top of page

Understanding Pharmaceutical Medicine: From Drug Discovery to Development




Introduction

Every year, hundreds of medications are pushed into the market globally, but more than twice of that number don't make it to the market. Why's that so? That's where pharmaceutical medicine comes in. Pharmaceutical medicine focuses on the development, evaluation, and regulation of medications. It aims to create drugs that effectively treat diseases and improve patient outcomes. This involves rigorous research and clinical trials to ensure that medications are both effective and safe for use. It also involves extensive testing to identify potential side effects and interactions, thereby minimizing risks to patients. It is important in navigating the regulatory landscape to ensure that new drugs meet the necessary legal and ethical standards before they can be marketed.



Drug Discovery and Target identification

Drug discovery is a complex process aimed at identifying new therapeutic agents to treat various diseases. A critical initial step in this process is target identification, which involves selecting a biological molecule that plays a significant role in a disease's pathology. Targets can include proteins, enzymes, receptors, and nucleic acids, which are integral to the disease mechanism. The identification of these targets is essential as it guides the subsequent stages of drug discovery, including hit identification and lead optimization.


In pharmaceutical medicine, the target identification process begins with a thorough understanding of the disease's etiology and pathophysiology. This process utilizes various approaches, such as genomics, proteomics, and bioinformatics, to uncover potential targets. This is often complemented by insights from clinical observations and patient data, which help in recognizing unmet medical needs and the biological relevance of the targets.


Once potential targets are identified, they undergo validation to confirm their role in the disease. This validation can involve genetic manipulation, biochemical assays, and animal models to demonstrate that modulating the target can lead to a therapeutic effect. Successful validation is crucial as it ensures that the drug development efforts are focused on biologically relevant targets, thereby increasing the likelihood of clinical success.



Lead identification and Optimization


Lead identification involves the selection of promising compounds, known as 'hits,' that demonstrate biological activity against a specific target identified during the earlier stages of drug discovery. These hits can be derived from various sources, including high-throughput screening of compound libraries, virtual screening using computational methods, or natural product extraction. The goal is to identify compounds that exhibit adequate potency, selectivity, and favorable pharmacokinetic properties. During this phase, medicinal chemists evaluate the structure-activity relationship (SAR) of the hits to understand how chemical modifications can enhance their efficacy and reduce toxicity.


Once suitable leads are identified, the process transitions to lead optimization. This phase aims to refine the lead compounds to improve their drug-like properties, ensuring they are safe and effective for clinical use. Optimization involves systematic chemical modifications to enhance potency, selectivity, and pharmacokinetics, including absorption, distribution, metabolism, and excretion (ADME) characteristics. Medicinal chemists employ various strategies, such as structure-based drug design and combinatorial chemistry, to generate analogs of the lead compounds and assess their biological activity. Additionally, lead optimization includes extensive in vitro and in vivo testing to evaluate the safety profile and therapeutic window of the compounds. This stage is crucial for identifying and mitigating potential toxicity issues, ensuring that the optimized leads can progress to preclinical development.


Preclinical Development

Preclinical development is a crucial phase in the drug development process that occurs after lead optimization and before clinical trials. This stage is designed to gather essential data on the safety, efficacy, and pharmacokinetics of a drug candidate, ensuring that it is suitable for testing in humans. The primary objectives of preclinical development are to assess the safety profile of the drug candidate, establish its pharmacological effects, and determine its pharmacokinetic properties. This phase aims to identify any potential toxicities, and understand the drug's mechanism of action, and evaluate its therapeutic effects.


Safety Pharmacology and Toxicology Studies

Safety pharmacology studies are conducted to evaluate the potential effects of the drug on vital organ systems, such as the cardiovascular, respiratory, and central nervous systems. These studies help identify any undesirable effects that could arise from the drug's administration. Toxicology studies, on the other hand, assess the potential for adverse effects at various doses and exposure durations. These studies typically involve multiple animal species to provide a comprehensive understanding of the drug's safety profile.

Toxicology studies are categorized into acute, sub-chronic, and chronic studies, depending on the duration of exposure. Acute studies assess the effects of a single dose, while sub-chronic and chronic studies evaluate the effects of repeated dosing over extended periods.


Pharmacokinetics and ADME Studies

Pharmacokinetics (PK) studies are essential for understanding how the drug is absorbed, distributed, metabolized, and excreted (ADME) in the body. These studies shed light into the drug's bioavailability, half-life, and clearance rates, which are critical for determining appropriate dosing regimens (Vrbanac & Slauter, 2017).


Formulation Development

Another critical aspect of preclinical development is formulation development. This involves creating a stable and effective dosage form of the drug candidate, such as tablets, capsules, or injectable solutions. The formulation must ensure that the drug is delivered effectively to the target site in the body while maintaining stability and bioavailability. Formulation scientists work to optimize the drug's solubility, stability, and release characteristics, which are vital for its success in clinical trials (Strickley, 2008).


Regulatory Considerations

Preclinical development is also closely tied to regulatory requirements. Before a drug candidate can enter clinical trials, sponsors must submit an Investigational New Drug (IND) application to regulatory agencies, such as the U.S. Food and Drug Administration (FDA) and National Agency for Food and Drug Administration and Control (NAFDAC). The IND application includes data from preclinical studies, proposed clinical trial protocols, and information on the drug's manufacturing process. Regulatory agencies review this information to ensure that the drug candidate is safe for human testing.


Clinical Development

Clinical development is a pivotal phase in the drug development process, following preclinical studies and preceding regulatory approval. This stage involves a series of carefully designed clinical trials that assess the safety, efficacy, and optimal dosing of a new drug candidate in human subjects. The clinical development process is typically divided into four phases—Phase I, Phase II, Phase III, and Phase IV—each with distinct objectives and methodologies.


Phase I Trials

Phase I trials are the first step in testing a new drug in humans. These studies primarily focus on assessing the safety and tolerability of the drug. Conducted with a small group of healthy volunteers (usually 10 to 100), Phase I trials aim to determine the maximum tolerated dose (MTD) and identify any potential side effects. Researchers monitor pharmacokinetics (ADME—absorption, distribution, metabolism, and excretion) to understand how the drug behaves in the human body. The trials are typically open-label, meaning both the researchers and participants know which drug is being administered. The data collected during this phase is crucial for designing subsequent trials.


Phase II Trials

Once a drug has demonstrated safety in Phase I, it progresses to Phase II trials, which focus on evaluating the drug's efficacy and further assessing its safety in a larger group of patients (usually 50 to 500). These trials are often randomized and controlled, comparing the investigational drug to a placebo or standard treatment. Phase II trials aim to determine the optimal dose and dosing regimen while continuing to monitor safety. This phase is critical for establishing whether the drug has a therapeutic effect and for identifying any dose-response relationships. Many drug candidates fail during this phase due to insufficient efficacy or unacceptable safety profiles.


Phase III Trials

Phase III trials are conducted on a much larger scale, typically involving several hundred to several thousand participants. The primary objective of this phase is to confirm the drug's efficacy and monitor its adverse effects in a diverse patient population. Phase III trials are randomized, double-blind, and controlled, providing robust data on the drug's performance compared to existing treatments or placebos. Successful completion of Phase III trials is often required for regulatory approval, as the data generated is critical for demonstrating the drug's benefit-risk profile. If the results are positive, the sponsor compiles all data into a New Drug Application (NDA) for submission to regulatory authorities.


Phase IV Trials

After a drug receives regulatory approval and is marketed, Phase IV trials, also known as post-marketing surveillance, begin. These studies aim to monitor the drug's long-term safety and effectiveness in the general population. Phase IV trials can identify rare or long-term adverse effects that may not have been evident in earlier trials due to smaller sample sizes or shorter durations. They also provide valuable information on drug-drug interactions, effects in special populations, and overall real-world effectiveness. The findings from Phase IV studies can lead to changes in drug labeling, dosage recommendations, or even withdrawal of the drug from the market if significant safety concerns arise.


CONCLUSION

Drug discovery is a systematic process aimed at identifying and developing new therapeutic agents. It involves target identification, hit discovery through screening, lead optimization, and preclinical evaluation, ultimately leading to clinical trials. The goal is to create effective and safe medications to address unmet medical needs and improve patient outcomes.




REFERENCES

Bhardwaj, S., & Gupta, D. (2012). Study of acute, sub acute and chronic toxicity test. International Journal of Advanced Research in Pharmaceutical & Bio Sciences1(3), 103-130.

Goodnow Jr, R. A. (2006). Hit and lead identification: Integrated technology-based approaches. Drug Discovery Today: Technologies3(4), 367-375.

Gupta, P., & Verma, A. K. (2023). Basics of Clinical Drug Development: Clinical Trial and Drug Development. In Drugs and a Methodological Compendium: From bench to bedside (pp. 213-234). Singapore: Springer Nature Singapore.

Hughes, J. P., Rees, S., Kalindjian, S. B., & Philpott, K. L. (2011). Principles of early drug discovery. British journal of pharmacology162(6), 1239-1249.

Mohs, R. C., & Greig, N. H. (2017). Drug discovery and development: Role of basic biological research. Alzheimer's & Dementia: Translational Research & Clinical Interventions3(4), 651-657.

Schenone, M., Dančík, V., Wagner, B. K., & Clemons, P. A. (2013). Target identification and mechanism of action in chemical biology and drug discovery. Nature chemical biology9(4), 232-240.

Singh, G. (2018). Preclinical Drug Development. In Pharmaceutical Medicine and Translational Clinical Research (pp. 47-63). Academic Press.

Strickley, R. G. (2008). Formulation in drug discovery. Annual Reports in Medicinal Chemistry43, 419-451.

Sun, D., Gao, W., Hu, H., & Zhou, S. (2022). Why 90% of clinical drug development fails and how to improve it?. Acta Pharmaceutica Sinica B12(7), 3049-3062.

Vrbanac, J., & Slauter, R. (2017). ADME in drug discovery. In A comprehensive guide to toxicology in nonclinical drug development (pp. 39-67). Academic Press. Assessed and Endorsed by the MedReport Medical Review Board





bottom of page