SND College of Pharmacy, Babhulgaon.
Clinical trials are the cornerstone of evidence-based medicine, providing scientific data on the safety, efficacy, and quality of new drugs, biologics, and medical devices. These trials are conducted in a phased manner – Phase I to Phase IV – to systematically assess pharmacokinetics, pharmacodynamics, therapeutic benefits, and long-term effects. Regulatory oversight plays a crucial role in ensuring that clinical research is conducted ethically, safely, and in compliance with Good Clinical Practice (GCP) guidelines. Global regulatory bodies such as the U.S. FDA, EMA, and CDSCO (India) monitor and approve trial protocols, informed consent processes, and data reporting to safeguard participant rights and ensure scientific validity. Recent developments include the rise of adaptive trial designs, decentralized clinical trials, and the use of digital health technologies for real-time monitoring. Despite significant progress, challenges remain, including under-reporting of adverse events, recruitment difficulties, and the need for harmonization of regulatory requirements across countries Streng.
Clinical research serves as the backbone of modern medicine by producing dependable evidence regarding the safety, effectiveness, and quality of new therapeutic options. Through clinical trials, discoveries made in preclinical studies are systematically converted into clinically approved drugs, vaccines, and medical devices. The clinical trial process is organized into four sequential phases: Phase I focuses on assessing safety and determining optimal dosage in a small group of healthy participants, Phase II examines preliminary effectiveness and identifies short-term side effects in patients, Phase III validates therapeutic benefits on a larger scale, and Phase IV conducts post-marketing surveillance to monitor rare or long-term adverse events.[1] Regulatory supervision plays a critical role in ensuring that every stage of this process is scientifically sound, ethically compliant, and prioritizes participant protection. Internationally recognized standards such as the ICH-GCP guidelines, along with regulations enforced by authorities including the U.S. FDA, EMA, and India’s CDSCO, provide clear guidance for trial approval, implementation, monitoring, and reporting.[2] With the growing complexity and globalization of clinical research, regulatory oversight has become even more significant. Key concerns include ensuring informed consent, safeguarding vulnerable populations, and maintaining transparency in data reporting. The COVID-19 pandemic further emphasized the need for streamlined yet safe research pathways.[3] This review seeks to present a detailed summary of the clinical trial framework, outline the responsibilities of regulatory bodies, discuss recent innovations such as adaptive and decentralized trial designs, and highlight existing challenges with suggestions for improving the future of clinical research.
Definition of clinical trials: -
A clinical trial is a systematically designed, prospective research study conducted in human participants to evaluate the safety, efficacy, and quality of new drugs, vaccines, devices, diagnostic tools, or treatment regimens. The primary goal is to generate reliable scientific data that supports approval for clinical use and improves patient care.
Objectives of clinical trials: -
Assessment of Safety: To systematically examine the safety profile of investigational drugs, vaccines, or medical devices by identifying both short-term and long-term risks in human subjects.
Evaluation of Efficacy: To verify whether the new intervention produces the expected therapeutic effect and contributes to better clinical outcomes.
Determination of Optimal Dosage and Administration: To establish the most appropriate dose, route, and frequency of administration for maximum benefit with minimal risk.
Detection of Adverse Reactions: To actively monitor, record, and analyze any adverse drug reactions or toxicities that may occur during or after treatment.
Comparison with Standard Therapy: To compare the new treatment with existing standard care to assess whether it offers superior, equivalent, or safer results.
Evidence Generation for Regulatory Decisions: To provide robust and scientifically valid data that support approval by regulatory authorities and guide clinical practice recommendations.
Contribution to Public Health: To expand medical knowledge, improve disease management, and ultimately enhance overall patient and population health outcomes.[4][5][6]
Phases of clinical Trials: -
Clinical trials are systematically divided into multiple stages, each with a specific purpose, sample size, and study design. Together, these phases generate evidence on the safety and effectiveness of new interventions.
Phase 0 – Exploratory or Microdosing Studies
This is an early, optional step performed on a very small number of participants (about 10–15). Its primary purpose is to study how the investigational drug behaves inside the human body—its absorption, distribution, metabolism, and elimination—using very small doses. There is no therapeutic or diagnostic intent at this stage, but it helps decide whether the drug should advance to further phases.
Phase I – First-in-Human Safety Studies
This phase primarily focuses on evaluating the safety, tolerability, and pharmacokinetics of the investigational product. It usually involves 20–100 healthy volunteers (or patients when studying drugs for cancer or life-threatening conditions). Phase I studies often use dose-escalation designs to determine the maximum tolerated dose (MTD) and observe early side effects.
Phase II – Therapeutic Exploration
Phase II trials examine the efficacy of the drug in patients with the targeted condition while continuing to collect safety data. These trials typically recruit 100–300 participants and are often randomized and controlled. The goal is to establish proof of concept—confirming that the drug produces the desired therapeutic effect.
Phase III – Large-Scale Confirmatory Trials
This phase is designed to provide definitive evidence of efficacy and further monitor side effects. Thousands of patients (often 1,000–3,000 or more) across multiple centres participate. These are generally double-blind randomized controlled trials (RCTs) comparing the new intervention to a placebo or standard treatment. The results form the basis for seeking marketing approval from regulatory agencies.
Phase IV – Post-Marketing Studies
Once the drug is approved and available to the public, Phase IV studies continue to monitor its long-term safety, rare side effects, and real-world effectiveness. These studies may involve thousands of patients and sometimes lead to updates in drug labelling, additional warnings, or even withdrawal from the market if significant risks are discovered.[7][8][9]
Types of clinical Trials: -
Clinical trials can be grouped according to their aim, design, and approach. These classifications help researchers choose the most appropriate method to answer a clinical research question.
1. Interventional (Experimental) Studies
In these studies, investigators actively assign participants to receive a treatment, drug, or procedure and then measure outcomes. Randomized Controlled Trials (RCTs) and crossover studies are common examples. Interventional trials are designed to establish cause–effect relationships.
2. Observational Studies
Here, researchers only observe participants in their natural setting without assigning any intervention. Cohort studies follow participants over time to study exposure–outcome relationships. Case–control studies compare people with a condition to those without to identify risk factors. Cross-sectional studies capture exposure and outcome data at a single time point.
3. Randomized Controlled Trials (RCTs)
Considered the gold standard for clinical research, RCTs randomly allocate participants to experimental and control groups. Blinding (single- or double-blind) minimizes bias and increases reliability.
4. Open-Label Trials
In open-label studies, both participants and investigators know which treatment is being administered. These are useful when blinding is impractical, such as surgical or device trials.
5. Crossover Trials
Each participant receives both the investigational treatment and the control/placebo at different time periods, with a washout period in between. This design reduces variability as each participant serves as their own control.
6. Pragmatic (Real-World) Trials
These studies focus on the effectiveness of interventions in everyday clinical practice, emphasizing external validity and generalizability rather than strict experimental control.
7. Adaptive Trials
Adaptive trials allow modifications to trial design—such as sample size adjustments or dropping/adding treatment arms based on interim results, without compromising study integrity.
8. Prevention Trials
These trials investigate strategies to prevent diseases or conditions, including vaccines, lifestyle interventions, or prophylactic drugs.
9. Screening and Diagnostic Trials
Such trials assess new diagnostic tools or screening methods to improve accuracy and early detection of diseases.
10. Quality of Life (QoL) or Supportive Care Trials
These focus on interventions that enhance patient comfort, reduce treatment side effects, and improve quality of life, particularly in chronic illness or palliative care settings.
11. Post-Marketing (Phase IV) Studies
Conducted after regulatory approval, these studies monitor long-term safety, rare adverse effects, and real-world effectiveness in a large population.[10][11][12]
Clinical Trials Designs: -
Clinical trial design represents the overall strategy or plan that determines how a study is structured, including participant allocation, interventions, comparisons, and outcome assessments. A well-planned design is crucial for maintaining scientific validity, minimizing bias, and generating reproducible evidence.
1. Randomized Controlled Trials (RCTs)
RCTs are considered the gold standard of clinical research. Participants are randomly assigned to one or more intervention groups or to a control/placebo group, ensuring baseline comparability and reducing selection bias. The most common format is the parallel-group design, where participants stay in their assigned group throughout the trial. Variants include cluster-randomized trials, in which groups (such as clinics or schools) are randomized, and crossover trials, where participants receive interventions in sequence with a washout period, serving as their own control and reducing variability.
Non-Randomized or Quasi-Experimental Designs
In some situations, randomization is not feasible or ethical. Non-randomized designs allocate participants using nonrandom methods, such as physician preference or availability of treatment. These studies are easier to conduct but have a higher risk of bias and confounding, requiring careful statistical adjustment.
2. Blinded and Open-Label Designs
Blinding is used to reduce bias by concealing treatment allocation. Single-blind trials hide allocation from participants but not investigators. Double-blind trials conceal allocation from both participants and investigators. Triple-blind trials extend blinding to outcome assessors or statisticians. In contrast, open-label designs disclose treatment assignments to everyone, which is practical in surgical, device-based, or behavioral studies but more prone to bias.
3. Factorial Designs
Factorial trials evaluate more than one intervention simultaneously in the same population, allowing researchers to study individual and combined effects efficiently. For example, a 2×2 factorial design could test Drug A, Drug B, both drugs together, and placebo.[13][14][15][6]
Ethical considerations in clinical Trials: -
1.Respect for Autonomy
Participants must be allowed to make voluntary and informed decisions about joining or continuing in a study.
Informed Consent: Study objectives, procedures, potential risks, benefits, and the right to withdraw should be communicated clearly and in an understandable language. Safeguards for Vulnerable Groups: Special protection is needed for children, pregnant women, prisoners, or individuals with impaired decision-making capacity.
2.Beneficence and Risk Minimization
Trials should aim to maximize potential benefits while reducing risks to the lowest feasible level. Risk–Benefit Evaluation: The anticipated benefit must justify participant exposure.
Ongoing Safety Oversight: Data Safety Monitoring Boards (DSMBs) and interim analyses help detect harm early and
stop unsafe studies.[19][20]
3.Principle of Justice
Research participation should be fair, avoiding exploitation any specific group. Equitable Recruitment: Study populations should be chosen to distribute risks and benefits fairly.
Post-Trial Benefits: Participants and host communities should have access to proven interventions once the study concludes.
4.Scientific and Ethical Integrity
Only scientifically valid studies should be conducted to avoid unnecessary risks. Sound Study Design: Adequate sample size, appropriate endpoints, and robust methodology are essential.
Result Transparency: Trials must be registered in public databases, and findings published irrespective of whether results are positive or negative.
5.Confidentiality and Data Protection
Protecting personal data is an ethical and legal requirement.
Secure Handling: Information should be coded, stored safely, and shared only with authorized personnel.
Regulatory Compliance: Follow global standards such as GDPR (Europe) or HIPAA (USA).[17][18]
Regulatory oversight in clinical Trials: -
1. Purpose of Oversight: - Ensures clinical trials are ethical, scientifically valid, and legally compliant. Protects participants’ rights and
1. Regulatory Authorities
FDA (USA), EMA (Europe), CDSCO (India), PMDA (Japan) – review, approve, and monitor trials. ICH-GCP –
WHO guidelines – promote trial registration and transparency.[21]
2. Regulatory Submissions
IND/CTA applications required before starting trials.
Clinical Study Reports (CSR) submitted after trial completion for final approval.
3. Safety Monitoring & Pharmacovigilance
Mandatory reporting of SAEs (Serious Adverse Events) and SUSARs.
Continuous risk–benefit monitoring by Data Safety Monitoring Boards (DSMBs).[22][23]
Good Clinical Practice (GCP) Compliance: -
GCP is an international ethical and scientific standard ensuring that clinical trials are participant-cantered, safe, and scientifically valid.
1.Core Principles
Ethical conduct: Trials must follow the Declaration of Helsinki.
Risk–benefit balance: Benefits must outweigh potential risks.
Informed consent: Clear, voluntary, and documented consent is mandatory.
Confidentiality: Protect participant data and privacy.
Protocol adherence: Follow only approved study protocols.
Qualified personnel: Trials must be conducted by trained investigators.
Accurate documentation: Maintain complete and timely records.
Quality systems: Regular monitoring and auditing are required.
2.Responsibilities
Investigators: Follow protocol, ensure participant safety, report data accurately.
Sponsors: Provide resources, monitor compliance, and submit regulatory documents.
IRB/IEC: Review protocols, approve informed consent forms, and oversee trial ethics.[24][25]
3.Key Requirements
Develop and register a protocol (CTRI/ClinicalTrials.gov). Monitor sites regularly and conduct audits. Report adverse events promptly. Archive essential documents for inspections.
4.Benefits
Protects participants’ rights and safety.
Improves credibility and reproducibility of trial data.
Facilitates global regulatory approval.
6.Challenges
High costs and administrative workload.
Need for continuous training of investigators.
Complexity with multicentre and decentralized trials.[26]
Recent Advance in Clinical Trials: -
1.Adaptive Designs – Allow mid-trial changes (sample size, dosage) to improve efficiency.
2.Decentralized/Virtual Trials – Use telemedicine, e-consent, and remote monitoring to increase accessibility.
3.AI & Machine Learning – Enhance patient recruitment, monitoring, and data analysis.[27]
4.Real-World Evidence (RWE) – Integrate data from EHRs, registries, and wearables for long-term insights.
5.Biomarker-Based & Precision Trials – Select patients based on genetic/molecular profiles for targeted therapies.
6.Platform, Basket & Umbrella Designs – Test multiple treatments or diseases under a single trial framework.[28]
7.Patient-Centric Models – Include patient-reported outcomes and digital engagement tools.
8.Blockchain & Digital Security – Ensure transparency and data integrity.
9.Regulatory Innovations – Fast-track approvals, rolling submissions, and remote inspections for quicker drug development.[29]
Challenges and Limitations: -
Future Direction: -
Greater Use of Digital Health Technologies
Expand telemedicine, e-consent, remote monitoring, and wearable devices to make trials more patient-friendly.
AI-Driven Trial Design & Data Analysis
Apply artificial intelligence and machine learning for protocol optimization, patient recruitment, and predictive analytics.
Personalized & Precision Medicine Approaches
Conduct trials based on genetic/molecular profiling to deliver targeted, individualized therapies.
Decentralized & Hybrid Trial Models
Combine site-based and home-based approaches to improve participation from diverse populations.
Real-World Evidence Integration
Use electronic health records and patient registries alongside trial data for better decision-making.
Global Harmonization of Regulations
Streamline approval processes across countries to reduce delays in multinational trials.[33][34]
Regulatory oversight: -
Regulatory oversight refers to the systematic monitoring, evaluation, and enforcement of laws, guidelines, and standards by authorized governmental or regulatory bodies to ensure the safety, quality, efficacy, and compliance of products, processes, or services. In the context of nutraceuticals and herbal formulations, it encompasses approval, labelling, manufacturing practices, safety assessment, and post-marketing surveillance to protect public health.[35][36][37] Classification: -
Classification of Regulatory Oversight in Nutraceuticals: -
Regulatory oversight ensures that nutraceutical products meet safety, quality, and efficacy standards throughout their lifecycle. It can be broadly categorized as follows:
1.Pre-Market Oversight
Before products reach the market, regulatory authorities evaluate safety and efficacy data. This includes:
Product Registration & Approval: Ensuring that formulations comply with regional safety and quality requirements, such as those outlined under the Dietary Supplement Health and Education Act (DSHEA) in the USA (FDA, 19947).[38]
Ingredient Approval: Assessment of new dietary ingredients for safety under recommended use.
Labelling Compliance: Accurate disclosure of ingredients, dosages, and health claims.
Good Manufacturing Practices (GMP): Verification that manufacturing follows standardized protocols to ensure consistent product quality.[39]
2.Post-Market Oversight
After commercialization, regulatory supervision focuses on:
Adverse Event Monitoring: Tracking and reporting safety issues associated with the product.
Product Recalls & Enforcement: Removal of unsafe or non-compliant products.
Facility Inspections & Audits: Ensuring ongoing compliance with GMP and regulatory standards (FDA, 2025).[40]
3.Risk-Based Oversight
4.Geographic and Jurisdictional Oversight
National Authorities: FDA (USA), EFSA (Europe), FSSAI (India) provide country-specific regulatory frameworks.
International Guidelines: Codex Alimentarius sets global food and supplement safety standards.
Regional Harmonization: Organizations like ASEAN aim to standardize regulations for cross-border safety and trade (Springer, 2025) Regulatory Oversight. [41]
Quality standard: -
1. Good Manufacturing Practices (GMP)
Standardized processes, trained personnel, and controlled equipment ensure consistent product quality and minimize contamination risks.
Regulatory bodies like FDA, EFSA, and FSSAI require GMP compliance for market authorization.
Standardization of Raw Materials
Active compounds (e.g., Moringa polyphenols or flavonoids) must meet defined concentration levels to reduce batchto-batch variability and maintain therapeutic efficacy.
2. Analytical Testing and Method Validation
Validated methods such as HPLC, UV spectroscopy, or GC-MS are used to quantify active ingredients and detect impurities.
Guarantees product safety and uniformity throughout shelf life.
1.Stability and Shelf-Life Assessment
Stability studies evaluate how storage conditions affect bioactive compound potency over time.
Regulatory authorities require evidence of stability to support labelled shelf life.[42]
2.Labelling and Documentation
Product labels must accurately reflect ingredients, dosage, and usage instructions.
Comprehensive records of manufacturing, testing, and quality control are necessary for inspections.
3.Compliance Inspections and Audits
Regulatory inspections verify adherence to GMP, quality testing, and documentation protocols. Non-compliance can lead to warnings, penalties, or product recalls.[43][44]
Safety Evaluation: -
1.Preclinical Safety Testing
Toxicological Evaluations: Conducting acute, sub-acute, and chronic toxicity studies in animal models to detect potential harmful effects.
Genotoxicity and Carcinogenicity Assessments: Screening ingredients for DNA damage or cancer risk.
Reproductive and Developmental Safety: Evaluating potential effects on fertility and fetal development (Perumalsamy et al., 2024).[47]
2.Clinical Safety Evaluation
Human Trials: Conducting clinical studies to confirm safety and tolerability in the target population.
Dose Determination: Establishing safe and effective dosage ranges for consumption (FDA, 2025).[45]
3.Safety of Ingredients and Formulations
Novel or New Dietary Ingredients (NDIs): Regulatory review of safety before market approval.
Advanced Formulations: Special scrutiny for nano encapsulated, fortified, or novel delivery systems to prevent potential bioaccumulation or toxicity (EFSA, 2020).[46]
Efficacy and Health claim: -
Regulatory oversight ensures that nutraceutical products not only are safe but also that any claims regarding their health benefits are scientifically validated, preventing misleading information for consumers.
1. Scientific Validation of Claims
Manufacturers must provide evidence supporting claims such as antioxidant, anti-inflammatory, or blood sugar– modulating effects.
Evidence may include in vitro experiments, animal studies, or human clinical trials (Perumalsamy et al., 2024).[50]
2. Types of Claims
Structure/Function Claims: Describe how a nutrient supports normal body functions (e.g., “Supports immune health”).
Disease Risk Reduction Claims: Indicate that a nutrient may lower the risk of a disease; these require strong scientific evidence.
General Well-Being Claims: Suggest maintenance or improvement of overall health; evidence is required but less stringent than for disease claims (FDA, 2025).[48]
3. Pre-Market Assessment
Regulatory agencies review the submitted evidence before permitting marketing.
Claims must be accurate, not misleading, and consistent with scientific data (EFSA, 2020).[49]
4. Labelling and Advertising
Health claims are strictly regulated on packaging and promotional material.
Misrepresentation or exaggerated claims may lead to fines, warnings, or product recalls (FDA, 2025).[48] Post-Market Surveillance
Continuous monitoring ensures claims remain substantiated as new scientific data emerge.
Labelling and consumer information: -
Labelling and consumer information are key components of regulatory oversight, ensuring transparency, safety, and informed use of nutraceutical products.
1. Ingredient Disclosure
Labels must clearly list all ingredients, including active compounds, excipients, and additives, allowing consumers to avoid allergens or undesired substances (Perumalsamy et al., 2024).[53]
2. Dosage and Administration
Clear instructions regarding recommended dose, frequency, and method of use help prevent misuse or overconsumption (FDA, 2025).[51]
3. Health and Function Claims
Claims regarding effects on health, body functions, or well-being must be scientifically substantiated and approved by regulatory authorities (EFSA, 2020).[52]
4. Warnings and Precautions
Labels should indicate potential side effects, contraindications, and interactions with other medications, especially for vulnerable populations such as children, pregnant women, or patients with chronic conditions (Perumalsamy et al., 2024).[53]
5. Regulatory Compliance
Authorities like FDA, EFSA, and FSSAI regulate labelling requirements, including clarity, language, font size, and placement. Misleading or false labelling can lead to fines, recalls, or marketing bans (FDA, 2025).[51]
Import, Export and Market Authorization: -
Regulatory oversight ensures that nutraceutical products are authorized, traded, and marketed safely while complying with national and international standards.
1. Market Authorization
Products must receive approval from relevant authorities (e.g., FDA, FSSAI, EFSA) before commercialization.
Approval confirms adherence to safety, efficacy, labeling, and manufacturing quality standards (Perumalsamy et al., 2024).[57]
2. Import Control
Imported nutraceuticals must satisfy the regulatory requirements of the destination country.
Required documentation includes product dossiers, GMP certificates, certificates of analysis, and safety/efficacy evidence (FDA, 2025).[54]
Customs inspections ensure compliance with labeling, packaging, and ingredient standards.
2. Export Compliance
Exported products must meet regulations of both the origin and destination countries.
Certificates of origin, GMP certifications, and quality testing reports are often mandatory.
Regulatory oversight maintains product safety, quality, and traceability in international trade (FSSAI, 2022).[56]
3. Enforcement and Compliance
Violation of import/export rules or lack of market authorization can result in fines, product seizure, or withdrawal of approval.
Regulatory authorities conduct audits, inspections, and testing to ensure ongoing compliance (EFSA, 2020).[55]
4. Global Harmonization
International frameworks such as Codex Alimentarius, ASEAN, and ICH provide guidance for standardizing import/export regulations.
Harmonization facilitates safe trade while ensuring quality and consumer protection (Perumalsamy et al., 2024).[57]
Post Marketing Surveillance: -
Post-marketing surveillance (PMS) is a crucial component of regulatory oversight, ensuring that nutraceutical products remain safe, effective, and of high quality after they are available to consumers.
1. Monitoring Adverse Events
Collection and analysis of reports on side effects or unexpected reactions from consumers.
Helps identify safety concerns that may not have been detected during pre-market evaluation (Perumalsamy et al., 2024) [60].
2. Quality Assurance
Periodic testing to confirm potency, purity, and compliance with labeling standards.
Ensures products maintain regulatory specifications throughout their shelf life (FDA, 2025).[58]
3. Risk Management and Corrective Actions
Regulatory authorities implement corrective measures based on PMS data.
Actions may include product recalls, safety warnings, or updates to labeling instructions (EFSA, 2020).[59]
Challenges in Regulation of Novel formulation:-
Novel nutraceutical formulations such as nanoparticles, liposomes, or fortified extracts face unique regulatory challenges due to their complexity and absence of specific guidelines.
1. Inadequate Regulatory Framework
Many countries lack tailored regulations for advanced formulations, making evaluation of safety, efficacy, and quality inconsistent (FDA, 2025).[61]
2. Safety Assessment Limitations
Altered bioavailability and pharmacokinetics in novel delivery systems may increase unforeseen risks.
Standard toxicological tests may not capture long-term or systemic effects (EFSA, 2020).[62]
3. Efficacy Evaluation Complexity
Enhanced absorption in new delivery systems complicates dose–response relationships.
Limited clinical data often restrict robust substantiation of health claims (Perumalsamy et al., 2024).[63]
4. Quality Control Issues
Maintaining consistency, stability, and reproducibility is difficult for complex formulations.
Analytical techniques require modification to accurately quantify actives in advanced matrices.
Recent Development: -
CONCLUSION: -
Clinical trials remain the cornerstone for generating high-quality evidence on the safety, efficacy, and therapeutic potential of novel interventions. Regulatory oversight plays an equally vital role in translating this evidence into safe and effective products for public use. A well-structured regulatory framework ensures Good Clinical Practice (GCP) compliance, ethical protection of participants, accurate labeling, and post-marketing surveillance, thereby safeguarding public health.
With the increasing complexity of novel formulations and global nutraceutical trade, harmonization of international standards, adoption of digital tools for traceability, and risk-based monitoring approaches are essential. Strengthening collaboration among regulators, researchers, and industry stakeholders can enhance transparency and accelerate innovation without compromising safety. Future directions should emphasize adaptive regulations, integration of AI for safety monitoring, and robust communication strategies to maintain consumer trust.
REFERENCES
Nyaharkar Varsha*, Darwade Amol, Divya Shinde, Aditya Shinde, Gaurav Zalte, Dnyaneshwar Shinde, Clinical Trials and Regulatory Oversight, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 11, 1470-1485 https://doi.org/10.5281/zenodo.17573151
10.5281/zenodo.17573151