1
|
Chen K, Zhao Y, Liu M, Lin J, Liu R. DOD-Combo: Bayesian dose finding design in combination trials with meta-analytic-predictive prior. J Biopharm Stat 2024:1-18. [PMID: 38468381 DOI: 10.1080/10543406.2024.2325142] [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: 10/01/2023] [Accepted: 02/24/2024] [Indexed: 03/13/2024]
Abstract
Combination therapy, a treatment modality that involves multiple treatment agents, has become imperative for improving treatment effectiveness and addressing resistance in the field of oncology. However, determining the most effective dose for these combinations, particularly when dealing with intricate drug interactions and diverse toxicity patterns, presents a substantial challenge. This paper introduces a novel Bayesian dose-finding design for combination therapies with information borrowing, named the DOD-Combo design. Leveraging historical single-agent trials and the meta-analytic-predictive (MAP) power prior, our approach utilizes a copula-type model to connect individual drug priors with joint toxicity probabilities in combination treatments. The MAP power prior allows the integration of information from multiple historical trials, constructing informative priors for each agent. Extensive simulations confirm our method's superior performance compared to combination designs with no information borrowing. By adaptively incorporating historical data, our approach reduces sample sizes and enhances efficiency in selecting the maximum tolerated dose (MTD), effectively addressing the intricate challenges presented by combination trials.
Collapse
Affiliation(s)
- Kai Chen
- Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, USA
| | - Yunqi Zhao
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, USA
| | - Meizi Liu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, USA
| | - Jianchang Lin
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, USA
| | - Rachael Liu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, USA
| |
Collapse
|
2
|
Bhattamisra SK, Banerjee P, Gupta P, Mayuren J, Patra S, Candasamy M. Artificial Intelligence in Pharmaceutical and Healthcare Research. BIG DATA AND COGNITIVE COMPUTING 2023; 7:10. [DOI: 10.3390/bdcc7010010] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Artificial intelligence (AI) is a branch of computer science that allows machines to work efficiently, can analyze complex data. The research focused on AI has increased tremendously, and its role in healthcare service and research is emerging at a greater pace. This review elaborates on the opportunities and challenges of AI in healthcare and pharmaceutical research. The literature was collected from domains such as PubMed, Science Direct and Google scholar using specific keywords and phrases such as ‘Artificial intelligence’, ‘Pharmaceutical research’, ‘drug discovery’, ‘clinical trial’, ‘disease diagnosis’, etc. to select the research and review articles published within the last five years. The application of AI in disease diagnosis, digital therapy, personalized treatment, drug discovery and forecasting epidemics or pandemics was extensively reviewed in this article. Deep learning and neural networks are the most used AI technologies; Bayesian nonparametric models are the potential technologies for clinical trial design; natural language processing and wearable devices are used in patient identification and clinical trial monitoring. Deep learning and neural networks were applied in predicting the outbreak of seasonal influenza, Zika, Ebola, Tuberculosis and COVID-19. With the advancement of AI technologies, the scientific community may witness rapid and cost-effective healthcare and pharmaceutical research as well as provide improved service to the general public.
Collapse
Affiliation(s)
- Subrat Kumar Bhattamisra
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to Be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Priyanka Banerjee
- Department of Pharmaceutical Technology, School of Medical Sciences, Adamas University, Kolkata 700126, West Bengal, India
| | - Pratibha Gupta
- Department of Pharmaceutical Technology, School of Medical Sciences, Adamas University, Kolkata 700126, West Bengal, India
| | - Jayashree Mayuren
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Susmita Patra
- Department of Pharmaceutical Technology, School of Medical Sciences, Adamas University, Kolkata 700126, West Bengal, India
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
3
|
Zhao Y, Liu R, Takeda K. Incorporating historical information to improve dose optimization design with toxicity and efficacy endpoints: iBOIN-ET. Pharm Stat 2022; 22:440-460. [PMID: 36514849 DOI: 10.1002/pst.2281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/13/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
In modern oncology drug development, adaptive designs have been proposed to identify the recommended phase 2 dose. The conventional dose finding designs focus on the identification of maximum tolerated dose (MTD). However, designs ignoring efficacy could put patients under risk by pushing to the MTD. Especially in immuno-oncology and cell therapy, the complex dose-toxicity and dose-efficacy relationships make such MTD driven designs more questionable. Additionally, it is not uncommon to have data available from other studies that target on similar mechanism of action and patient population. Due to the high variability from phase I trial, it is beneficial to borrow historical study information into the design when available. This will help to increase the model efficiency and accuracy and provide dose specific recommendation rules to avoid toxic dose level and increase the chance of patient allocation at potential efficacious dose levels. In this paper, we propose iBOIN-ET design that uses prior distribution extracted from historical studies to minimize the probability of decision error. The proposed design utilizes the concept of skeleton from both toxicity and efficacy data, coupled with prior effective sample size to control the amount of historical information to be incorporated. Extensive simulation studies across a variety of realistic settings are reported including a comparison of iBOIN-ET design to other model based and assisted approaches. The proposed novel design demonstrates the superior performances in percentage of selecting the correct optimal dose (OD), average number of patients allocated to the correct OD, and overdosing control during dose escalation process.
Collapse
Affiliation(s)
- Yunqi Zhao
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Rachael Liu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| |
Collapse
|
4
|
He T, Liu R, Liu M, Lin J. PMED: Optimal Bayesian Platform Trial Design with Multiple Endpoints. J Biopharm Stat 2022; 32:567-581. [PMID: 36000260 DOI: 10.1080/10543406.2022.2080692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/11/2022] [Indexed: 10/18/2022]
Abstract
In oncology drug development, indication selection and optimal dose identification are the primary objectives for the early phase of clinical trials and could significantly impact the probability of success. Master protocols, e.g., basket trial, umbrella trial, and platform trial, have become popular in practice considering the connection of trial designs with multiple indications and treatment candidates. They also enable the optimization of operational resources and maximize the capability of data-driven decision-making. However, most of the available designs are developed with the efficacy endpoint only for treatment effect estimation and testing, without consideration of the safety end point. Thus, it often lacks a comprehensive quantitative framework to allow optimal treatment selection, which could put future development at risk. We propose an optimal Bayesian platform trial design with multiple end points (PMED) to characterize the overall benefit-risk profile. The design is further extended to allow treatment and indication selection within and across arms, with continuous monitoring on multiple interim analyses for futility. In addition, we propose dynamic borrowing across arms to increase the efficiency and accuracy of estimation given the level of similarity across arms. A hierarchical hypothesis structure is utilized to achieve optimal indication and treatment combination selection by controlling family-wise error. Through simulation studies, we show that PMED is a robust design under the studied scenarios with superb power and controlled family-wise error rate.
Collapse
Affiliation(s)
- Tian He
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, Indiana, USA
| | - Rachael Liu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Meizi Liu
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Jianchang Lin
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| |
Collapse
|
5
|
Pan J, Bunn V, Hupf B, Lin J. Bayesian Additive Regression Trees (BART) with covariate adjusted borrowing in subgroup analyses. J Biopharm Stat 2022; 32:613-626. [PMID: 35737650 DOI: 10.1080/10543406.2022.2089160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
It is crucial in clinical trials to investigate treatment effect consistency across subgroups defined by patient baseline characteristics. However, there may be treatment effect variability across subgroups due to small subgroup sample size. Various Bayesian models have been proposed to incorporate this variability when borrowing information across subgroups. These models rely on the underlying assumption that patients with similar characteristics will have similar outcomes to the same treatment. Patient populations within each subgroup must subjectively be deemed similar enough Pocock (1976) to borrow response information across subgroups. We propose utilizing the machine learning method of Bayesian Additive Regression Trees (BART) to provide a method for subgroup borrowing that does not rely on an underlying assumption of homogeneity between subgroups. BART is a data-driven approach that utilizes patient-level observations. The amount of borrowing between subgroups automatically adjusts as BART learns the covariate-response relationships. Modeling patient-level data rather than treating the subgroup as a single unit minimizes assumptions regarding homogeneity across subgroups. We illustrate the use of BART in this context by comparing performance from existing subgroup borrowing methods in a simulation study and a case study in non-small cell lung cancer. The application of BART in the context of subgroup analyses alleviates the need to subjectively choose how much information to borrow based on subgroup similarity. Having the amount of borrowing be analytically determined and controlled for based on the similarity of individual patient-level characteristics allows for more objective decision making in the drug development process with many other applications including basket trials.
Collapse
Affiliation(s)
- Jane Pan
- Department of Biostatistics, University of California, Los Angeles, California, USA
| | - Veronica Bunn
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Bradley Hupf
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Jianchang Lin
- Statistical and Quantitative Sciences, Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
de las Heras B, Bouyoucef‐Cherchalli D, Reeve L, Reichl A, Mandarino D, Flach S, Vidal L, van Brummelen EMJ, Steeghs N. Healthy volunteers in first-in-human oncology drug development for small molecules. Br J Clin Pharmacol 2022; 88:1773-1784. [PMID: 34558113 PMCID: PMC10234445 DOI: 10.1111/bcp.15092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
This review provides tools to consider the inclusion of healthy volunteers (HVs) in first-in-human (FIH) oncology clinical trials with small molecules, including targeted and immunomodulatory agents, a strategy that was not envisioned with classic chemotherapy. To enable an FIH oncology trial in HVs compared to cancer patients (CPs), a robust nonclinical package must be generated, which includes toxicokinetic and pharmacokinetic studies, as well as more extensive safety pharmacology, toxicology and genotoxicity studies. This strategy could provide an early clinical characterization of the pharmacokinetic parameters and clinical safety profile in the absence of comorbidities and concomitant medication. It also avoids the ethical issue of administrating subtherapeutic doses to CPs, and could potentially help to accelerate the timelines of clinical drug development for patient care. That being said, stakeholders involved in these studies need to proceed with caution, fully understand the regulatory guidance and thoroughly evaluate the benefits and risks. This paper serves to address the regulatory guidance and other considerations needed when using healthy volunteers in early oncology trials.
Collapse
Affiliation(s)
- Begoña de las Heras
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
- Madrid Medical Doctors AssociationMadridSpain
| | | | - Lesley Reeve
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Andreas Reichl
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Debra Mandarino
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Stephen Flach
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Laura Vidal
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | | | | |
Collapse
|
7
|
Kolluri S, Lin J, Liu R, Zhang Y, Zhang W. Machine Learning and Artificial Intelligence in Pharmaceutical Research and Development: a Review. AAPS J 2022; 24:19. [PMID: 34984579 PMCID: PMC8726514 DOI: 10.1208/s12248-021-00644-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022] Open
Abstract
Over the past decade, artificial intelligence (AI) and machine learning (ML) have become the breakthrough technology most anticipated to have a transformative effect on pharmaceutical research and development (R&D). This is partially driven by revolutionary advances in computational technology and the parallel dissipation of previous constraints to the collection/processing of large volumes of data. Meanwhile, the cost of bringing new drugs to market and to patients has become prohibitively expensive. Recognizing these headwinds, AI/ML techniques are appealing to the pharmaceutical industry due to their automated nature, predictive capabilities, and the consequent expected increase in efficiency. ML approaches have been used in drug discovery over the past 15–20 years with increasing sophistication. The most recent aspect of drug development where positive disruption from AI/ML is starting to occur, is in clinical trial design, conduct, and analysis. The COVID-19 pandemic may further accelerate utilization of AI/ML in clinical trials due to an increased reliance on digital technology in clinical trial conduct. As we move towards a world where there is a growing integration of AI/ML into R&D, it is critical to get past the related buzz-words and noise. It is equally important to recognize that the scientific method is not obsolete when making inferences about data. Doing so will help in separating hope from hype and lead to informed decision-making on the optimal use of AI/ML in drug development. This manuscript aims to demystify key concepts, present use-cases and finally offer insights and a balanced view on the optimal use of AI/ML methods in R&D.
Collapse
Affiliation(s)
- Sheela Kolluri
- Global Clinical & Real World Evidence Statistics, Global Biometrics, Teva Pharmaceuticals, 145 Brandywine Pkwy, PA, 19380, West Chester, USA.
| | - Jianchang Lin
- Statistical and Quantitative Science, Data Sciences Institute, Takeda Pharmaceutical Co. Limited, 300 Mass Ave, West Chester, PA, 19380, USA
| | - Rachael Liu
- Statistical and Quantitative Science, Data Sciences Institute, Takeda Pharmaceutical Co. Limited, 300 Mass Ave, West Chester, PA, 19380, USA
| | - Yanwei Zhang
- Statistical and Quantitative Science, Data Sciences Institute, Takeda Pharmaceutical Co. Limited, 300 Mass Ave, West Chester, PA, 19380, USA
| | - Wenwen Zhang
- Statistical and Quantitative Science, Data Sciences Institute, Takeda Pharmaceutical Co. Limited, 300 Mass Ave, West Chester, PA, 19380, USA
| |
Collapse
|
8
|
Li Q, Lin J, Lin Y. Adaptive design implementation in confirmatory trials: methods, practical considerations and case studies. Contemp Clin Trials 2020; 98:106096. [PMID: 32739496 DOI: 10.1016/j.cct.2020.106096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/13/2020] [Accepted: 07/27/2020] [Indexed: 10/23/2022]
Abstract
The rapidly changing drug development landscapes have brought unique challenges to sponsors in designing clinical trials in a faster and more efficient way. With the ability to accelerate development timeline, reduce redundant sample size, and select the right dose and patient population during the clinical trial, adaptive designs help to increase the probability of success of clinical trials and eventually contribute to bringing the promising drugs to patients earlier and fulfilling their unmet medical needs. Although extensive adaptive design methods have been proposed in recent years, a comprehensive review of how to implement adaptive design in the practical confirmatory trials is still lacking. In this paper, we will review the evolving history of adaptive designs, updates of newly released regulatory guidance and emerging practical adaptive designs, including but not limited to sample size re-estimation, seamless design and surrogate endpoint used in the interim analysis. Furthermore, we will discuss the current practice of adaptive design implementation by demonstrating a complex oncology seamless phase 2/3 adaptive design case study. Through this example, we will introduce the critical roles of each cross disciplinary function, communication process and important documents when adaptive designs are implemented in real-world setting.
Collapse
Affiliation(s)
- Qing Li
- Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA 02139, United States of America.
| | - Jianchang Lin
- Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA 02139, United States of America
| | - Yunzhi Lin
- Sanofi, 50 Binney Street, Cambridge, MA 02142, United States of America
| |
Collapse
|