×

Clinical Studies in Drug Development: Ensuring Safety and Approval

Clinical Studies in Drug Development: Ensuring Safety and Approval

Introduction

Clinical studies are an essential component of the drug development process, playing a critical role in ensuring the safety and effectiveness of new treatments. These studies involve testing experimental drugs or therapies on human volunteers to assess their safety, efficacy, and potential side effects. By generating valuable data, clinical trials provide the evidence needed to secure approval from regulatory agencies such as the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), and Health Canada (HC). In this overview, we’ll take a closer look at the structured process of clinical trials and their importance in bringing new, life-saving medications to the public.

1. Pre-clinical Phase

The main objective of the pre-clinical assessment is to explore primary Pharmaco-dynamic data and early toxicity, to identify hazards, evaluate risks and provide basis for risk management.

The animal studies are conducted to determine the test article does not have a toxicity profile that could cause adverse experiences in humans at pharmacological doses for which toxicological characterization of discovery lead is necessary.

  1. In-vitro toxicology assessment: The potential for toxic effects (cell death, change in cell function) to be determined using in vitro techniques, such as cell-based systems or micro-arrays.
  2. Acute (single dose) toxicity: Deal with the potential adverse effects of single doses providing information about possible dose levels for first applications to humans, and the possible effects to be expected with (accidental or intentional) over-dosing.
  3. Dose-Range-Finding Studies: These studies are performed to determine an MTD.
  4. Pilot 14-day Study: A dose level that causes toxic changes, such as morbidity or salivation, and one that produces the no-toxic-effect are determined during 14-day studies.

The results from the pharmacology and pre-clinical studies are documented in technical reports and used to prepare a regulatory agency submission for the initiation of human clinical trials.

Non-Clinical Assessment:

The non-clinical development is highly regulated by regional, national, and international guidelines and must be carried out in stringent environment in compliance to GLP requirements. Research studies intended for submission to a regulatory agency are to be conducted according to GLP regulations, as published by regulatory authorities in all the leading pharmaceutical markets.

Safety pharmacology:

The early pharmacological/pharmacodynamic studies are intended to investigate the mode of action or effects of substance in relation to desired therapeutic target.

Sub-chronic/chronic (repeated dose) toxicity:

To characterize the toxicological profile of the candidate on repeated administration, and gives information on toxic effects, potential target organs, effects on physiological functions, hematology, clinical chemistry, pathology and histopathology.

Genotoxicity:

These studies are conducted to detect compounds which induce genetic damage directly or indirectly, the usual approach is to carry out a battery of tests which provide information on gene mutations, structural chromosome aberrations and numerical chromosome aberrations.

Carcinogenicity:

To identify the tumorogenic potential in animals and to assess the relevant risk to humans.

Reproductive toxicity:

The primary goal of reproductive toxicity studies is therefore to characterize the toxicological profile of the pharmaceutical with respect to effects on:

  • Fertility and early embryonic development
  • Embryo-foetal development
  • Pre- and post-natal development, including maternal function

2. Phase 0: Exploratory Studies

Phase 0 studies are conducted in response to exploratory IND investigations are intended to expedite the clinical evaluation of new candidates to support the performance of first-in-human testing of new investigational drugs at sub-therapeutic doses based on reduced manufacturing and toxicological requirements, allowing the demonstration of drug-target effects and assessment of pharmacokinetic-pharmacodynamic relationships in humans earlier in clinical development.

Phase 0 trials include a small number of subjects (10 to 15) to gather preliminary data on the lead’s pharmacokinetics (what the body does to the drugs) that can be utilized to rank drug candidates in order to decide which has the best pharmacokinetic parameters in humans to take forward into further development.

Phase 0 studies offer the molecular proof-of-concept investigations ensuring promise of rational selection of agents for large-scale development as well as the molecular identification of potential therapeutic failures early in the development process.

3. Phase 1: Safety and Dosage

Phase I trials are the first stage of testing in human subjects involving a group of 20–100 healthy volunteers designed to assess the safety, tolerability, pharmacokinetics, and pharmaco-dynamics of a drug including dose-ranging, also called dose escalation studies, so that the best and safest dose can be found and to discover the point at which a compound is too poisonous to administer.

The tested range of doses will usually be a fraction of the dose that caused harm in animal testing.

Phase Ia:

A small group of subjects are given a single dose of the drug and observed for a period of time to confirm safety. Typically, 03 participants are monitored sequentially at a particular dose for adverse side effects, and the pharmacokinetic data are roughly in line with predicted safe values, the dose is escalated, and a new group of subjects is given a higher dose.

If unacceptable toxicity is observed in any of the participants, an additional number of participants are treated at the same dose until pre-calculated pharmacokinetic safety levels are reached, or intolerable side effects start showing up (at which point the drug is said to have reached the maximum tolerated dose (MTD)).

Phase Ib:

Multiple ascending dose studies investigate the pharmacokinetics and pharmacodynamics of multiple doses of the drug, looking at safety and tolerability. In these studies, a group of patients receives multiple low doses of the drug, while samples are collected at various time points and analyzed to acquire information on how the drug is processed within the body. The dose is subsequently escalated for further groups, up to a predetermined level.

Food effect:

A short trial designed to investigate any differences in absorption of the drug by the body, caused by eating (pre-determined meal) before the drug is given. These studies are usually run as a crossover study, with volunteers being given two identical doses of the drug while fasted, and after being fed.

4. Phase 2: Efficacy and Side Effects

Once a dose or range of doses is determined, the next goal is to evaluate whether the drug has any biological activity or effect. Phase II trials are performed on larger groups (100-300) and are designed to assess how well the drug works, as well as to continue Phase I safety assessments in a larger group of volunteers and patients.

Genetic testing is common, particularly when there is evidence of variation in metabolic rate. When the development process for a new drug fails, this usually occurs during Phase II trials when the drug is discovered not to work as planned, or to have toxic effects.

Phase IIa is specifically designed to assess dosing requirements.

Phase IIb is specifically designed to study efficacy. 

5. Phase 3: Confirmatory Studies

Phase III is designed to assess the effectiveness of the new intervention and, thereby, its value in clinical practice. Phase III studies are randomized controlled multicenter trials on large patient groups (300–3,000 or more depending upon the disease/medical condition studied) and are aimed at being the definitive assessment of how effective the drug is, in comparison with current ‘gold standard’ treatment.

Phase III trials are the most expensive, time-consuming and difficult trials to design and run, especially in therapies for chronic conditions determined as “pre-marketing phase” as it measures consumer response to the drug.

In addition, trials are performed for “label expansion” to obtain additional safety data, or to support marketing claims for the drug that can be designated as “Phase IIIB studies.”

During the clinical development of an investigational drug, periodic analysis of safety information is crucial to the ongoing assessment of risk and the comprehensive evaluation of safety information, exploring new safety issues and management of potential risks should be submitted as DSUR to assure regulators that safety profile is adequately monitored. 

Safety information to support clinical development phases

Stage Information Required
Phase 0 Single dose toxicity (sub acute study) 02 rodent species
Phase I Repeated dose toxicity 02 species (01 rodent and 01 non-rodent mammal)
Phase II Repeated dose toxicity 02 mammalian species (01 rodent and 01 non-rodent)
Phase III Carcinogenicity
Product Label Mutagenicity, Carcinogenicity, Reproductive toxicology (Impairment of fertility, Teratogenicity, and peri/post-natal developmental effects)

Product Label Mutagenicity, Carcinogenicity, Reproductive toxicology (Impairment of fertility, Teratogenicity, and peri/post-natal developmental effects).

6. Phase 4: Post-Marketing Surveillance

Once a drug is approved for market use, Phase 4 trials continue to monitor the drug’s long-term safety and effectiveness in the general population. These trials are often called post-marketing surveillance or real-world evidence (RWE) studies. This phase helps detect any rare or long-term side effects that may not have been evident during earlier trials.

Phase 4 trials can also involve further studies to explore additional indications, dosages, or combinations with other treatments. The key features of PMS include:

  • Long-Term Safety: Monitoring the drug’s effects over a longer period to identify rare or delayed side effects not seen in earlier phases.

  • Additional Indications: Researchers may explore the drug’s use in different populations, combinations with other drugs, or for new conditions that were not included in the original trials.

  • Usage Patterns: Assess how the drug is being used in the real world, including off-label uses and adherence rates.

  • Pharmacovigilance: Continuous collection and analysis of adverse event data from patients using the drug in routine clinical practice. Regulatory agencies can issue safety warnings or require changes to drug labeling based on Phase 4 findings.

Phase 4 is also where drugs that demonstrate significant benefits in real-world use may undergo further trials for new indications or approvals for broader use. 

Summary of the Clinical trails in a Nutshell:

Phase Purpose Participants Outcome
Phase 0 Explore pharmacokinetics and pharmacodynamics 0-15 Preliminary data on drug action and metabolism
Phase 1a Assess initial safety and tolerability (single dose) 20–80 Identify safe dose range, pharmacokinetics, and effects
Phase 1b Assess safety with repeated dosing 20–80 Establish safety profile under multiple dosing
Phase 2 Evaluate effectiveness, refine dose 100–300 Preliminary efficacy data, side effect profile
Phase 3 Confirm effectiveness, monitor reactions 300-3000 Definitive efficacy, risk-benefit assessment
Phase 4 Post-marketing surveillance Public

Long-term safety and effectiveness in real-world use

Challenges and Considerations 

  • Recruitment Issues: Finding enough participants who meet the inclusion criteria for clinical trials can be difficult, particularly for rare diseases or conditions. Recruitment delays can extend trial timelines.

  • Ethical Considerations: Ensuring that participants provide informed consent, understanding the risks and benefits of participation, and adhering to ethical guidelines are critical components of clinical trial design.

  • Cost and Time: Clinical trials can be extremely costly, sometimes running into the billions of dollars, especially for Phase 3 trials. This makes drug development a high-risk and resource-intensive process.

  • Failure Rates: Most drug candidates fail to make it through clinical trials, particularly in Phase 2 and Phase 3, where the drug may not demonstrate sufficient efficacy or may reveal serious safety issues. On average, fewer than 10% of experimental drugs ever reach the market.

Conclusion

In summary, clinical trials are the cornerstone of drug development, offering a rigorous, evidence-based process that validates the safety and effectiveness of new treatments. From the initial preclinical stages to post-marketing surveillance, these trials play a pivotal role in shaping public health by ensuring that only those therapies that meet the highest standards of safety and efficacy reach the market. As the pharmaceutical industry continues to innovate, clinical trials remain indispensable in safeguarding the well-being of patients and advancing medical science for future generations.

    Post Comment

    You May Have Missed

    Don’t copy the text; How about upgrading it with your genius touch!