 445 Eastern Point Rd, Groton, CT 06340 USA
| |
Collapse
|
35
|
Llanos-Paez C, Ambery C, Yang S, Beerahee M, Plan EL, Karlsson MO. Improved Confidence in a Confirmatory Stage by Application of Item-Based Pharmacometrics Model: Illustration with a Phase III Active Comparator-Controlled Trial in COPD Patients. Pharm Res 2022; 39:1779-1787. [PMID: 35233731 PMCID: PMC9314306 DOI: 10.1007/s11095-022-03194-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/09/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE The current study aimed to illustrate how a non-linear mixed effect (NLME) model-based analysis may improve confidence in a Phase III trial through more precise estimates of the drug effect. METHODS The FULFIL clinical trial was a Phase III study that compared 24 weeks of once daily inhaled triple therapy with twice daily inhaled dual therapy in patients with chronic obstructive pulmonary disease (COPD). Patient reported outcome data, obtained by using The Evaluating Respiratory Symptoms in COPD (E-RS:COPD) questionnaire, from the FULFIL study were analyzed using an NLME item-based response theory model (IRT). The change from baseline (CFB) in E-RS:COPD total score over 4-week intervals for each treatment arm was obtained using the IRT and compared with published results obtained with a mixed model repeated measures (MMRM) analysis. RESULTS The IRT included a graded response model characterizing item parameters and a Weibull function combined with an offset function to describe the COPD symptoms-time course in patients receiving either triple therapy (n = 907) or dual therapy (n = 894). The IRT improved precision of the estimated drug effect compared to MMRM, resulting in a sample size of at least 3.64 times larger for the MMRM analysis to achieve the IRT precision in the CFB estimate. CONCLUSION This study shows the advantage of IRT over MMRM with a direct comparison of the same primary endpoint for the two analyses using the same observed clinical trial data, resulting in an increased confidence in Phase III.
Collapse
Affiliation(s)
- Carolina Llanos-Paez
- Department of Pharmacy, Uppsala University, BMC, Box 580, 751 23, Uppsala, Sweden
| | - Claire Ambery
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline plc, London, UK
| | - Shuying Yang
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline plc, London, UK
| | - Misba Beerahee
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline plc, London, UK
| | - Elodie L Plan
- Department of Pharmacy, Uppsala University, BMC, Box 580, 751 23, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmacy, Uppsala University, BMC, Box 580, 751 23, Uppsala, Sweden.
| |
Collapse
|
36
|
San-Martín-Martínez D, Serrano-Lemus D, Cornejo V, Gajardo AIJ, Rodrigo R. Pharmacological Basis for Abrogating Myocardial Reperfusion Injury Through a Multi-Target Combined Antioxidant Therapy. Clin Pharmacokinet 2022; 61:1203-1218. [DOI: 10.1007/s40262-022-01151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2022] [Indexed: 11/29/2022]
|
37
|
Applications of Model Informed Drug Development (MIDD) in Drug Development Lifecycle and Regulatory Review. Pharm Res 2022; 39:1663-1667. [PMID: 35790617 DOI: 10.1007/s11095-022-03327-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
|
38
|
Okwuokenye M. Quantitative Decision Under Unequal Covariances and Post-Treatment Variances: A Kidney Disease Application. Stat Biopharm Res 2022. [DOI: 10.1080/19466315.2020.1864464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
39
|
Azer K, Barrett JS. Quantitative system pharmacology as a legitimate approach to examine extrapolation strategies used to support pediatric drug development. CPT Pharmacometrics Syst Pharmacol 2022; 11:797-804. [PMID: 35411657 PMCID: PMC9286717 DOI: 10.1002/psp4.12801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 11/15/2022] Open
Abstract
Extrapolation strategies from adult data for designing pediatric drug development programs are explored using the quantitative systems pharmacology (QSP) modeling approach, a mechanistic drug and disease modeling framework that can predict clinical response and guide pediatric drug development in general. This innovative model‐informed drug discovery and development approach can leverage adult‐pediatric pharmacology and disease similarity metrics to validate extrapolation assumptions. We describe the QSP model strategy and framework for extrapolation to design pediatric drug development programs by leveraging adult data across a wide range of therapeutic areas and illustrating stage‐gate decisions informed by such an approach.
Collapse
Affiliation(s)
- Karim Azer
- Axcella Therapeutics Cambridge Massachusetts USA
| | | |
Collapse
|
40
|
Masters JC, Cook JA, Anderson G, Nucci G, Colzi A, Hellio MP, Corrigan B. Ensuring diversity in clinical trials: The role of clinical pharmacology. Contemp Clin Trials 2022; 118:106807. [DOI: 10.1016/j.cct.2022.106807] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/16/2022] [Accepted: 05/21/2022] [Indexed: 01/16/2023]
|
41
|
Marcantonio DH, Matteson A, Presler M, Burke JM, Hagen DR, Hua F, Apgar JF. Early Feasibility Assessment: A Method for Accurately Predicting Biotherapeutic Dosing to Inform Early Drug Discovery Decisions. Front Pharmacol 2022; 13:864768. [PMID: 35754500 PMCID: PMC9214263 DOI: 10.3389/fphar.2022.864768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
The application of model-informed drug discovery and development (MID3) approaches in the early stages of drug discovery can help determine feasibility of drugging a target, prioritize between targets, or define optimal drug properties for a target product profile (TPP). However, applying MID3 in early discovery can be challenging due to the lack of pharmacokinetic (PK) and pharmacodynamic (PD) data at this stage. Early Feasibility Assessment (EFA) is the application of mechanistic PKPD models, built from first principles, and parameterized by data that is readily available early in drug discovery to make effective dose predictions. This manuscript demonstrates the ability of EFA to make accurate predictions of clinical effective doses for nine approved biotherapeutics and outlines the potential of extending this approach to novel therapeutics to impact early drug discovery decisions.
Collapse
|
42
|
Zhang Y, Wo SK, Leng W, Gao F, Yan X, Zuo Z. Population pharmacokinetics and IVIVC for mesalazine enteric-coated tablets. J Control Release 2022; 346:275-288. [PMID: 35461968 DOI: 10.1016/j.jconrel.2022.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/17/2022] [Accepted: 04/15/2022] [Indexed: 10/18/2022]
Abstract
Although in-vivo bioequivalence (BE) study serves as a golden standard for establishing interchangeability of oral dosage forms, it remains challenging for products with high inter-subject variability such as mesalazine enteric-coated tablet to fulfil the BE criteria set by regulatory authorities. Mesalazine, as a BCS class IV drug, targets to be delivered to distal ileum or colon with a pH-sensitive polymer coating for the remission of ulcerative colitis. Through population pharmacokinetic (PK) analysis and in-vitro in-vivo correlation (IVIVC) modeling on the dissolution and BE data of a generic enteric-coated product (EM) and its reference Salofalk® 250 mg tablet (SM), we for the first time revealed the underlying mechanism of the high inter-subject variability for such delayed-release formulation. It was also noted that the in-vivo start time of absorption (Ts) for EM and SM was positively correlated with their in-vitro lag time (Tlag) under the USP three-stage dissolution condition and reversely correlated with their in-vivo bioavailability. The varied oral bioavailability of mesalazine enteric-coated tablet was mainly due to the varied N-acetyltransferase activities along GI tract. Although such extensive intestinal first-pass metabolism with large individual differences led to a significant variation of mesalazine Cmax (coefficient of variation: 60-63.5%) and AUC0-t (coefficient of variation: 37.5-46.9%), the corresponding variations in the total absorbed mesalazine (mesalazine and its metabolite N-acetyl mesalazine) were significantly reduced by 12 to 45%. Since the BE purpose for mesalazine enteric-coated tablet focused on their comparable safety profiles, total absorbed mesalazine was recommended to be adopted for the development of the IVIVC model and BE evaluation for EM. All in all, our model-based approach has not only successfully identified the key factors that affect the BE of EM to guide its further formulation optimization, but also demonstrated the indispensable role of modeling in the development of generic pharmaceutical product at its early stages.
Collapse
Affiliation(s)
- Yufeng Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Siu Kwan Wo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Wei Leng
- Europharm Laboratoires Co. Ltd., 12-14 Dai Wang Street, Tai Po Industrial Estate, Tai Po, New Territories, Hong Kong Special Administrative Region
| | - Fang Gao
- Europharm Laboratoires Co. Ltd., 12-14 Dai Wang Street, Tai Po Industrial Estate, Tai Po, New Territories, Hong Kong Special Administrative Region
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region.
| |
Collapse
|
43
|
Fernando K, Menon S, Jansen K, Naik P, Nucci G, Roberts J, Wu SS, Dolsten M. Achieving end-to-end success in the clinic: Pfizer's learnings on R&D productivity. Drug Discov Today 2022; 27:697-704. [PMID: 34922020 PMCID: PMC8719639 DOI: 10.1016/j.drudis.2021.12.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022]
Abstract
Over the past decade, Pfizer has focused efforts to improve its research and development (R&D) productivity. By the end of 2020, Pfizer had achieved an industry-leading clinical success rate of 21%, a tenfold increase from 2% in 2010 and well above the industry benchmark of ∼11%. The company had also maintained the quality of innovation, because 75% of its approvals between 2016 and 2020 had at least one expedited regulatory designation (e.g., Breakthrough Therapy). Pfizer's Signs of Clinical Activity (SOCA) paradigm enabled better decision-making and, along with other drivers (biology and modality), contributed to this productivity improvement. These laid a strong foundation for the rapid and effective development of the Coronavirus 2019 (COVID-19) vaccine with BioNTech, as well as the antiviral candidate Paxlovid™, under the company's 'lightspeed' paradigm.
Collapse
Affiliation(s)
- Kathy Fernando
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - Sandeep Menon
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - Kathrin Jansen
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - Prakash Naik
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - Gianluca Nucci
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - John Roberts
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - Shuang Sarah Wu
- Worldwide Research, Development and Medical, Pfizer Inc, United States
| | - Mikael Dolsten
- Worldwide Research, Development and Medical, Pfizer Inc, United States.
| |
Collapse
|
44
|
Broglio K, Marshall J, Yu B, Frewer P. Comparing Go/No-Go Decision-Making Properties Between Single Arm Phase II Trial Designs in Oncology. Ther Innov Regul Sci 2022; 56:291-300. [PMID: 34988927 DOI: 10.1007/s43441-021-00360-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/24/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Simon's design has been widely used in oncology to conduct single arm phase II trials and to make Go/No-Go development decision. Other authors have proposed designs with decision-making frameworks that include a third, "Consider" outcome. For results in the Consider zone, a final Go/No-Go development decision must still be made; however it is typically a subjective decision based on the totality of data and the development landscape. Under this framework, the probability of continuing development when the candidate therapy is truly ineffective or the probability of stopping development when the candidate therapy is truly effective is undefined. METHODS We use a motivating example to compare end of trial decision-making between Simon's two-stage approach and a Multilevel outcome approach. We present the minimum and maximum development decision error probabilities by varying whether candidates that end in the Consider zone would ultimately continue with development or not. RESULTS The Multilevel approach typically requires fewer patients, but the risk of making an incorrect drug development decision is inflated above the statistically defined Type I and Type II error rates. Compared to a Type I error rate of 20%, the Multilevel trial's maximum probability of moving forward with an ineffective therapy is 22%, 27%, and 36% for Consider zone sizes of 10%, 20%, and 30%, respectively. CONCLUSION The Multilevel approach provides flexibility in interpreting moderate efficacy results. However, the flexibility is accomplished with a lower sample size and corresponding uncertainty in the trial outcome that increases the risk of incorrect drug development decisions.
Collapse
Affiliation(s)
- Kristine Broglio
- Oncology Data Science and Analytics, AstraZeneca, 1 Medimmune Way, Gaithersburg, MD, 20878, USA.
| | - Jayne Marshall
- Early Oncology Statistics, AstraZeneca, Melbourn Science Park, Melbourn, UK
| | - Binbing Yu
- Oncology Data Science and Analytics, AstraZeneca, 1 Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Paul Frewer
- Early Oncology Statistics, AstraZeneca, Melbourn Science Park, Melbourn, UK
| |
Collapse
|
45
|
Golhen K, Winskill C, Yeh C, Zhang N, Welzel T, Pfister M. Value of Literature Review to Inform Development and Use of Biologics in Juvenile Idiopathic Arthritis. Front Pediatr 2022; 10:909118. [PMID: 35799700 PMCID: PMC9253535 DOI: 10.3389/fped.2022.909118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/24/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) is one of the most common pediatric inflammatory rheumatic diseases (PiRDs). Uncontrolled disease activity is associated with decreased quality of life and chronic morbidity. Biologic disease-modifying antirheumatic drugs (bDMARDs) and Janus kinase inhibitors (JAKi) have considerably improved clinical outcomes. For optimized patient care, understanding the efficacy-safety profile of biologics in subgroups of JIA is crucial. This systematic review based on published randomized controlled trials (RCTs) aims to assess efficacy and safety data for bDMARDs and JAKi with various JIA subgroups after 3 months of treatment. METHODS Data for American College of Rheumatology (ACR) pediatric (Pedi) 30, 50, and/or 70 responses after 3 months of treatment were selected from RCTs investigating bDMARDs or JAKi in JIA according to predefined inclusion/exclusion criteria. Treatment and control arms were compared by calculating risk ratios (RRs) with 95% confidence intervals (CIs), and proportions of overall, serious adverse events (AEs) and infections were analyzed. Forest plots were generated to summarize efficacy and safety endpoints across studies, JIA subgroups, and type of biologics. RESULTS Twenty-eight out of 41 PiRD RCTs investigated bDMARD or JAKi treatments in JIA. 9 parallel RCTs reported ACR Pedi 30, 50, and/or 70 responses 3 months after treatment initiation. All treatment arms showed improved ACR Pedi responses over controls. RRs ranged from 1.05 to 3.73 in ACR Pedi 30, from 1.20 to 7.90 in ACR Pedi 50, and from 1.19 to 8.73 in ACR Pedi 70. An enhanced effect for ACR Pedi 70 was observed with infliximab combined with methotrexate in PJIA vs. methotrexate monotherapy. A slightly higher risk of gastrointestinal AEs and infections was observed with treatment arms compared to placebo or methotrexate monotherapy. CONCLUSION Investigated bDMARDs and JAKi showed superior treatment responses compared to controls after 3 months of treatment, which were more pronounced in ACR Pedi 50 and 70 than in ACR Pedi 30. Higher susceptibility to infections associated with bDMARDs or JAKi vs. control arms must be weighed against efficacious treatment of the underlying disease and prevention of disease-related damage. Additional RCTs are warranted to further inform development and utilization of biologics in JIA.
Collapse
Affiliation(s)
- Klervi Golhen
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Carolyn Winskill
- Integrated Drug Development, Certara LP, Princeton, NJ, United States
| | - Cynthia Yeh
- Integrated Drug Development, Certara LP, Princeton, NJ, United States
| | - Nancy Zhang
- Integrated Drug Development, Certara LP, Princeton, NJ, United States
| | - Tatjana Welzel
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.,Pediatric Rheumatology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.,Integrated Drug Development, Certara LP, Princeton, NJ, United States
| |
Collapse
|
46
|
Kaushik AC, Sahi S, Wei DQ. Computational Methods for Structure-Based Drug Design Through System Biology. Methods Mol Biol 2022; 2385:161-174. [PMID: 34888721 DOI: 10.1007/978-1-0716-1767-0_9] [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] [Indexed: 06/13/2023]
Abstract
The advances in computational chemistry and biology, computer science, structural biology, and molecular biology go in parallel with the rapid progress in target-based systems. This technique has become a powerful tool in medicinal chemistry for the identification of hit molecules. The recent developments in target-based systems have played a major role in the creation of libraries of compounds, and it has also been widely applied for the design of molecular docking methods. The main advantage of this method is that it hits the fragment that has the strongest binding, has relatively small size, and leads to better compounds in terms of pharmacokinetic properties when compared with virtual screening (VS) and high-throughput screening (HTS) hits. De novo design is an essential aspect of target-based systems and requires the synthesis of chemical to allow the design of promising compound.
Collapse
Affiliation(s)
| | - Shakti Sahi
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
- Peng Cheng Laboratory, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
47
|
De Simone A, Davani L, Montanari S, Tumiatti V, Avanessian S, Testi F, Andrisano V. Combined Methodologies for Determining In Vitro Bioavailability of Drugs and Prediction of In Vivo Bioequivalence From Pharmaceutical Oral Formulations. Front Chem 2021; 9:741876. [PMID: 34805090 PMCID: PMC8597939 DOI: 10.3389/fchem.2021.741876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/15/2021] [Indexed: 11/26/2022] Open
Abstract
With the aim of developing an in vitro model for the bioavailability (BA) prediction of drugs, we focused on the study of levonorgestrel (LVN) released by 1.5 mg generic and brand-name tablets. The developed method consisted in combining a standard dissolution test with an optimized parallel artificial membrane permeability assay (PAMPA) to gain insights into both drug release and gastrointestinal absorption. Interestingly, the obtained results revealed that the tablet standard dissolution test, combined with an optimized PAMPA, highlighted a significant decrease in the release (15 ± 0.01 μg min−1 vs 30 ± 0.01 μg min−1) and absorption (19 ± 7 × 10–6 ± 7 cm/s Pe vs 41 ± 15 × 10–6 cm/s Pe) profiles of a generic LVN tablet when compared to the brand-name formulation, explaining unbalanced in vivo bioequivalence (BE). By using this new approach, we could determine the actual LVN drug concentration dissolved in the medium, which theoretically can permeate the gastrointestinal (GI) barrier. In fact, insoluble LVN/excipient aggregates were found in the dissolution media giving rise to non-superimposable dissolution profiles between generic and brand-name LVN tablets. Hence, the results obtained by combining the dissolution test and PAMPA method provided important insights confirming that the combined methods can be useful in revealing crucial issues in the prediction of in vivo BE of drugs.
Collapse
Affiliation(s)
- A De Simone
- Department of Drug Science and Technology, University of Turin, Torino, Italy
| | - L Davani
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - S Montanari
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - V Tumiatti
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | | | - F Testi
- Valpharma International S.p.A., Rimini, Italy
| | - V Andrisano
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
48
|
Maier C, de Wiljes J, Hartung N, Kloft C, Huisinga W. A continued learning approach for model-informed precision dosing: updating models in clinical practice. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 11:185-198. [PMID: 34779144 PMCID: PMC8846635 DOI: 10.1002/psp4.12745] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 11/12/2022]
Abstract
Model-informed precision dosing (MIPD) is a quantitative dosing framework that combines prior knowledge on the drug-disease-patient system with patient data from therapeutic drug/biomarker monitoring (TDM) to support individualized dosing in ongoing treatment. Structural models and prior parameter distributions used in MIPD approaches typically build on prior clinical trials that involve only a limited number of patients selected according to some exclusion/inclusion criteria. Compared to the prior clinical trial population, the patient population in clinical practice can be expected to include also altered behavior and/or increased interindividual variability, the extent of which, however, is typically unknown. Here, we address the question of how to adapt and refine models on the level of the model parameters to better reflect this real-world diversity. We propose an approach for continued learning across patients during MIPD using a sequential hierarchical Bayesian framework. The approach builds on two stages to separate the update of the individual patient parameters from updating the population parameters. Consequently, it enables continued learning across hospitals or study centers, since only summary patient data (on the level of model parameters) need to be shared, but no individual TDM data. We illustrate this continued learning approach with neutrophil-guided dosing of paclitaxel. The present study constitutes an important step towards building confidence in MIPD and eventually establishing MIPD increasingly in everyday therapeutic use.
Collapse
Affiliation(s)
- Corinna Maier
- Institute of Mathematics, University of Potsdam, Germany.,Graduate Research Training Program PharMetrX: Pharmacometrics & Computational Disease Modelling, Freie Universität Berlin and University of Potsdam, Germany
| | - Jana de Wiljes
- Institute of Mathematics, University of Potsdam, Germany
| | - Niklas Hartung
- Institute of Mathematics, University of Potsdam, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Germany
| | | |
Collapse
|
49
|
Hampson LV, Holzhauer B, Bornkamp B, Kahn J, Lange MR, Luo WL, Singh P, Ballerstedt S, Cioppa GD. A New Comprehensive Approach to Assess the Probability of Success of Development Programs Before Pivotal Trials. Clin Pharmacol Ther 2021; 111:1050-1060. [PMID: 34762298 DOI: 10.1002/cpt.2488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/30/2021] [Indexed: 01/01/2023]
Abstract
The point at which clinical development programs transition from early phase to pivotal trials is a critical milestone. Substantial uncertainty about the outcome of pivotal trials may remain even after seeing positive early phase data, and companies may need to make difficult prioritization decisions for their portfolio. The probability of success (PoS) of a program, a single number expressed as a percentage reflecting the multitude of risks that may influence the final program outcome, is a key decision-making tool. Despite its importance, companies often rely on crude industry benchmarks that may be "adjusted" by experts based on undocumented criteria and which are typically misaligned with the definition of success used to drive commercial forecasts, leading to overly optimistic expected net present value calculations. We developed a new framework to assess the PoS of a program before pivotal trials begin. Our definition of success encompasses the successful outcome of pivotal trials, regulatory approval and meeting the requirements for market access as outlined in the target product profile. The proposed approach is organized in four steps and uses an innovative Bayesian approach to synthesize all relevant evidence. The new PoS framework is systematic and transparent. It will help organizations to make more informed decisions. In this paper, we outline the rationale and elaborate on the structure of the proposed framework, provide examples, and discuss the benefits and challenges associated with its adoption.
Collapse
Affiliation(s)
| | | | | | - Joseph Kahn
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | - Wen-Lin Luo
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | | | | |
Collapse
|
50
|
Song E, Lee W, Kim BH. Model-Based Approach for Designing an Efficient Bioequivalence Study for Highly Variable Drugs. Pharmaceuticals (Basel) 2021; 14:1101. [PMID: 34832883 PMCID: PMC8624447 DOI: 10.3390/ph14111101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 02/05/2023] Open
Abstract
The statistical procedures as outlined by the European Medicines Agency (EMA) and United States Food and Drug Administration (FDA) guidelines for bioequivalence testing of highly variable drugs (HVDs) are complex. Additionally, the sample size is affected by clinical study designs or practical real-world problems, such as dropout rate or study budget. To overcome these difficulties, we propose a model-based approach for the selection of a study design with a sample size that satisfies the bioequivalence criteria using simulation studies based on a pharmacokinetic (PK) model. The designed approach was implemented using a simulation procedure considering some conventionally measured factors, such as geometric mean ratio and within-subject coefficient of variation, with various PK information important in determining bioequivalence. All simulation results were assessed according to the EMA and FDA guidelines. Furthermore, power calculations from simulation results were interpreted with regard to PK characteristics and compared among 2 × 2, 3 × 3, and 2 × 4 crossover designs to determine the efficient design considering appropriate sample size and duration of the clinical study. The proposed approach can be applied to bioequivalence studies of all drugs. However, the current study was targeted at HVDs, which are highly likely to require detailed decision making for sample size and study design.
Collapse
Affiliation(s)
- Eunjung Song
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Medical Center, Seoul 02447, Korea;
| | - Woojoo Lee
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Korea
| | - Bo-Hyung Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Medical Center, Seoul 02447, Korea;
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|