Drug development is a complex and lengthy process that involves extensive preclinical and clinical studies. Preclinical studies evaluate the safety and efficacy of a drug candidate before clinical trials in humans. Animal models are commonly used in preclinical studies to predict drug behavior in humans. Bioanalytical methods and in vitro assays are also used to evaluate the pharmacokinetic and pharmacodynamic properties of a drug candidate.
The objective of bridging preclinical data to clinical studies is to ensure that the safety and efficacy of a drug candidate in humans are properly evaluated before the drug is approved for use.
In this blog post, we’ll discuss the importance of bioanalysis and animal studies in drug development, the objective of bridging preclinical data to clinical studies, and the strategies used for integrating preclinical and clinical data.
Bioanalytical methods evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) properties of a drug candidate. Here are the two commonly used bioanalytical methods in drug development:
Ligand-binding assays (LBAs):
Ligand-binding assays provide high sensitivity, specificity, and accuracy. These assays are designed to detect and quantify the drug and its metabolites in biological matrices. LBAs are commonly used to measure drug concentrations in plasma, serum, or other biological fluids.
The two types of LBAs used in bioanalysis are competitive and non-competitive assays. Competitive assays are used for small molecules, while non-competitive assays are used for large molecules like proteins.
Liquid chromatography-mass spectrometry (LC-MS):
Liquid chromatography-mass spectrometry determines the concentration of a drug and its metabolites in biological matrices. LC-MS combines the separation power of liquid chromatography with the detection and identification capabilities of mass spectrometry. It provides high selectivity and sensitivity, making it a preferred method for quantifying small molecules and their metabolites.
In vitro assays are used to assess the PK and PD properties of a drug candidate before in vivo studies. Here are some commonly used in vitro assays:
Absorption, Distribution, Metabolism, and Excretion (ADME) assays:
ADME assays are used to evaluate the PK properties of a drug candidate. These assays assess how a drug is absorbed, distributed, metabolized, and eliminated by the body. ADME assays help identify potential safety and efficacy issues early in drug development.
Receptor binding assays:
Receptor binding assays are used to evaluate the PD properties of a drug candidate. These assays determine how a drug interacts with its target receptor and whether it has the desired effect. Receptor binding assays help identify potential safety and efficacy issues early in drug development.
Animal models are used to evaluate the safety and efficacy of a drug candidate in a living organism.
Selection of appropriate species:
The chosen species should have similar anatomical, physiological, and pharmacological properties as humans. For example, monkeys are commonly used in drug development as their anatomy and physiology are similar to humans.
Consideration of ethical guidelines and animal welfare:
The use of animals in drug development is subject to ethical guidelines and regulations to ensure animal welfare. These guidelines aim to minimize the use of animals, reduce pain and distress, and provide appropriate housing and care.
Let’s break down some of the strategies used for bridging preclinical data to clinical studies:
Allometric scaling:
Allometric scaling predicts drug dosing in humans based on preclinical data. This method involves scaling the animal dose by the animal’s body weight to obtain a human equivalent dose (HED). This approach assumes that the pharmacokinetic and pharmacodynamic properties of the drug are similar across species, and the animal data can be extrapolated to humans.
Physiologically-based pharmacokinetic (PBPK) modeling:
PBPK modeling is a mathematical approach used to predict drug dosing in humans based on preclinical data. This method uses physiological data, such as organ weight and blood flow, to simulate drug distribution in the body. PBPK modeling can also account for inter-species differences in pharmacokinetic and pharmacodynamic properties, making it a more accurate method for predicting drug dosing in humans.
In vitro-in vivo correlation (IVIVC):
IVIVC is a method used to predict drug performance in vivo based on in vitro data. This approach involves correlating the drug concentration in vitro with the drug concentration in vivo. IVIVC can be used to predict the pharmacokinetic and pharmacodynamic properties of a drug candidate in humans based on in vitro data.
Maximum tolerated dose (MTD) from animal studies:
The MTD is the highest dose of a drug that can be administered to animals without causing significant toxicity. The MTD is used as a starting point for determining the FIH dose in humans.
No-observed-adverse-effect-level (NOAEL):
The NOAEL is the highest dose of a drug that does not cause any significant toxicity in animals. It is used to establish the safety margin for the FIH dose in humans.
Human equivalent dose (HED) calculation:
The HED is the dose of a drug that is predicted to have similar pharmacokinetic and pharmacodynamic properties in humans as in animals. It is calculated using allometric scaling or PBPK modeling.
Types of biomarkers:
There are two types of biomarkers: efficacy biomarkers and safety biomarkers. Efficacy biomarkers are used to evaluate the drug’s effectiveness in treating a specific disease, while safety biomarkers are used to evaluate the drug’s safety profile.
Validation and qualification of biomarkers:
Biomarkers need to be validated and qualified before they can be used in clinical studies. Validation involves demonstrating that the biomarker is reliable, accurate, and reproducible. Qualification involves demonstrating that the biomarker is linked to a specific disease and is predictive of clinical outcomes.
Implementing biomarkers in clinical study design:
Biomarkers can be used to guide clinical study design and to monitor the drug’s safety and efficacy. They can be used as inclusion/exclusion criteria for study subjects, as surrogate endpoints for clinical outcomes, and as a tool for dose selection.
Electronic data capture (EDC) systems:
EDC systems allow for real-time data collection and monitoring, increasing the accuracy and efficiency of data analysis.
Biostatistical methods for data analysis:
Biostatistical methods are used to analyze preclinical and clinical data. These methods allow for the identification of trends, patterns, and relationships in the data.
Dose-escalation and de-escalation:
Adaptive clinical trial designs allow for the modification of the study protocol during the trial. Dose-escalation and de-escalation are commonly used in adaptive trial designs to determine the optimal dose of a drug candidate.
Seamless phase transitions:
Seamless phase transitions are used to streamline the drug development process. This approach involves conducting multiple phases of clinical trials concurrently, reducing the time and resources needed to complete the drug development process.
Sample size re-estimation:
Sample size re-estimation is used to modify the sample size of a clinical trial during the trial.
Population PK/PD modeling:
Population PK/PD modeling is a method used to analyze drug exposure and response data from clinical trials. This approach allows for the identification of patient-specific factors that may affect drug exposure and response. Population PK/PD modeling can also be used to optimize dosing regimens for different patient populations.
Model-based drug development:
Model-based drug development integrates preclinical and clinical data to develop predictive models of drug exposure and response.
Identifying potential adverse events:
Preclinical data is used to identify potential adverse events before they occur in clinical trials. This approach allows for the implementation of risk mitigation strategies to prevent adverse events from occurring.
Developing safety monitoring plans:
Preclinical data can be used to develop safety monitoring plans by identifying potential adverse events and developing strategies to mitigate risks.
Implementing risk mitigation strategies:
Preclinical data can be used to develop risk mitigation strategies by identifying potential adverse events and developing strategies to mitigate risks.
Here are the regulatory considerations for bridging preclinical and clinical studies:
Preclinical data requirements:
Preclinical data requirements are necessary for the submission of an IND application. These data requirements include information on the drug’s pharmacology, toxicology, and pharmacokinetics.
Clinical study protocol:
The clinical study protocol is a critical component of the IND application. The protocol should include detailed information on the study design, patient population, dosing regimen, and safety monitoring plan.
Chemistry, manufacturing, and controls (CMC) information:
The CMC information includes information on the drug substance and drug product, manufacturing process, and quality control.
Pre-IND meetings:
Pre-IND meetings with regulatory agencies are important for discussing the drug development plan and obtaining feedback from regulatory agencies.
Interactions during clinical study progression:
Interactions with regulatory agencies during the clinical study progression are important for addressing safety concerns and ensuring compliance with regulatory requirements.
Post-marketing safety monitoring:
Post-marketing safety monitoring is critical for identifying and addressing safety concerns that may arise after the drug is approved for use.
Examples of Successful Drug Development Programs:
Imatinib (Gleevec):
Imatinib is a small molecule drug used to treat chronic myeloid leukemia (CML) and gastrointestinal stromal tumors (GISTs). Imatinib was developed using a translational research approach that involved preclinical studies in animal models and clinical studies in humans.
The preclinical studies demonstrated the drug’s efficacy and safety, and the clinical studies confirmed these findings. Imatinib was approved by the FDA in 2001 and has since become a standard treatment for CML and GISTs.
Pembrolizumab (Keytruda):
Pembrolizumab is a monoclonal antibody used to treat various types of cancer, including melanoma, non-small cell lung cancer, and head and neck cancer. Pembrolizumab was developed using a translational research approach that involved preclinical studies in animal models and clinical studies in humans.
The preclinical studies demonstrated the drug’s efficacy and safety, and the clinical studies confirmed these findings. Pembrolizumab was approved by the FDA in 2014 and has since become a standard treatment for various types of cancer.
Translational research is essential for successful drug development:
Both Imatinib and Pembrolizumab were developed using a translational research approach that involved preclinical studies in animal models and clinical studies in humans.
Preclinical data should be sufficient to support clinical studies:
The preclinical data for both Imatinib and Pembrolizumab were sufficient to support clinical studies in humans.
Utilizing advanced technologies:
Advances in technologies such as in vitro organ models, microfluidic devices, and artificial intelligence can improve the predictive value of preclinical data, accelerating drug development and improving the translational success rate.
Enhancing collaborations:
Collaborations between industry, academia, and regulatory agencies can improve the design and execution of preclinical and clinical studies.
Early engagement with regulatory agencies:
Early engagement with regulatory agencies can ensure that the drug development plan meets regulatory requirements and can accelerate the drug development process.
In conclusion, bridging preclinical data to clinical studies is critical for the successful development of safe and effective drugs. It allows for the identification of potential safety concerns and optimization of dosing regimens for different patient populations.
Throughout this blog post, we have discussed the key points in designing bioanalytical and animal studies, translating preclinical data to clinical studies, integrating preclinical and clinical data, regulatory considerations, and case studies of successful drug development programs. We have also highlighted the importance of using advanced technologies, enhancing collaborations, and early engagement with regulatory agencies to improve the translational success rate of drug development.
At InfinixBio, we have a proven track record of overcoming lab-related challenges while efficiently guiding clients on a successful regulatory pathway. Our workforce consists of scientists and professionals who have diverse backgrounds. Located in the Midwest, our team has experience in fields ranging from drug discovery, to pharmacology, and clinical diagnostics.
Get in touch with us today.
Our experienced lab team is here to help. Reach out today to learn more.