Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Pharm Res ; 41(9): 1775-1786, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39231907

ABSTRACT

PURPOSE: The concept of a Design Space (DSp) was introduced in ICH Q8 as a tool within the quality-by-design (QbD) approach to pharmaceutical development with the intent of being globally applicable. However, there appears to be variance in the regulatory agency expectations in pharmaceutical product filing and implementation of DSp. This paper presents some of the current industry perspective on design space. METHODS: The Utilization of Design Space for Filings (UDSpF) Working Group in the Innovation and Quality (IQ) Consortium conducted a survey to establish a baseline for the current understanding of DSp among IQ member companies and assess the similarities and/or differences in strategies when filing a DSp. The survey focused on how IQ member companies approach DSp development, the primary drivers for the DSp, the presentation of the DSp in the filing, DSp verification and the benefits and flexibility anticipated and/or realized. RESULTS: A total of 14 responses were received and analyzed representing a small sample size but a large proportion of the innovator industry/large pharmaceutical companies. The survey results revealed that DSp is not yet a commonplace for small molecule drug products and may not even be utilized as much in large molecule drug products. The benefits of DSp, with respect to enhanced process understanding, are well understood by the sponsors; however, the benefits of filed DSp with respect to manufacturing flexibility are not fully realized in the commercial lifecycle of the product. There are also challenges in gaining consistent buy-in/value proposition for DSp among cross-functional teams within organizations. CONCLUSIONS: There are still gaps in design space implementation for its full benefit in the pharmaceutical industry. The WG has presented a unified view from member companies on the approach to DSp considering when/where the DSp experiments are conducted and on the extent of the DSp development proposed in a dossier.


Subject(s)
Drug Development , Drug Industry , Drug Industry/methods , Drug Development/methods , Drug Development/legislation & jurisprudence , Surveys and Questionnaires , Humans , Pharmaceutical Preparations/chemistry , Drug Design , Quality Control
2.
Clin Ther ; 46(8): e6-e14, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38981791

ABSTRACT

PURPOSE: Artificial intelligence (AI) refers to technology capable of mimicking human cognitive functions and has important applications across all sectors and industries, including drug development. This has considerable implications for the regulation of drug development processes, as it is expected to transform both the way drugs are brought to market and the systems through which this process is controlled. There is currently insufficient evidence in published literature of the real-world applications of AI. Therefore, this narrative review investigated, collated, and elucidated the applications of AI in drug development and its regulatory processes. METHODS: A narrative review was conducted to ascertain the role of AI in streamlining drug development and regulatory processes. FINDINGS: The findings of this review revealed that machine learning or deep learning, natural language processing, and robotic process automation were favored applications of AI. Each of them had considerable implications on the operations they were intended to support. Overall, the AI tools facilitated access and provided manageability of information for decision-making across the drug development lifecycle. However, the findings also indicate that additional work is required by regulatory authorities to set out appropriate guidance on applications of the technology, which has critical implications for safety, regulatory process workflow and product development costs. IMPLICATIONS: AI has adequately proven its utility in drug development, prompting further investigations into the translational value of its utility based on cost and time saved for the delivery of essential drugs.


Subject(s)
Artificial Intelligence , Drug Development , Drug Development/legislation & jurisprudence , Drug Development/methods , Humans , Deep Learning , Machine Learning , Natural Language Processing
3.
Clin Pharmacol Ther ; 116(4): 902-913, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38973127

ABSTRACT

The 2022 United States Food and Drug Administration (US FDA) draft guidance on diversity plan (DP), which will be implemented through the Diversity Action Plans by December 2025, under the 21st Century Cures Act, marks a pivotal effort by the FDA to ensure that registrational studies adequately reflect the target patient populations based on diversity in demographics and baseline characteristics. This white paper represents the culminated efforts of the International Consortium of Quality and Innovation (IQ) Diversity and Inclusion (D&I) Working Group (WG) to assess the implementation of the draft FDA guidance by members of the IQ consortium in the discipline of clinical pharmacology (CP). This article describes current practices in the industry and emphasizes the tools and techniques of quantitative pharmacology that can be applied to support the inclusion of a diverse population during global drug development, to support diversity and inclusion of underrepresented patient populations, in multiregional clinical trials (MRCTs). It outlines strategic and technical recommendations to integrate demographics, including age, sex/gender, race/ethnicity, and comorbidities, in multiregional phase III registrational studies, through the application of quantitative pharmacology. Finally, this article discusses the challenges faced during global drug development, which may otherwise limit the enrollment of a broader, potentially diverse population in registrational trials. Based on the outcomes of the IQ survey that provided the current awareness of diversity planning, it is envisioned that in the future, industry efforts in the inclusion of previously underrepresented populations during global drug development will culminate in drug labels that apply to the intended patient populations at the time of new drug application or biologics license application rather than through post-marketing requirements.


Subject(s)
Drug Development , Drug Industry , Pharmacology, Clinical , United States Food and Drug Administration , Humans , Drug Development/legislation & jurisprudence , Drug Development/methods , United States , Clinical Trials as Topic/legislation & jurisprudence , Cultural Diversity
5.
J Pediatric Infect Dis Soc ; 13(Supplement_2): S93-S102, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38995086

ABSTRACT

The United States (U.S.) Food and Drug Administration (FDA) oversees the safety and quality of drugs and vaccines that are used in the U.S. Administration of the FDA falls under the jurisdiction of the U.S. Department of Health and Human Services (HHS). The regulatory oversight of the FDA is complex and comprehensive, requiring the various roles and responsibilities to be divided across six main centers. The activities of two of these centers, the Center for Drug Evaluation and Research (CDER) and the Center for Biologics Evaluation and Research (CBER) are the primary focus of this review.


Subject(s)
Drug Approval , United States Food and Drug Administration , Vaccines , United States , Drug Approval/legislation & jurisprudence , Humans , Vaccines/therapeutic use , Drug Development/legislation & jurisprudence , United States Dept. of Health and Human Services
6.
Clin Pharmacol Ther ; 116(3): 577-591, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39072758

ABSTRACT

Optimized dosages provide a secure foundation for the development of well-tolerated and effective oncology drugs. Project Optimus, an initiative within the Oncology Center of Excellence, strives to reform the dosage optimization and dosage selection paradigm in oncology. This initiative stems from the availability of targeted drugs and from the demand for more tolerable dosages from patients, caregivers, and advocates. Reforming dosage optimization for oncology drugs requires a paradigm shift from the one employed for cytotoxic chemotherapy to one that understands and considers the unique attributes of targeted therapy, immunotherapy, and cellular therapy. Limited characterization of dosage during drug development may result in (1) patients not staying on a therapy long-term due to poor tolerability, (2) failure to establish positive benefit-risk in clinical trials for regulatory approval (3) withdrawal of drugs from the market (4) removal of indications of drugs from the market. Timely access to drugs is important for all patients with cancer, so it is vital to iteratively analyze all nonclinical and clinically relevant data at each stage of development and leverage quantitative approaches, innovative trial designs, and regulatory support to arrive at dosages with favorable benefit-risk. This review highlights the key challenges and opportunities to embracing this new paradigm and realizing the full potential of new oncology therapies.


Subject(s)
Antineoplastic Agents , Drug Approval , Drug Development , Neoplasms , Humans , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Drug Development/legislation & jurisprudence , Drug Development/methods , Neoplasms/drug therapy , Drug Approval/legislation & jurisprudence , Dose-Response Relationship, Drug , Drug Dosage Calculations
8.
Paediatr Drugs ; 26(5): 555-563, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38837008

ABSTRACT

For more than two decades, regulatory agencies throughout the world released guidelines, rules and laws to stimulate and assist in paediatric drug development. In 2014, the National Health and Family Planning Commission (now known as the National Health Commission, NHC) and five other departments in China jointly issued 'Several Opinions on Safeguarding Medication for Children', after which several policies and regulations were issued to implement the priority review and approval of paediatric medicinal products and support the development of new drugs, including new dosage forms and strengths, for children. A total of 172 special medicinal products for children were approved from 2018 to 2022. Since 2016, the NHC, together with relevant administrative departments, has formulated and issued four paediatric drug lists containing 129 medicinal products to encourage research and development. At present, approximately 25 of these drugs (at exactly the same dosage forms and strengths as on the lists) have been approved for marketing, including antitumour drugs and immunomodulators, nervous system drugs, drugs for mental disorders and drugs for rare diseases. In this review, we analysed the regulations issued for promoting paediatric drug development in China, including the priority review and approval system, technical guidelines, data protection and financial support policies and general profiles of paediatric drug approval, clinical trials and the addition of information for children in the labels of marketed medicinal products. Finally, we discussed the challenges and possible strategies in the research and development of paediatric drugs in China.


Subject(s)
Drug Development , Humans , China , Drug Development/legislation & jurisprudence , Child , Pediatrics/legislation & jurisprudence , Pediatrics/organization & administration , Drug Approval/legislation & jurisprudence
9.
Pediatrics ; 154(1)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38841764

ABSTRACT

Despite >1 in 5 children taking prescription drugs in the United States, off-label drug use is common. To increase the study of drugs in children, regulatory bodies have enacted legislation to incentivize and require pediatric drug studies. As a result of this legislation, novel trial approaches, and an increase in personnel with pediatric expertise, there have been numerous advancements in pediatric drug development. With this review, we aim to highlight developments in pediatric pharmacology over the past 6 years for the most common disease processes that may be treated pharmacologically by the pediatric primary care provider. Using information extracted from label changes between 2018 and 2023, the published literature, and Clinicaltrials.gov, we discuss advances across multiple therapeutic areas relevant to the pediatric primary care provider, including asthma, obesity and related disorders, mental health disorders, infections, and dermatologic conditions. We highlight instances in which new drugs have been developed on the basis of a deeper mechanistic understanding of illness and instances in which labels have been expanded in older drugs on the basis of newly available data. We then consider additional factors that affect pediatric drug use, including cost and nonpharmacologic therapies. Although there is work to be done, efforts focused on pediatric-specific drug development will increase the availability of evidence-based, labeled guidance for commonly prescribed drugs and improve outcomes through the safe and effective use of drugs in children.


Subject(s)
Off-Label Use , Primary Health Care , Humans , Child , Off-Label Use/legislation & jurisprudence , Pediatrics , United States , Drug Development/legislation & jurisprudence , Asthma/drug therapy
10.
Ther Innov Regul Sci ; 58(5): 845-854, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38736019

ABSTRACT

The ICH E17 guidelines (2014-2017) on Multiregional Clinical Trials (MRCT) was a joint effort by the regulators and industry to facilitate simultaneous global drug development and registration through taking a strategic approach for clinical trials. In other words, the objective was to reduce the time it takes to bringing medications to patients around the world through minimizing unnecessary duplication of local or regional studies, which may add the regulatory burden to cost and time of bringing new therapies to patients. Under the auspices of ICH, training materials were created and provided to various stakeholders. Despite the successful promotion of the benefits of ICH E17 MRCT guidelines across the different regions, the uptake of some concepts (e.g., pooling strategy) in the ICH E17 guidelines has been slow. This paper describes various factors which could affect the conduct of MRCT at a global level, including ambiguity in definition of "region" (in MRCT), new regulatory requirements to enroll a diverse patient population, the use of decentralized clinical trials, use of data sources other than randomized clinical trials (e.g., use of Real Word Data), and the impact of the COVID-19 pandemic on the conduct of MRCT.


Subject(s)
Drug Development , Drug Development/legislation & jurisprudence , Humans , Clinical Trials as Topic , Guidelines as Topic , Multicenter Studies as Topic , COVID-19
11.
Pharmaceut Med ; 38(3): 179-204, 2024 May.
Article in English | MEDLINE | ID: mdl-38730200

ABSTRACT

Children were often referred to as "therapeutic orphans" in the past due to different reasons such as ethical, regulatory, economic, scientific, etc., ones. They were exposed to avoidable risks while missing out on therapeutic advances. Pediatric patients have suffered from a lack of scientific and regulatory standards (e.g., proper drug testing, authorization of medicines for their use, etc.), although the pharmaceutical legislative framework, which ensures the high standards of safety, quality, and efficacy of medicinal products for use in adults, was developed primarily in response to past "drug disasters," mainly involving children. The adoption of pediatric regulatory initiatives first in the USA and then in Europe and other countries and regions has significantly changed the worldwide frameworks and permanently changed pediatric drug research and development. This article tries to give various perspectives with historical context, a review of the different challenges and opportunities as well as important stakeholders in pediatric drug development. The pediatric trial networks are probably the most important stakeholder that enables efficient patient recruitment, access to better resource utilization, and global collaboration of different stakeholders necessary for performing quality and well-designed clinical trials.


Subject(s)
Clinical Trials as Topic , Drug Development , Humans , Child , Drug Development/legislation & jurisprudence , Orphan Drug Production/legislation & jurisprudence
12.
Clin Pharmacol Ther ; 116(1): 42-51, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38698592

ABSTRACT

Cardiac safety regulatory guidance for drug development has undergone several monumental shifts over the past decade as technological advancements, analysis models and study best practices have transformed this landscape. Once, clinical proarrhythmic risk assessment of a new chemical entity (NCE) was nearly exclusively evaluated in a dedicated thorough QT (TQT) study. However, since the introduction of the International Council for Harmonisation (ICH) E14/S7B Q&A 5.1 and 6.1 TQT substitutions, drug developers are offered an alternative pathway to evaluate proarrhythmic risk during an ascending dose study in healthy volunteers or during a powered patient study, respectively. In addition, the findings as well as the manner in which nonclinical studies are conducted (i.e., utilizing best practices) can dictate the need for a positive control in the clinical study and/or affect the labeling outcome. Drug sponsors are now faced with the option of pursuing a dedicated TQT study or requesting a TQT substitution. Potential factors influencing the choice of pathway include the NCE mechanism of action, pharmacokinetic properties, and safety profile, as well as business considerations. This tutorial will highlight the regulatory framework for integrated arrhythmia risk prediction models to outline drug safety, delineate potential reasons why a TQT substitution request may be rejected and discuss when a standalone TQT is recommended.


Subject(s)
Arrhythmias, Cardiac , Long QT Syndrome , Humans , Risk Assessment/methods , Long QT Syndrome/chemically induced , Arrhythmias, Cardiac/chemically induced , Drug Development/legislation & jurisprudence , Drug Development/methods , Electrocardiography/drug effects , Clinical Trials as Topic/legislation & jurisprudence , Clinical Trials as Topic/methods , Drug-Related Side Effects and Adverse Reactions
13.
Clin Ther ; 46(5): 424-428, 2024 May.
Article in English | MEDLINE | ID: mdl-38677979

ABSTRACT

Pharmaceutical development is a highly regulated industry through numerous worldwide guidance, laws, and regulations. Issues related to the safety of pharmaceutical products have been the most common cause of withdrawals from the market, as well as restrictions on distribution and limitations on labeling. Collaboration (hereafter referred to as partnership) between pharmaceutical companies in drug development has been recognized as critically significant to maximize the efficiency of drug development. In general, pharmaceutical companies might benefit from partnering in conducting pharmacovigilance (PV) activities, resulting in enhanced safety monitoring, improved clinical outcomes, and support of optimal benefit-risk assessment. However, some challenges exist. Differences between partners in strategy, culture, and processes can impact the harmonization of safety practices and decision-making processes, necessitating open communications and consensus-building to effectively address safety concerns. Both successful and unsuccessful partnership attempts within the pharmaceutical industry provide valuable business cases and lessons for the future. This paper sheds light on some of the critical aspects of PV in partnerships within the pharmaceutical industry. It addresses issues of the benefits and risks of partnerships, regulatory/legal expectations, and best practices for safety teams' integration.


Subject(s)
Drug Industry , Pharmacovigilance , Humans , Cooperative Behavior , Drug Development/legislation & jurisprudence , Drug Industry/legislation & jurisprudence , Drug Industry/organization & administration , Drug-Related Side Effects and Adverse Reactions/prevention & control , Risk Assessment
14.
Clin Pharmacol Ther ; 116(2): 282-288, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38519861

ABSTRACT

A recent industry perspective published in this journal describes the benefits received by drug companies from participation in the MIDD Pilot Program. Along with the primary objectives of supporting good decision-making in drug development, there were substantial savings in time and development costs. Furthermore, many sponsors reported qualitative benefits such as new learnings and clarity on MIDD strategies and methodology that could be applied to other development programs. Based on the success of the Pilot Program, the FDA recently announced the continuation of the MIDD Paired Meeting Program as part of the Prescription Drug User Fee Act (PDUFA VII). In this report, we describe the collective experiences of industry participants in the MIDD Program to date, including all aspects of the process from meeting request submission to follow-up actions. The purpose is to provide future participants with information to optimize the value of the MIDD Program.


Subject(s)
Drug Development , Drug Industry , United States Food and Drug Administration , United States , Drug Development/legislation & jurisprudence , Drug Development/methods , Humans , Drug Industry/legislation & jurisprudence , Pilot Projects , Drug Approval
15.
Clin Pharmacol Ther ; 115(6): 1400-1407, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38429944

ABSTRACT

In recent years, there has been significant focus on China's new drug lag, but relevant research is limited. This study explores the reasons for drug lag by assessing the impact of reforms in China's drug review system, particularly focusing on the influence of clinical development strategies. This study selected drugs first launched in the United States between 2017 and 2022, examining absolute and relative lag between China and the first-launch country (including submission and review lag). These delays with drugs approved in the European Union and Japan during the same period were compared with uncover the roots of delays in China, further identifying potential factors that could reduce these delays. The results indicate that the National Medical Products Administration (NMPA) has a longer relative lag compared with the European Medicines Agency (EMA) and the Pharmaceuticals and Medical Devices Agency (PMDA). The submission lag time of the NMPA significantly surpasses that of the EMA and PMDA, whereas the review lag time of the NMPA exceeds that of the PMDA but falls short of the EMA. Focusing on clinical trial strategies, bridging trials and multiregional clinical trials (MRCTs) are typically required by the NMPA in East Asia, resulting in longer clinical delay time. Whereas the EMA and PMDA primarily require international MRCTs in Europe and America, with a clinical delay of < 5 months. It is evident that there is a significant gap in clinical trial durations between China and other countries. Further optimization of clinical trial management is necessary to address the lag for new drugs in China.


Subject(s)
Drug Approval , Drug Development , China , Humans , Drug Approval/legislation & jurisprudence , Drug Development/legislation & jurisprudence , United States , European Union , Clinical Trials as Topic/legislation & jurisprudence , Time Factors , Japan
16.
Clin Pharmacol Ther ; 116(1): 52-63, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38529786

ABSTRACT

Although platform trials have many benefits, the complexity of these designs may result not only in increased methodological but also regulatory and ethical challenges. These aspects were addressed as part of the IMI project EU Patient-Centric Clinical Trial Platforms (EU-PEARL). We reviewed the available guidelines on platform trials in the European Union and the United States. This is supported and complemented by feedback received from regulatory interactions with the European Medicines Agency and the US Food and Drug Administration. Throughout the project we collected the needs of all relevant stakeholders including ethics committees, regulators, and health technology assessment bodies through active dialog and dedicated stakeholder workshops. Furthermore, we focused on methodological aspects and where applicable identified the corresponding guidance. Learnings from the guideline review, regulatory interactions, and workshops are provided. Based on these, a master protocol template was developed. Issues that still need harmonization or clarification in guidelines or where further methodological research is needed are also presented. These include questions around clinical trial submissions in Europe, the need for multiplicity control across the whole master protocol, the use of non-concurrent controls, and the impact of different randomization schemes. Master protocols are an efficient and patient-centered clinical trial design that can expedite drug development. However, they can also introduce additional operational and regulatory complexities. It is important to understand the different requirements of stakeholders upfront and address them in the trial. While relevant guidance is increasing, early dialog with relevant stakeholders can help to further support such designs.


Subject(s)
Clinical Trials as Topic , Drug Development , European Union , United States Food and Drug Administration , Humans , Clinical Trials as Topic/legislation & jurisprudence , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , United States , Drug Development/legislation & jurisprudence , Drug Development/methods , Research Design , Guidelines as Topic , Technology Assessment, Biomedical/legislation & jurisprudence
19.
Pharm Stat ; 22(6): 1135-1140, 2023.
Article in English | MEDLINE | ID: mdl-37431704

ABSTRACT

The role and value of statistical contributions in drug development up to the point of health authority approval are well understood. But health authority approval is only a true 'win' if the evidence enables access and adoption into clinical practice. In today's complex and evolving healthcare environment, there is additional strategic evidence generation, communication, and decision support that can benefit from statistical contributions. In this article, we describe the history of medical affairs in the context of drug development, the factors driving post-approval evidence generation needs, and the opportunities for statisticians to optimize evidence generation for stakeholders beyond health authorities in order to ensure that new medicines reach appropriate patients.


Subject(s)
Drug Development , Drug Development/legislation & jurisprudence , Statistics as Topic
20.
Int J Health Serv ; 52(3): 363-371, 2022 07.
Article in English | MEDLINE | ID: mdl-35546103

ABSTRACT

The process of developing and marketing new pharmaceuticals in the United States is driven by a need to maximize returns to shareholders. This results all too often in the production of new medications that are expensive and of marginal value to patients and society. In line with our heightened awareness of the importance of social justice and public health-and in light of our government's alliance with private companies in bringing us COVID-19 vaccines-we need to reconsider how new pharmaceuticals are developed and distributed. Accordingly, we propose the creation of a new agency of the Food and Drug Administration (FDA) that would direct the whole process. This agency would fund the research and development of high-value medications, closely monitor the clinical studies of these new drugs, and manage their distribution at prices that are value-based, fair, and equitable.


Subject(s)
Drug Development , Drug Industry , United States Food and Drug Administration , COVID-19 Vaccines , Drug Development/legislation & jurisprudence , Drug Development/organization & administration , Humans , Marketing , Pharmaceutical Preparations , United States
SELECTION OF CITATIONS
SEARCH DETAIL