View Article

  • Navigating Regulatory Challenges In Multi-Regional Clinical Trials: A Comprehensive Overview
  • Pharm. D, MS Quality Assurance Associate

Abstract

Multi-regional clinical trials (MRCTs) are pivotal in accelerating the development of new drugs and therapies, allowing for more comprehensive data collection across diverse populations. However, conducting MRCTs comes with significant regulatory challenges due to the varying legal, ethical, and procedural frameworks across different regions. This article provides a comprehensive overview of these challenges, focusing on regulatory harmonization efforts, ethical considerations, and patient safety concerns. We discuss the roles of regulatory agencies like the FDA, EMA, and PMDA in ensuring compliance, while exploring strategies for overcoming obstacles such as approval delays, differing trial requirements, and variations in Good Clinical Practice (GCP) standards. By identifying best practices and collaborative approaches, this review aims to guide sponsors and researchers in navigating the complex regulatory landscape of MRCTs, ultimately enhancing the efficiency and global reach of clinical trials.

Keywords

Clinical trial, Regulatory authority, Good clinical practice.

Introduction

The globalization of drug development has led to a growing reliance on multi-regional clinical trials (MRCTs), an essential strategy for pharmaceutical companies aiming to introduce innovative therapies to global markets 1. MRCTs offer several benefits, including the ability to gather data from diverse populations, expedite patient recruitment, and shorten the time required to bring drugs to market. However, these trials also present significant regulatory challenges that complicate efforts to achieve successful and timely outcomes 2. Regulatory bodies worldwide have differing standards regarding clinical trial protocols, patient safety, and data requirements, forcing sponsors to navigate a complex network of regulations to secure approvals across multiple jurisdictions 3. The complexity of MRCTs is further compounded by varying clinical trial regulations, ethical considerations, patient consent laws, and data transparency requirements between countries 4. Efforts to achieve regulatory harmonization, such as those led by the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), have made progress, but challenges remain in creating a unified framework for all stakeholders, including regulatory authorities, pharmaceutical companies, and research organizations. Additionally, disparities in healthcare infrastructure, cultural attitudes toward clinical research, and regional readiness to implement new regulations further complicate MRCTs 5. This article delves into the specific regulatory obstacles associated with MRCTs, addressing issues related to trial design, approval processes, data standardization, and post-trial evaluations. It also examines strategies to overcome these hurdles, emphasizing the need for stronger regulatory cooperation and innovative solutions to improve the efficiency and global relevance of clinical trials.

ICH E17 GUIDELINE 6:

The ICH E17 Guideline is an important document developed by the International Council for Harmonisation (ICH), aimed at facilitating the conduct of multi-regional clinical trials (MRCTs) for pharmaceutical development. It is titled "General Principles for Planning and Design of Multi-Regional Clinical Trials," and it was finalized in 2017.

Purpose:

The purpose of this guideline is to describe general principles for the planning and design of MRCTs with the aim of increasing the acceptability of MRCTs in global regulatory submissions. The guideline addresses strategic programme issues as well as issues that are specific to the planning and design of confirmatory MRCTs, and it should be used together with other ICH guidelines, including E5, E6, E8, E9, E10, and E18. To provide recommendations for designing MRCTs that will generate data acceptable across different regulatory regions. This allows pharmaceutical companies to pursue simultaneous drug approvals in multiple regions, improving efficiency. To promote consistency in clinical trial design and ensure that results from MRCTs can be applied across regions, making the clinical data more globally relevant. The guideline aims to improve the precision of drug evaluations by accounting for regional differences, such as genetic, environmental, and healthcare factors.

PRINCIPLES:

  • The strategic use of MRCTs in drug development can enhance efficiency by enabling concurrent submission of marketing applications and supporting regulatory decisions across regions, leading to faster global access to new drugs. While MRCTs are often favored for multinational regulatory submissions, careful attention should be given to regional variations that could affect the study’s interpretability.
  • Key intrinsic and extrinsic factors essential to a drug development program should be identified early and assessed during exploratory phases before confirmatory MRCT design. These factors must be monitored throughout the confirmatory trials to evaluate their impact on treatment outcomes.
  • MRCTs assume the treatment effect is applicable to the entire target population, including all regions involved. Strategically allocating sample sizes across regions enables assessment of this assumption.
  • Pre-determined pooling of regions or subpopulations can enhance flexibility in sample size distribution, ensuring consistent treatment effects and aiding regulatory decisions.
  • A unified primary analysis approach, accepted by all regulatory authorities, should guide hypothesis testing and overall treatment effect estimation.
  • Ensuring quality trial design and conduct across all regions per ICH E6 guidelines is vital for result interpretation. Effective communication between sponsors and regulatory bodies during MRCT planning is crucial to support a globally accepted study design.

Consideration:

1. Planning of MRCTs:

  • Consideration of Regional Differences:

When planning MRCTs, sponsors must consider potential regional differences that could affect the interpretation of the trial results, such as variations in medical practices, disease prevalence, or pharmacogenomics.

  • Involvement of Regulatory Authorities:

Early involvement and consultation with regulatory authorities from the regions involved in the MRCT are encouraged. This helps ensure that the study design meets regulatory requirements in all regions.

2. Study Design

  • Randomization and Control:

Randomized controlled trial designs remain the gold standard. The guideline emphasizes careful planning of the randomization process to ensure the reliability of results across different regions.

  • Population Representation:

The guideline recommends that the study population should reflect the diversity of regions included in the trial. This ensures that the results are generalizable to a wider population.

  • Sample Size:

The sample size for each region should be sufficient to evaluate the drug’s efficacy and safety across different regions. The guideline emphasizes using proper statistical methods to determine sample sizes for regional subgroups.

3. Consistency of Treatment

Effect Pooling of Data:

One of the key issues addressed in the guideline is the pooling of data from different regions. It encourages the use of proper statistical methods to demonstrate that the treatment effect is consistent across different regions.

Handling Regional Differences:

If regional differences are observed in the treatment effect, sponsors should provide a clear explanation of the factors contributing to the difference. Subgroup analyses may be necessary to identify the causes of variability.

4. Ethical and Practical Considerations

Ethical Standards:

MRCTs must be conducted according to internationally recognized ethical standards, such as those outlined in the Declaration of Helsinki, and must respect the rights and welfare of participants in all regions.

Practical Considerations:

The guideline discusses logistical issues that can arise in MRCTs, such as differences in regulatory timelines, infrastructure, and resource availability between regions. Careful planning is needed to overcome these practical barriers.

5. Data Analysis and Interpretation

Primary and Secondary Endpoints:

The guideline emphasizes the importance of clearly defining primary and secondary endpoints that are meaningful across regions. These endpoints should be agreed upon before the trial begins.

Handling Missing Data:

Strategies for dealing with missing data should be established early in the trial planning process to avoid biases in the analysis.

6. Regulatory Review

One of the goals of the guideline is to allow simultaneous submissions of clinical trial data to multiple regulatory authorities. The harmonized design of MRCTs supports this by ensuring that the data is acceptable across regions.

REGULATORY DIVERSITY ACROSS REGION:

In 1993, the U.S. Congress passed the NIH Revitalization Act, mandating the inclusion of women and racial and ethnic minority groups in all federally funded clinical research to improve the generalizability of findings 7. However, despite this directive, these groups remain underrepresented. For instance, Black individuals make up only 8.2% of participants in pancreatic cancer trials, even though they account for 12.4% of diagnoses. Similar disparities are seen across various medical fields, including cardiovascular studies and NIH-funded respiratory disease research, where fewer than 5% of studies from 1993-2013 reported minority inclusion 8. This trend continues, as only 58% of COVID-19 vaccine trials reported participants' race. Diversity in clinical research is critical for the validity and relevance of findings, as it ensures that results are applicable to the populations most affected by the conditions under study. The U.S. population is becoming more diverse, with Asian and Hispanic groups growing rapidly, as shown by the 2020 Census 9. To achieve scientific rigor and trust, clinical trials must mirror this demographic shift. Furthermore, racial and ethnic minorities often experience poorer health outcomes, making their inclusion in research essential for developing better-targeted therapies. Regulatory agencies, like the FDA, are addressing these gaps by issuing guidance to trial sponsors on enhancing diversity through broader eligibility and improved accessibility. Initiatives like the NIH's CEAL Against COVID-19 Disparities work to dismantle barriers such as misinformation and mistrust by engaging with community leaders. Additionally, the industry must overcome logistical barriers and focus on participant recruitment, retention, and engagement. Academic institutions are also called to foster community partnerships by understanding local perceptions and addressing community needs 10.

European Union:

Regulatory diversity across the European Union (EU) stems from the complex interplay between harmonized EU-wide regulations and individual member states' legal systems. While the EU has established unified frameworks through directives and regulations, particularly in areas such as pharmaceuticals, food safety, and environmental protection, each country retains some autonomy in implementation. This leads to variations in administrative practices, compliance requirements, and enforcement 11. For example, the European Medicines Agency (EMA) provides centralized procedures for drug approval, but national agencies may still handle certain aspects of market authorization or post-market surveillance. Differences can also arise in how member states interpret directives or integrate them into national law, leading to discrepancies in areas like taxation, labor laws, or consumer protection. These regional regulatory differences require companies and stakeholders operating across the EU to navigate a complex legal landscape, balancing uniform standards with localized nuances 12.

India:

Regulatory diversity in India arises due to the complex federal structure where both the central and state governments play significant roles in policy and regulation. At the national level, bodies like the Central Drugs Standard Control Organization (CDSCO) regulate the approval, import, and quality of drugs, while the Food Safety and Standards Authority of India (FSSAI) oversees food safety. However, states also have their own drug control departments and food safety officers, leading to regional variations in enforcement, interpretation of guidelines, and implementation of policies. Additionally, factors such as local governance dynamics, resource allocation, and regional economic conditions contribute to disparities in how regulations are applied across states. For example, certain states may have stricter rules for clinical trials, pharmaceutical manufacturing, or health services, creating a patchwork of regulatory environments. This diversity necessitates a thorough understanding of both national laws and regional regulations for compliance, especially in sectors like healthcare, pharmaceuticals, and food safety, where regulations impact public health directly 13.

POOLING STRATEGIES AND DATA CONSISTENCY:      

Pooling strategies in clinical trials involve the aggregation of data from various studies, trial sites, or different phases of a trial to improve statistical power and offer broader insights into the research question. This approach is commonly employed in meta-analyses or integrated analyses, where data from multiple sources are combined to evaluate the efficacy, safety, or other clinical outcomes of interventions across diverse populations. Pooling is especially beneficial when individual studies are underpowered or when larger sample sizes are needed to detect significant effects or rare outcomes 14. However, a major challenge in pooling data is ensuring consistency and comparability across datasets. This requires harmonizing study designs, standardizing variables such as inclusion criteria, treatment protocols, and outcome measures, and maintaining uniform data collection methods across sites or trial phases. If these elements are not carefully aligned, data heterogeneity can arise, potentially introducing biases and compromising the reliability of the pooled results. Statistical models like fixed-effects and random-effects are frequently used to manage variability across studies or populations. Furthermore, attention must be given to differences in patient populations, such as demographic factors and disease severity, as well as any variations in study settings or methodologies that could affect the comparability of results. Through meticulous standardization and alignment of methods, pooling strategies can yield more comprehensive and generalizable conclusions, ultimately leading to stronger, more reliable outcomes in clinical trials 15.

CULTURAL AND LOGISTICAL CHALLENGES IN MRCTS:

Multi-Regional Clinical Trials encounter a myriad of cultural and logistical challenges that can profoundly influence their planning, execution, and overall success. Culturally, researchers must navigate diverse healthcare practices, societal norms, and patient expectations that vary from region to region. These differences can significantly affect patient recruitment, as individuals from various backgrounds may have varying levels of trust in clinical research, influenced by past experiences, local healthcare practices, or societal attitudes towards medical interventions 16. Furthermore, language barriers can complicate effective communication, creating potential misunderstandings during the informed consent process and hindering participant comprehension of study protocols and potential risks. Cultural perceptions regarding clinical trials may also lead to differences in participant engagement and retention, with some cultures valuing personal relationships and trust over standard recruitment practices. Logistically, MRCTs require meticulous coordination across multiple countries, which introduces complexities related to regulatory compliance. Each participating region has its own regulatory body with distinct requirements, timelines, and approval processes, making it challenging to maintain a unified protocol while adhering to local laws 17. Variability in clinical trial infrastructure—such as the availability of medical resources, technological capabilities, and the presence of trained personnel—can lead to disparities in data collection and management. Moreover, differences in healthcare systems can influence the standard of care provided, which may impact the trial's outcomes. For instance, variations in access to medications, treatment protocols, and patient follow-up care can introduce confounding factors that compromise the validity of the study results. To address these multifaceted challenges, it is crucial for MRCT planners to adopt culturally sensitive strategies, foster local collaborations, and establish robust logistical frameworks that ensure consistency and quality across diverse study sites, ultimately leading to more reliable and generalizable findings. Ethical considerations in research encompass the protection of subjects, informed consent, research integrity, accessibility, transparency, and the quality of reviews by local Ethics Committees and Institutional Review Boards. They also involve ensuring data privacy, adherence to Good Clinical Practice (GCP) standards, compliance with relevant laws regarding ethical reviews, informed consent, and the protection of human participants in biomedical studies 18.

HARMONIZATION OF ETHICAL STANDARDS:

Harmonization of ethical standards in clinical trials refers to the process of aligning and standardizing the ethical guidelines and regulatory requirements across different countries and regions to ensure the protection of human subjects while promoting scientific integrity. This effort is critical due to the global nature of clinical research, where trials often involve participants from various jurisdictions, each with its own ethical frameworks and regulations. Harmonization aims to create a unified approach to informed consent, risk assessment, data privacy, and the equitable selection of participants, minimizing discrepancies that can arise from differing national laws. By fostering collaboration among international regulatory bodies, such as the International Conference on Harmonisation (ICH) and the World Health Organization (WHO), the goal is to enhance the efficiency of trial conduct, facilitate faster access to innovative treatments, and uphold the highest ethical standards to safeguard participants' rights and welfare across diverse cultural and legal landscapes 19-21.

REGULATORY COLLABORATION AND MUTUAL RECOGNITION:

Regulatory collaboration and mutual recognition are key strategies utilized by nations and regions to improve the efficiency and effectiveness of regulatory frameworks, particularly in sectors like pharmaceuticals, medical devices, and food safety. Regulatory collaboration involves the exchange of information, best practices, and resources among regulatory agencies, facilitating the harmonization of standards, the streamlining of approval procedures, and a collective response to global challenges, including emerging health threats. Conversely, mutual recognition entails the acceptance of regulatory decisions made by one jurisdiction by another, enabling products approved in one country to be acknowledged and marketed in another without the need for repetitive evaluations. This partnership can alleviate regulatory burdens, expedite the market entry of innovative products, and enhance consumer safety while fostering international trade and cooperation among regulatory authorities. Collectively, these strategies aim to establish a more integrated and responsive regulatory landscape that promotes public health and stimulates economic growth 22-23.

REAL-WORLD DATA (RWD) IN MRCTS:

Real-world data (RWD) encompasses information gathered outside conventional clinical trial settings, drawing from a wide array of sources, including electronic health records, insurance claims, patient registries, and patient-reported outcomes. In the context of clinical trials, RWD enhances the comprehension of treatment effects by offering insights into the performance of therapies across varied patient populations, which often include diverse demographics, comorbid conditions, and levels of treatment adherence that are typically not fully represented in controlled trial environments. The use of RWD enables researchers to discern patterns in treatment outcomes, evaluate long-term effects, and analyze the economic implications of interventions. Furthermore, incorporating RWD into clinical trials facilitates the design and execution of more pragmatic studies, promoting a thorough assessment of therapies that resonates with real-world healthcare practices. This approach ultimately supports regulatory decision-making and enhances patient care outcomes. Makady et al. found in RWD policies among six European HTA organizations, that all six accepted any source of data, including RWD, albeit with a certain hierarchy. Based on our results from 21 countries, it seems that other European countries may be more reluctant to accept RWD 24-27.

CONCLUSION:

In summary, multi-regional clinical trials face significant regulatory hurdles that can delay the development of new therapies. The complexities arising from differing regulations, ethical standards, data privacy issues, and diverse patient populations complicate the trial process and may affect the validity of findings. To mitigate these challenges, collaboration among stakeholders regulatory authorities, pharmaceutical companies, and research institutions is essential for creating streamlined and harmonized regulatory frameworks. Enhancing communication and understanding among regulatory bodies will facilitate smoother trial operations and improve the integrity of clinical research across regions, ultimately expediting the development of innovative therapies to address global health needs.

REFERENCES:

  1. Meekings K.N., Williams C.S., Fau-Arrowsmith J.E., Arrowsmith J.E. Orphan drug development: An economically viable strategy for biopharma R&D. Drug Discov. Today. 2012;17:660–664. doi: 10.1016/j.drudis.2012.02.005.
  2. Parker J. Who Has a Drug Lag? Manag. Decis. Econ. 1989;10:299–309. doi: 10.1002/mde.4090100407.  
  3. Vogel D. The Globalization of Pharmaceutical Regulation. Governance. 1998;11:1–22. doi: 10.1111/0952-1895.551998055.     
  4. Andersson F. The drug lag issue: The debate seen from an international perspective. Int. J. Health Serv. 1992;22:53–72. doi: 10.2190/9Y32-X86Y-M3F0-JQFC.
  5. Binkowitz B, Ibia E. Multiregional clinical trials: An introduction from an industry perspective. Ther Innov Regul Sci. 2011;45:569–73
  6. Guideline I.H. General Principles for Planning and Design of Multi-Regional Clinical Trials E17; The International Council for Harmonisation. 2017. [(accessed on 9 February 2021)]. Available online: https://www.ich.org/page/efficacy-guidelines 
  7. National Institutes of Health. NIH policy and guidelines on the inclusion of women and minorities as subjects in clinical research. https://grants.nih.gov/policy/inclusion/women-and-minorities/guidelines.htm
  8. Vespa J, Armstrong DM, Medina L. Demographic turning points for the United States: Population projections for 2020 to 2060: US Department of Commerce, Economics and Statistics Administration. https://www.census.gov/content/dam/Census/library/publications/2020/demo/p25-1144.pdf
  9. U.S. Food and Drug Administration. Enhancing the Diversity of Clinical Trial Populations — Eligibility Criteria, Enrollment Practices, and Trial Designs Guidance for Industry. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enhancing-diversity-clinical-trial-populations-eligibility-criteria-enrollment-practices-and-trial
  10. Woodcock J, Araojo R, Thompson T, Puckrein GA. Integrating Research into Community Practice — Toward Increased Diversity in Clinical Trials. N Engl J Med. 385 (2021) 1351–1353.
  11. Molnár-Gábor F, Sellner J, Pagil S, Slokenberga S, Tzortzatou-Nanopoulou O, Nyström K. Harmonization after the GDPR? Divergences in the rules for genetic and health data sharing in four member states and ways to overcome them by EU measures: Insights from Germany, Greece, Latvia and Sweden. Semin Cancer Biol. 2022 Sep;84:271-283. doi: 10.1016/j.semcancer.2021.12.001
  12. Assessment of the EU Member States’ Rules on Health Data in the Light of GDPR.  Available at https://health.ec.europa.eu/publications/assessment-eu-member-states-rules-health-data-light-gdpr_en
  13. Central Drugs Standard Control Organization. Available in https://cdsco.gov.in/opencms/opencms/en/Home/
  14. Abraham Yeh, Xiaohong (Grace) Zhang, Shin-Ru Wang, Novartis Pharmaceuticals Corporation, East Hanover, NJ. Pooling strategy of clinical data. Available in https://www.lexjansen.com/nesug/nesug13/21_Final_Paper.pdf
  15. Bangdiwala SI, Bhargava A, O'Connor DP, Robinson TN, Michie S, Murray DM, Stevens J, Belle SH, Templin TN, Pratt CA. Statistical methodologies to pool across multiple intervention studies. Transl Behav Med. 2016 Jun;6(2):228-35. doi: 10.1007/s13142-016-0386-8.
  16. Shenoy P. Multi-regional clinical trials and global drug development. Perspect Clin Res. 2016 Apr-Jun;7(2):62-7. doi: 10.4103/2229-3485.179430.     
  17. Tsou HH, Tsong Y, Chang WJ, Dong X, Hsiao CF. Design and analysis issues of multiregional clinical trials with different regional primary endpoints. J Biopharm Stat. 2012;22:1051–9.     
  18. The Globalization of Clinical Trials. 2009. Available from: http://www.pfizer.com/files/research/nejmpaper_globalization_of_trials.pdf
  19. Schafer A. The ethics of the randomized clinical trial. N. Engl. J. Med. 1982;307:719–724. doi: 10.1056/NEJM198209163071204.     
  20. Riley W.T., Riddle M., Lauer M. NIH policies on experimental studies with humans. Nat. Hum. Behav. 2018;2:103–106. doi: 10.1038/s41562-017-0265-4.
  21. Young MJ, Bodien YG, Edlow BL. Ethical Considerations in Clinical Trials for Disorders of Consciousness. Brain Sci. 2022 Feb 2;12(2):211. doi: 10.3390/brainsci12020211.
  22. Mutual Recognition Agreements (MRA). Available in https://www.fda.gov/international-programs/international-arrangements/mutual-recognition-agreements-mra
  23. Regulatory collaboration. World health organixation. Available in https://iris.who.int/bitstream/handle/10665/331013/DI303-361-369-eng.pdf?sequence=1&isAllowed=y
  24. Makady A., van Veelen A., Jonsson P., Moseley O., D'Andon A., de Boer A., et al. (2018). Using Real-World Data in Health Technology Assessment (HTA) Practice: A Comparative Study of Five HTA Agencies. PharmacoEconomics 36 (3), 359–368. 10.1007/s40273-017-0596-z
  25. Facey K. M., Rannanheimo P., Batchelor L., Borchardt M., de Cock J. (2020). Real-world Evidence to Support Payer/HTA Decisions about Highly Innovative Technologies in the EU-Actions for Stakeholders. Int. J. Technol. Assess. Health Care, 1–10. 10.1017/S026646232000063X
  26. Makady A., Stegenga H., Ciaglia A., Debray T. P., Lees M., Happich M., et al. (2017). Practical Implications of Using Real-World Evidence (RWE) in Comparative Effectiveness Research: Learnings from IMI-GetReal. J. Comp. Eff. Res. 6 (6), 485–490. 10.2217/cer-2017-0044.
  27. Makutam V, Yashashwini Achanti S, Doostan M. INTEGRATION OF ARTIFICIAL INTELLIGENCE IN ADAPTIVE TRIAL DESIGNS: ENHANCING EFFICIENCY AND PATIENT-CENTRIC OUTCOMES. IJAR. 2024 Aug 31;12(08):205-15.

Reference

  1. Meekings K.N., Williams C.S., Fau-Arrowsmith J.E., Arrowsmith J.E. Orphan drug development: An economically viable strategy for biopharma R&D. Drug Discov. Today. 2012;17:660–664. doi: 10.1016/j.drudis.2012.02.005.
  2. Parker J. Who Has a Drug Lag? Manag. Decis. Econ. 1989;10:299–309. doi: 10.1002/mde.4090100407.  
  3. Vogel D. The Globalization of Pharmaceutical Regulation. Governance. 1998;11:1–22. doi: 10.1111/0952-1895.551998055.     
  4. Andersson F. The drug lag issue: The debate seen from an international perspective. Int. J. Health Serv. 1992;22:53–72. doi: 10.2190/9Y32-X86Y-M3F0-JQFC.
  5. Binkowitz B, Ibia E. Multiregional clinical trials: An introduction from an industry perspective. Ther Innov Regul Sci. 2011;45:569–73
  6. Guideline I.H. General Principles for Planning and Design of Multi-Regional Clinical Trials E17; The International Council for Harmonisation. 2017. [(accessed on 9 February 2021)]. Available online: https://www.ich.org/page/efficacy-guidelines 
  7. National Institutes of Health. NIH policy and guidelines on the inclusion of women and minorities as subjects in clinical research. https://grants.nih.gov/policy/inclusion/women-and-minorities/guidelines.htm
  8. Vespa J, Armstrong DM, Medina L. Demographic turning points for the United States: Population projections for 2020 to 2060: US Department of Commerce, Economics and Statistics Administration. https://www.census.gov/content/dam/Census/library/publications/2020/demo/p25-1144.pdf
  9. U.S. Food and Drug Administration. Enhancing the Diversity of Clinical Trial Populations — Eligibility Criteria, Enrollment Practices, and Trial Designs Guidance for Industry. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enhancing-diversity-clinical-trial-populations-eligibility-criteria-enrollment-practices-and-trial
  10. Woodcock J, Araojo R, Thompson T, Puckrein GA. Integrating Research into Community Practice — Toward Increased Diversity in Clinical Trials. N Engl J Med. 385 (2021) 1351–1353.
  11. Molnár-Gábor F, Sellner J, Pagil S, Slokenberga S, Tzortzatou-Nanopoulou O, Nyström K. Harmonization after the GDPR? Divergences in the rules for genetic and health data sharing in four member states and ways to overcome them by EU measures: Insights from Germany, Greece, Latvia and Sweden. Semin Cancer Biol. 2022 Sep;84:271-283. doi: 10.1016/j.semcancer.2021.12.001
  12. Assessment of the EU Member States’ Rules on Health Data in the Light of GDPR.  Available at https://health.ec.europa.eu/publications/assessment-eu-member-states-rules-health-data-light-gdpr_en
  13. Central Drugs Standard Control Organization. Available in https://cdsco.gov.in/opencms/opencms/en/Home/
  14. Abraham Yeh, Xiaohong (Grace) Zhang, Shin-Ru Wang, Novartis Pharmaceuticals Corporation, East Hanover, NJ. Pooling strategy of clinical data. Available in https://www.lexjansen.com/nesug/nesug13/21_Final_Paper.pdf
  15. Bangdiwala SI, Bhargava A, O'Connor DP, Robinson TN, Michie S, Murray DM, Stevens J, Belle SH, Templin TN, Pratt CA. Statistical methodologies to pool across multiple intervention studies. Transl Behav Med. 2016 Jun;6(2):228-35. doi: 10.1007/s13142-016-0386-8.
  16. Shenoy P. Multi-regional clinical trials and global drug development. Perspect Clin Res. 2016 Apr-Jun;7(2):62-7. doi: 10.4103/2229-3485.179430.     
  17. Tsou HH, Tsong Y, Chang WJ, Dong X, Hsiao CF. Design and analysis issues of multiregional clinical trials with different regional primary endpoints. J Biopharm Stat. 2012;22:1051–9.     
  18. The Globalization of Clinical Trials. 2009. Available from: http://www.pfizer.com/files/research/nejmpaper_globalization_of_trials.pdf
  19. Schafer A. The ethics of the randomized clinical trial. N. Engl. J. Med. 1982;307:719–724. doi: 10.1056/NEJM198209163071204.     
  20. Riley W.T., Riddle M., Lauer M. NIH policies on experimental studies with humans. Nat. Hum. Behav. 2018;2:103–106. doi: 10.1038/s41562-017-0265-4.
  21. Young MJ, Bodien YG, Edlow BL. Ethical Considerations in Clinical Trials for Disorders of Consciousness. Brain Sci. 2022 Feb 2;12(2):211. doi: 10.3390/brainsci12020211.
  22. Mutual Recognition Agreements (MRA). Available in https://www.fda.gov/international-programs/international-arrangements/mutual-recognition-agreements-mra
  23. Regulatory collaboration. World health organixation. Available in https://iris.who.int/bitstream/handle/10665/331013/DI303-361-369-eng.pdf?sequence=1&isAllowed=y
  24. Makady A., van Veelen A., Jonsson P., Moseley O., D'Andon A., de Boer A., et al. (2018). Using Real-World Data in Health Technology Assessment (HTA) Practice: A Comparative Study of Five HTA Agencies. PharmacoEconomics 36 (3), 359–368. 10.1007/s40273-017-0596-z
  25. Facey K. M., Rannanheimo P., Batchelor L., Borchardt M., de Cock J. (2020). Real-world Evidence to Support Payer/HTA Decisions about Highly Innovative Technologies in the EU-Actions for Stakeholders. Int. J. Technol. Assess. Health Care, 1–10. 10.1017/S026646232000063X
  26. Makady A., Stegenga H., Ciaglia A., Debray T. P., Lees M., Happich M., et al. (2017). Practical Implications of Using Real-World Evidence (RWE) in Comparative Effectiveness Research: Learnings from IMI-GetReal. J. Comp. Eff. Res. 6 (6), 485–490. 10.2217/cer-2017-0044.
  27. Makutam V, Yashashwini Achanti S, Doostan M. INTEGRATION OF ARTIFICIAL INTELLIGENCE IN ADAPTIVE TRIAL DESIGNS: ENHANCING EFFICIENCY AND PATIENT-CENTRIC OUTCOMES. IJAR. 2024 Aug 31;12(08):205-15.

Photo
Viswakanth makutam
Corresponding author

Pharm D, MS

Viswakanth Makutam , Navigating Regulatory Challenges In Multi-Regional Clinical Trials: A Comprehensive Overview, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 10, 978-986. https://doi.org/10.5281/zenodo.13955331

More related articles
A Review On Nanotechnology As Transformative Parad...
Vinit Khairnar, Sudarshan kale, shraddha vaishnav, chaitali marka...
A Review On The Metabolic Syndrome: Risk Factors, ...
Sakshi machindra shinde , Ashwini satalkar , Dipali shegar , ...
Formulation Optimization & Evaluation Of Oro-Dispe...
Yogesh Sonawane, M. M. Bari, S. D. Barhate, Reema Jagnit, Abhay S...
Formulation and Evaluation of Polyherbal Facial Scrub...
Roshni Uikey, Rahul Kulsange, Arti Wagh, Priya Nikam, Namrata Mane, ...
Pharmaceutical Aspects Of Accelerated Stability Studies...
Ramya Teja Medarametla, K. Venkata Gopaiah, J. N. Suresh Kumar, . Lakshmi Pravallika4, P. Mouli, S. ...
Organoids As Promising Tools In Therapy...
Dr. Pittu Vishnu Priya, Keshineni Shravani, Pabbathi Sai Divya, AVSSS Gupta, ...
Related Articles
Formulation And Evaluation of Activated Charcoal Face Pack...
Mr.Kishan Ashroba Kukar, Prof. Ram Baban Ingle, Mr.Chaukale Mahesh Manikrao, Miss.Vaishnavi Vilas Ja...
Simultaneous Estimation Of Vildagliptin And Pioglitazone In Bulk And Pharmaceuti...
Aishwarya Aijinath Ubale , Monika gopal Shinde, Punam Nivrutti Bandgar, Pradnya Prashant Shinde , Ba...
Formulation And Evaluation Of Herbal Face Mask Sheet...
Shashank Tiwari, Prabha Shukla, Sunil Mishra, Sushil Kumar Pal, ...
Development And Validation Of Reverse Phase-High-Performance Liquid Chromatograp...
S. P. Kumbhar, S. S. Patil , C. V. Panchal, P. M. Jadhav, ...
A Review On Nanotechnology As Transformative Paradigms In Drug Delivery And Ther...
Vinit Khairnar, Sudarshan kale, shraddha vaishnav, chaitali markand, ...
More related articles
A Review On Nanotechnology As Transformative Paradigms In Drug Delivery And Ther...
Vinit Khairnar, Sudarshan kale, shraddha vaishnav, chaitali markand, ...
A Review On The Metabolic Syndrome: Risk Factors, Pathophysiology, Causes Of Met...
Sakshi machindra shinde , Ashwini satalkar , Dipali shegar , ...
Formulation Optimization & Evaluation Of Oro-Dispersible Film Of Tadalafil...
Yogesh Sonawane, M. M. Bari, S. D. Barhate, Reema Jagnit, Abhay Sawant, Amol Chaudhari, ...
A Review On Nanotechnology As Transformative Paradigms In Drug Delivery And Ther...
Vinit Khairnar, Sudarshan kale, shraddha vaishnav, chaitali markand, ...
A Review On The Metabolic Syndrome: Risk Factors, Pathophysiology, Causes Of Met...
Sakshi machindra shinde , Ashwini satalkar , Dipali shegar , ...
Formulation Optimization & Evaluation Of Oro-Dispersible Film Of Tadalafil...
Yogesh Sonawane, M. M. Bari, S. D. Barhate, Reema Jagnit, Abhay Sawant, Amol Chaudhari, ...