The Scientist’s Guide to Understanding FDA Drug Approval

Last Updated on 

February 18, 2025

By 

Excedr
FDA drug approval process
Table of Contents

Other Posts About Industry Insights

The journey from discovery to FDA approval is one of the most rigorous and comprehensive processes in the pharmaceutical and biotech industries. While drug development promises innovative treatments, from life-saving vaccines to gene therapies, it involves an intensive series of steps and regulatory evaluations. Each stage in the process is designed to guarantee that new therapies meet strict standards for safety, efficacy, and quality before reaching patients.

Understanding the FDA drug approval process is essential for biotech and pharmaceutical companies as they navigate challenges related to clinical trials, regulatory hurdles, and commercialization. This guide will provide an in-depth overview of the FDA’s role, the steps involved in securing drug approval, and the significance of various application types, from New Drug Applications (NDAs) to Emergency Use Authorizations (EUAs).

A Brief History of the FDA

Founded in 1930, the US Food and Drug Administration (FDA) is a federal agency responsible for protecting public health by ensuring the safety, efficacy, and security of human and veterinary drugs, biological products, medical devices, and other products subject to regulation.

In 1938, Congress passed the Federal Food, Drug, and Cosmetics (FDC) Act, which requires that drugs be safe before they are allowed on the market. The Kefauver-Harris Drug Amendment of 1962 further ensured that drugs not only be safe but also effective before they receive market authorization. 

In 1966, the drug division of the FDA was reorganized into the Office of New Drugs (OND), which began reviewing new drug applications. In 1982, the Bureau of Biologics was merged with the FDA. In 1987, two separate entities were established: the Center for Drug Evaluation and Research (CDER) and the Center for Biologics Evaluation and Research (CBER)

Initially formed with six offices, the Center for Drug Evaluation and Research (CDER) has grown to 13 offices to better fulfill its mission of safeguarding public health. Acting as a consumer watchdog, CDER oversees thousands of prescription and over-the-counter drugs, ensuring their safety and efficacy before they reach the market. This includes assessing new drug applications, monitoring post-market safety, and providing scientific support through the FDA’s review process.

The FDA’s drug approval process involves rigorous scientific and clinical testing to evaluate a drug’s benefits, risks, and side effects. Beginning with preclinical testing and advancing through clinical trials, this multi-step process ensures that new medicines are safe and effective for their intended use. CDER continues to monitor drugs after approval, taking corrective action, including removal from the market, if necessary.

Through these standards, the FDA strives to ensure that approved drugs not only meet safety and efficacy benchmarks but also deliver meaningful patient benefits. Post-approval, the FDA can intervene if new safety concerns arise, prioritizing public health at every step.

How Does the Drug Approval Process Work?

The FDA’s drug approval process is carefully designed to ensure that drugs are safe, effective, and appropriate for their intended uses before they reach the public. The process for taking a potential therapeutic compound from initial discovery to final approval spans multiple stages and can last 10-15 years, costing biotech and pharmaceutical companies hundreds of millions, if not billions, of dollars. While estimates vary, studies in publications like the Journal of Health Economics and JAMA Network have placed the median cost of drug development anywhere from $985 million to $2.8 billion.

Here’s a high-level look at the steps involved in the FDA’s drug approval process:

  1. Preclinical Research: Before any human testing, a drug undergoes rigorous laboratory and animal studies to establish baseline data on its safety, pharmacodynamics, and pharmacokinetics. This preclinical phase provides critical insights into whether the drug could potentially be safe and effective in humans.
  2. Investigational New Drug (IND) Application: With promising preclinical results, the sponsor submits an IND application to the FDA. This application must include preclinical data, plans for human trials, and details about the drug’s manufacturing and chemistry. Upon approval, the sponsor can proceed to clinical trials.
  3. Clinical Trials: The drug undergoes three phases of clinical trials:
    • Phase I: Focuses on safety, involving a small group of healthy volunteers (or patients, for specific cases). Researchers look for adverse effects, dosing limits, and basic pharmacology.
    • Phase II: Expands to a larger patient group and starts evaluating the drug’s effectiveness and optimal dosing, while still monitoring for side effects.
    • Phase III: Conducted on an even larger scale with hundreds to thousands of patients, this phase provides the substantial evidence of efficacy and safety needed for FDA approval.
  4. New Drug Application (NDA) Submission: After completing clinical trials, the sponsor submits an NDA, which contains extensive data from all phases of testing, along with the drug’s proposed labeling and manufacturing information.
  5. FDA Review: The FDA’s review teams, often with input from advisory committees, rigorously assess the data, ensuring the benefits outweigh any risks. Depending on the drug’s complexity and novelty, the review process may last months to years.
  6. Post-Market Safety Monitoring: Even after a drug is approved, the FDA continues to monitor its safety through programs like the Adverse Event Reporting System and additional post-market studies. This vigilance helps ensure that any long-term or rare side effects are identified and addressed.

Additional Pathways for Drug Applications

While INDs and NDAs are common, the FDA offers different drug application pathways depending on the drug’s intended use, nature, and novelty. For instance, biologics require a Biologics License Application (BLA), while generic drugs are submitted through an Abbreviated New Drug Application (ANDA). These options, along with mechanisms like Emergency Use Authorizations (EUAs), provide customized routes for drugs to reach the market under various circumstances. Each type of application allows the FDA to cater its review process to the specific drug and its potential role in healthcare.

Through these steps, the FDA balances the need to bring new, effective drugs to patients quickly with the obligation to ensure safety and efficacy, providing essential safeguards for public health.

How Has the FDA’s Approval Methods Changed Over the Years?

The FDA’s drug approval process has evolved considerably throughout the 20th and early 21st centuries. This evolution has been largely driven by the need to accelerate and simplify approvals, especially for drugs addressing unmet medical needs. Key shifts have included incorporating new technologies, adjusting regulatory pathways, and refining assessment standards.

Several factors have influenced these changes. Federal regulations have expanded, drug and device complexities have increased, and the pharmaceutical industry has grown into a major economic force in the United States. To keep pace, the FDA has adopted electronic submissions and integrated real-world data to streamline review processes. Additionally, the agency has introduced a priority review program, expediting approvals for drugs that offer significant advantages over existing treatments. Another major step is the accelerated approval process, which allows drugs to be approved based on surrogate endpoints—early indicators of efficacy—without waiting for complete clinical results.

In recent years, the FDA has also shifted toward a risk-based approach. This approach emphasizes balancing benefits and risks rather than solely evaluating safety and efficacy. The agency has further bolstered international collaboration, working closely with global regulators to enhance access to new drugs worldwide.

A recent study in the Journal of Family Medicine and Primary Care highlights trends in drug approvals over the last two decades. The study notes an uptick in the expedited approval of anticancer drugs and biologics, contrasting with slower approval rates for drugs treating neurological disorders (such as depression and psychosis) and lifestyle diseases like obesity and diabetes. This trend reflects the heightened research focus on developing anticancer therapies and biologics.

While opinions vary on whether these FDA changes are ultimately beneficial or pose risks, they have undoubtedly accelerated the approval timeline, bringing innovative treatments to patients faster while maintaining standards of safety and efficacy.

But First, What Is a Drug?

We all know what a drug is, but to appease the SEO best practices laid out by Google, let's include a quick definition. Who knows, it might even be helpful to review the exact definition, in the eyes of the FDA. According to the agency, a drug is any substance "intended to diagnose, treat, cure, or prevent a disease or condition." This includes prescription medications, over-the-counter remedies, and biologics, such as vaccines and blood products. Importantly, the definition covers substances for human and animal use, as both fall under the FDA’s purview.

However, the FDA does make some distinguishments. For example, it considered drugs to be different from dietary supplements, a category that includes herbal products, vitamins, and complementary therapies. These products, regulated by the Center for Drug Evaluation and Research (CDER) under the Dietary Supplement Health and Education Act of 1994, do not undergo the same rigorous approval process as drugs. Instead, dietary supplements are evaluated based on guidelines for safe manufacturing practices and labeling but are not required to demonstrate efficacy in treating diseases or conditions.

Types of FDA Drug Applications

There are some common types of drug applications the FDA reviews that sponsors can submit to receive approval to sell and market a medical product, including the New Drug Application (NDA), Abbreviated New Drug Application (ANDA), Investigational New Drug application, Biologics License Application (BLA), and Over-the-Counter (OTC) Drug Review.

Additionally, there are mechanisms the FDA can use to quickly authorize the use of a medical product or modify an approved drug that include the Emergency Use Authorization (EUA) and Supplement to a Previously Approved Application.

New Drug Application (NDA)

An NDA is submitted to gain approval to market a new drug. Typically, the sponsor, often a pharmaceutical company, files an NDA when they believe sufficient data on safety and efficacy has been collected to meet FDA requirements.

The process is comprehensive, often taking years and substantial resources to complete, and involves submitting extensive preclinical and clinical data. This application must include study results, detailed manufacturing information, and a proposed drug label. The NDA process works as follows:

  1. Preclinical Studies: Safety, pharmacology, and toxicology are assessed.
  2. Clinical Trials: Safety and efficacy in humans are demonstrated.
  3. Manufacturing Information: Chemistry, manufacturing, and controls (CMC) details are submitted.
  4. Submission and Review: The NDA is reviewed for safety, efficacy, and quality standards. Labeling and manufacturing processes are also evaluated.
  5. Advisory Committee (if necessary): Additional expert opinions may be sought.
  6. Approval: If approved, the drug can be marketed.

NDAs are essential, providing the FDA with crucial data to make informed decisions about new drugs.

Abbreviated New Drug Application (ANDA)

An ANDA is filed to market a generic drug equivalent to an existing brand-name drug. Unlike NDAs, ANDAs typically do not require preclinical and clinical data for safety and efficacy, allowing a faster, more cost-effective approval process.

To obtain approval, the application must prove the generic drug’s equivalence to the brand-name product in safety, efficacy, and quality. Some cases may require bioequivalence studies or additional trials. Once approved, generic drugs provide affordable alternatives and increase competition, lowering prices for consumers.

The ANDA process generally works as follows:

  1. Identify Reference Drug: The sponsor selects a brand-name drug for equivalence testing.
  2. Establish Equivalence: Demonstrate the generic’s similarity in safety, efficacy, and quality.
  3. Manufacturing Data: Detailed CMC information is provided.
  4. Bioequivalence Studies (if needed): Additional testing may confirm biological similarity.
  5. Submission and Review: The FDA reviews for equivalency and regulatory compliance.
  6. Approval: The generic can be marketed, and the reference drug’s exclusivity may end.

ANDAs play a vital role in increasing access to medications and reducing healthcare costs.

Investigational New Drug (IND)

An IND application is submitted to gain approval for testing an investigational drug in human clinical trials, marking the first step toward market authorization. The application, submitted by the drug’s sponsor, provides the FDA with detailed preclinical data on the drug’s safety, pharmacology, and manufacturing processes to confirm its suitability for human trials.

IND applications also permit the legal transport of an investigational drug across state lines for testing. These applications include a proposed trial design, a risk management plan, and an outline of potential benefits, which together assure the FDA that human testing will prioritize safety. The two primary IND types are Commercial INDs (intended for market approval) and Research INDs (aimed at safety and efficacy data collection). Specialized IND categories include:

  1. Investigator IND: Submitted by a physician to study new uses for an approved drug or to test a new drug in a specific patient population.
  2. Emergency Use IND: Allows access to experimental drugs in emergencies when no approved treatments are available.
  3. Treatment IND: Used for drugs showing promise in clinical trials for serious or life-threatening conditions.

The IND application process generally involves:

  1. Preclinical Studies: Conducted in labs and with animals to assess initial safety.
  2. IND Submission: Includes all preclinical data, manufacturing information, and a trial proposal.
  3. FDA Review: The FDA assesses the data to determine if human testing can proceed safely.
  4. Clinical Trials: With IND approval, trials begin in stages, with the FDA actively monitoring safety.

This process is a cornerstone of drug development, ensuring that only drugs with sufficient safety data progress to human testing.

Biologic License Application (BLA)

A Biologic License Application (BLA) is submitted to obtain FDA approval for biologic products—complex molecules produced from living cells, such as vaccines, blood products, and gene therapies. Governed by the Public Health Service Act (PHS Act), BLAs focus on demonstrating the product’s safety, efficacy, and consistent quality for its intended use.

Unlike drugs synthesized chemically, biologics are produced through living systems, making their development and manufacturing more complex. To ensure product integrity, sponsors must provide the FDA with detailed information on the product’s development, manufacturing processes, and clinical trials that establish safety and effectiveness.

The BLA process generally includes the following steps:

  1. Product Development: The biologic is created using living cells or biological systems.
  2. Preclinical and Clinical Studies: Safety and efficacy are evaluated in both preclinical (lab and animal) and clinical (human) studies.
  3. Manufacturing Data: The sponsor details manufacturing processes, facilities, and quality control measures.
  4. Submission and Review: The BLA is reviewed by the FDA to evaluate safety, efficacy, and quality standards.
  5. Approval Decision: If the FDA approves, the biologic can be marketed.

BLAs are critical for bringing innovative biologics to the public, allowing treatments for complex and previously untreatable conditions.

Over-the-Counter (OTC) Drug Review

OTC drugs are non-prescription medications used to treat common ailments, such as headaches, colds, and allergies. To ensure public safety, the FDA runs an OTC Drug Review program, which assesses each drug’s safety, efficacy, and labeling to make certain it’s suitable for self-medication.

OTC drugs evaluated under this program are subject to OTC monographs—guidelines for each therapeutic category that outline ingredients, dosages, labeling, and testing requirements. Drugs that comply with a monograph can be marketed without an NDA, while those that don’t require separate FDA approval.

The OTC Drug Review process typically includes:

  1. Assessment and Classification: Drugs are evaluated, and therapeutic categories are created.
  2. Monograph Development: The FDA establishes monographs detailing safe usage standards.
  3. Public Comment: The FDA opens monographs for public input before finalization.
  4. Final Monograph Issuance: The finalized monograph sets conditions for marketing without a prescription.
  5. Post-Market Monitoring: The FDA monitors OTC drug safety through adverse event reporting and periodic evaluations.

The OTC Drug Review allows manufacturers to bring safe, effective non-prescription drugs to market, enhancing consumer access to affordable healthcare options.

Other Categorizations & Types of FDA Approvals

Beyond traditional drug applications, FDA approvals can also be categorized by product type, including name-brand drugs, generic drugs, and over-the-counter drugs.

  1. Name-Brand Drugs: Approval follows a five-step process—discovery, preclinical research, clinical trials, FDA review, and post-market safety monitoring. Name-brand drugs require a New Drug Application (NDA) for market entry.
  2. Generic Drugs: Generics are submitted through an Abbreviated New Drug Application (ANDA). They bypass certain trials required for name-brand drugs, needing only proof that the active ingredient is identical to the original product.
  3. Over-the-Counter Drugs: OTC products can be approved through an NDA or evaluated under the OTC Drug Review monograph process, which specifies safe usage standards.

In addition to drug approvals, the FDA oversees the approval of medical devices. Device testing often mirrors that of drugs, beginning with laboratory assessments, progressing to animal studies, and finally undergoing human trials. Approved devices are classified by risk level, with higher-risk devices facing more stringent requirements.

This classification system enhances the FDA’s ability to regulate a wide range of products, ensuring safety and effectiveness across pharmaceuticals and medical devices.

Additional FDA Mechanisms

In addition to traditional applications, the FDA has pathways to authorize medical products more quickly, including the Emergency Use Authorization (EUA) and Supplement to a Previously Approved Application.

  • Emergency Use Authorization (EUA): The EUA allows the FDA to temporarily authorize unapproved medical products during a public health emergency, such as a pandemic, when no approved alternatives are available. EUAs expedite access to essential diagnostics, treatments, and vaccines without full FDA approval. The process typically includes:
    1. Declaration of a public health emergency.
    2. Submission of data on safety and efficacy.
    3. FDA review and, if approved, conditional authorization.
    4. Ongoing safety monitoring and potential revision or revocation as new data emerges.
  • Supplement to a Previously Approved Application: A supplement application allows approved drugs to undergo changes—such as label updates, manufacturing adjustments, or new indications—without requiring a completely new approval. This process ensures modifications do not affect the drug’s safety or efficacy. Key steps include:
    1. Submission of data supporting the change.
    2. FDA review to assess the impact on safety and efficacy.
    3. Implementation of changes if approved.

Resources for Submissions

The FDA provides a variety of resources to assist sponsors in navigating the complex submission process, from early research stages through to final application. These resources include guidance documents, regulatory requirements, forms, and specific information on pathways like accelerated approval. Together, they establish clear standards for every aspect of drug development, from preclinical studies and clinical trials to submission requirements for New Drug Applications (NDAs), Biologics License Applications (BLAs), and more.

  • Guidance Documents: FDA guidance documents offer detailed explanations of the agency’s regulatory expectations for each type of application. Although they are not legally binding, these documents help sponsors prepare applications that align with FDA standards, potentially reducing delays in the review process. Key topics covered include clinical trial design, safety reporting, chemistry and manufacturing controls, and specialized pathways like accelerated approval. Sponsors can find guidance documents specific to Investigational New Drug (IND) applications, 505(b)(2) submissions, and Emergency Use Authorizations (EUAs) on the FDA’s website. Regularly updated, these documents reflect evolving scientific and regulatory standards, keeping sponsors informed of best practices. You can search for specific documents as well.
  • Laws, Regulations, & Procedures: The FDA operates under the Federal Food, Drug, and Cosmetic Act, which mandates the protection of public health by ensuring the safety, effectiveness, and quality of drugs and medical devices. The agency enforces these regulations through the Code of Federal Regulations (CFR), particularly in Title 21, which covers food and drug requirements. Title 21 includes binding rules for drug sponsors, covering everything from good manufacturing practices (GMP) to clinical trial standards. By adhering to CFR guidelines, sponsors can ensure compliance with federal laws, reducing the risk of regulatory setbacks.
  • Application Forms and Templates: The FDA provides standardized forms and templates for each stage of the application process, making it easier for sponsors to organize and submit information. For example, Form FDA 1571 is required for IND submissions, while Form FDA 356h is essential for NDAs and BLAs. These forms help structure applications to meet FDA review criteria, ensuring that essential data is presented in an accessible, organized format.
  • Educational Workshops and Webinars: To further support sponsors, the FDA offers educational workshops and webinars on regulatory science, application requirements, and emerging topics in drug development. These sessions provide direct access to FDA experts, who can clarify complex regulatory topics, answer questions, and share insights on recent policy updates. Sponsors can benefit from these events to stay current on regulatory expectations and optimize their submission strategies.
  • Online Portals for Submission Tracking: The FDA has developed online portals, like the Electronic Submission Gateway (ESG) and the CDER NextGen Portal, where sponsors can submit and track their applications. These platforms streamline communication with the FDA, providing updates on submission status and enabling sponsors to respond to additional information requests in real time. These tools improve efficiency, helping sponsors stay on track during the review process.

With these resources, sponsors can prepare applications that not only meet FDA standards but also reduce the likelihood of delays or denials. Utilizing the available guidance and tools allows sponsors to streamline the submission process, increase compliance, and enhance the chances of a successful review.

Why Your Application May Get Approved, Delayed, or Denied

Approval decisions are complex and influenced by multiple critical factors. Primarily, the FDA assesses a drug's safety and efficacy as demonstrated through robust clinical trials. If a drug shows compelling evidence of effectiveness in treating its target condition with manageable risks, it stands a stronger chance of approval. However, several other elements can affect the outcome:

  1. Clinical Trial Design and Data Integrity: Trials must be well-designed, adhere to rigorous standards, and be free from bias or data inconsistencies. The FDA scrutinizes the study design, participant selection, control measures, and statistical analyses used to evaluate efficacy. Poorly designed trials or insufficient data can lead to delays or denials.
  2. Manufacturing Process and Quality Control: The FDA evaluates the proposed manufacturing process to ensure that the drug can be consistently produced to meet safety and quality standards. Any concerns about production methods, equipment, or facilities can result in application delays until compliance with Good Manufacturing Practice (GMP) standards is confirmed.
  3. Drug Stability and Formulation: Drugs must remain stable and effective throughout their intended shelf life. If stability studies or formulations raise concerns about a drug’s potency over time, the FDA may require additional testing to ensure that each batch remains effective and safe for patients.
  4. Benefit-Risk Profile: For each application, the FDA conducts a benefit-risk assessment, weighing the therapeutic advantages of the drug against potential risks. This assessment is particularly relevant for drugs targeting serious, life-threatening, or unmet medical needs. In cases where risks are significant but the drug offers a substantial improvement over existing treatments, the FDA may approve the drug with additional monitoring requirements.
  5. Patient Population and Unmet Medical Need: Drugs that address critical or rare conditions with limited treatment options are often prioritized. For these cases, the FDA may consider a more flexible approach, especially if the drug demonstrates significant potential benefits. Applications for drugs serving high-priority populations may qualify for expedited pathways, including accelerated approval or priority review.
  6. Post-Market Requirements and Risk Management Plans: The FDA may approve a drug on the condition that the sponsor implements a risk management strategy, such as a Risk Evaluation and Mitigation Strategy (REMS). These plans ensure ongoing monitoring of the drug’s safety profile in the real world and can be essential for high-risk medications.
  7. Additional Data Requests and Amendments: During the review process, the FDA may request additional data or clarifications. While not necessarily a denial, these requests can delay the approval timeline as sponsors gather the required information or amend their applications to meet FDA standards.

Approval, delay, or denial is a dynamic process where thorough evaluation safeguards public health, ensuring that only safe, effective, and high-quality drugs reach the market. By meeting FDA requirements and preparing for potential issues, sponsors can improve their chances of a timely and favorable outcome.

Need Essential Lab Equipment to Support FDA Approval?

Navigating the FDA’s drug approval process demands significant resources, from preclinical research through clinical trials. Excedr’s leasing program offers a cost-effective way to secure high-quality lab equipment without depleting capital reserves. By leasing instead of purchasing, labs can maintain financial flexibility, avoid large upfront expenses, and allocate funds more strategically to advance critical research milestones.

If you’re looking for a streamlined approach to acquiring the equipment necessary for rigorous FDA testing and compliance, contact Excedr to learn how our leasing program can support your lab’s success in every phase of the approval process.

Other Posts About Industry Insights

Resources for founders, scientists, and the life sciences community.