🛄Pharma and Biotech Industry Management Unit 2 – Drug & Device Development Process

Drug and device development is a complex, multi-stage process that transforms promising compounds into safe, effective treatments. It involves rigorous testing, regulatory oversight, and collaboration between scientists, clinicians, and regulatory agencies. The process spans discovery, preclinical testing, clinical trials, FDA review, and post-market surveillance. Key concepts include INDs, NDAs, GMPs, and pharmacovigilance. Understanding this process is crucial for professionals in pharma and biotech management.

Key Concepts and Terminology

  • New Drug Application (NDA) submitted to the FDA for approval to market a new drug in the United States
  • Investigational New Drug (IND) application filed with the FDA to begin clinical trials on a new drug
  • Good Manufacturing Practices (GMP) ensure consistent quality and safety in drug manufacturing
  • Pharmacovigilance involves monitoring and reporting adverse drug reactions and side effects
  • Orphan drugs treat rare diseases affecting fewer than 200,000 people in the United States
  • Biomarkers are measurable indicators of biological processes or responses to therapeutic interventions
  • Pharmacokinetics studies how the body absorbs, distributes, metabolizes, and excretes a drug
  • Pharmacodynamics examines the biochemical and physiological effects of a drug on the body

Stages of Drug Development

  • Discovery and Development involves identifying new drug targets and creating compounds that interact with those targets
    • Includes screening large libraries of compounds to find promising lead candidates
    • Preclinical testing assesses safety and efficacy in animal models before human trials
  • Clinical Trials test the safety and effectiveness of a drug in human subjects
    • Phase 1 trials evaluate safety and determine appropriate dosing in a small group of healthy volunteers
    • Phase 2 trials assess efficacy and side effects in a larger group of patients with the targeted condition
    • Phase 3 trials involve large-scale, randomized, controlled studies to confirm safety and efficacy
  • FDA Review and Approval process evaluates the data from clinical trials to determine if the benefits outweigh the risks
    • NDA is submitted with comprehensive data on the drug's safety, efficacy, and manufacturing process
    • FDA may request additional information or studies before making a decision
  • Post-Market Surveillance monitors the drug's safety and effectiveness after it is approved and marketed
    • Adverse events are reported and investigated to ensure ongoing patient safety

Regulatory Framework and Approval Process

  • Food and Drug Administration (FDA) oversees the approval and regulation of drugs and medical devices in the United States
  • European Medicines Agency (EMA) serves a similar role in the European Union
  • Investigational New Drug (IND) application must be filed with the FDA before beginning clinical trials
    • Includes data from preclinical studies, manufacturing information, and clinical trial protocols
  • New Drug Application (NDA) is submitted to the FDA for approval to market a new drug
    • Includes comprehensive data from clinical trials, manufacturing processes, and proposed labeling
  • Priority Review designation accelerates the FDA review process for drugs that offer significant improvements over existing treatments
  • Accelerated Approval pathway allows for earlier approval of drugs for serious conditions based on surrogate endpoints
  • Fast Track designation facilitates the development and expedites the review of drugs for serious conditions with unmet medical needs

Clinical Trials and Testing

  • Randomized Controlled Trials (RCTs) are the gold standard for evaluating the safety and efficacy of new drugs
    • Participants are randomly assigned to receive either the investigational drug or a placebo/standard treatment
    • Double-blind design ensures that neither the participants nor the researchers know who is receiving the active drug
  • Inclusion and Exclusion Criteria define the characteristics of the study population
    • Ensures that the trial results are applicable to the intended patient population
  • Informed Consent is obtained from all participants, ensuring they understand the risks and benefits of the trial
  • Data and Safety Monitoring Boards (DSMBs) independently monitor the trial data to ensure participant safety
  • Adverse Event reporting is crucial for identifying and addressing any safety concerns that arise during the trial
    • Serious Adverse Events (SAEs) must be promptly reported to the FDA and other regulatory authorities

Manufacturing and Quality Control

  • Good Manufacturing Practices (GMP) are a set of regulations and guidelines that ensure the quality and consistency of drug products
    • Covers all aspects of the manufacturing process, from raw materials to finished products
    • Includes requirements for facility design, equipment maintenance, personnel training, and record-keeping
  • Validation is the process of establishing that a manufacturing process consistently produces a product that meets predetermined specifications
    • Includes validation of equipment, analytical methods, and cleaning procedures
  • Quality Control (QC) involves testing and monitoring the manufacturing process to ensure that the product meets the required specifications
    • Includes testing of raw materials, in-process samples, and finished products
  • Stability Testing assesses the ability of a drug product to maintain its quality and potency over its shelf life
    • Conducted under various environmental conditions (temperature, humidity, light) to simulate real-world storage and use

Market Analysis and Commercialization

  • Market Research assesses the potential demand, target patient population, and competitive landscape for a new drug
    • Helps determine pricing, distribution, and promotional strategies
  • Pharmacoeconomics evaluates the cost-effectiveness of a drug in comparison to existing treatments
    • Considers both the direct costs (drug price) and indirect costs (hospitalizations, lost productivity) associated with a disease and its treatment
  • Reimbursement and Pricing strategies aim to balance the need for patient access with the recovery of research and development costs
    • Payers (insurance companies, government programs) often negotiate prices and establish formularies that determine which drugs are covered
  • Marketing and Promotion efforts raise awareness of a new drug among healthcare providers and patients
    • Includes direct-to-consumer advertising (where permitted), physician detailing, and educational programs
  • Patent Protection and Exclusivity periods grant the drug developer a temporary monopoly to recoup their investment
    • Patents typically last 20 years from the date of filing, but exclusivity periods can extend beyond patent expiration for certain drugs (orphan drugs, pediatric indications)

Ethical Considerations and Patient Safety

  • Informed Consent ensures that clinical trial participants understand the risks and benefits of the study and voluntarily agree to participate
    • Includes providing clear, understandable information about the trial and the participant's rights
  • Institutional Review Boards (IRBs) review and approve clinical trial protocols to ensure they meet ethical standards and protect participant rights and welfare
  • Equitable Access to new drugs is a concern, particularly for expensive or limited-supply treatments
    • Compassionate Use programs allow access to investigational drugs for patients with serious conditions who have exhausted other treatment options
  • Post-Market Surveillance is critical for identifying and addressing any safety issues that may not have been detected in clinical trials
    • Adverse Event reporting by healthcare providers and patients helps monitor real-world safety and effectiveness
  • Risk Management Plans outline the strategies for minimizing and mitigating any identified risks associated with a drug
    • Includes labeling requirements, patient education, and restrictions on distribution or use
  • Personalized Medicine tailors drug therapy to an individual's genetic profile, medical history, and other unique characteristics
    • Pharmacogenomics studies how genetic variations influence drug response and helps identify the most effective and safe treatments for each patient
  • Gene Therapy involves modifying or replacing faulty genes to treat or prevent disease
    • Includes techniques such as gene replacement, gene editing (CRISPR), and CAR T-cell therapy for cancer
  • Biologic Drugs are derived from living organisms and include antibodies, enzymes, and vaccines
    • Biosimilars are highly similar versions of approved biologic drugs, offering the potential for increased access and affordability
  • Digital Health Technologies such as wearables, mobile apps, and telemedicine are transforming drug development and patient care
    • Remote monitoring and real-time data collection can improve clinical trial efficiency and post-market surveillance
  • Artificial Intelligence and Machine Learning are being applied to various aspects of drug development, from drug discovery to clinical trial design and analysis
    • Can accelerate the identification of new drug targets, optimize compound selection, and predict patient responses to therapy


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.