Fact-checked by Grok 2 weeks ago

Center for Drug Evaluation and Research

The Center for Drug Evaluation and Research (CDER) is a division of the United States Food and Drug Administration (FDA) tasked with regulating the majority of prescription, over-the-counter, generic, and certain biological drug products to ensure their safety, efficacy, and quality for public use. Established through the evolution of FDA's drug regulatory functions dating back to the early 20th century, CDER evaluates new drug applications, oversees clinical trials, and monitors post-approval safety via adverse event reporting and risk management programs. Its core mandate involves reviewing scientific data from manufacturers to approve only those products demonstrating benefits outweighing risks, thereby preventing the marketing of ineffective or hazardous medications while facilitating access to beneficial therapies. CDER's operations encompass thousands of annual reviews, including expedited pathways for therapies and initiatives to modernize processes through enhanced , , and of advanced technologies like . Notable achievements include the approval of life-saving treatments across therapeutic areas, such as antivirals and biologics, which have significantly improved outcomes by providing evidence-based options previously unavailable. However, the center has faced scrutiny for systemic challenges, including resource constraints, lengthy approval timelines that can delay innovation, and instances where post-market issues revealed gaps in pre-approval risk assessments, as highlighted in broader FDA critiques of underfunding and organizational hurdles. These tensions underscore ongoing debates about striking an optimal balance between rigorous safety standards and timely access to new drugs, with CDER continually adapting through policy reforms and stakeholder engagement to address such concerns.

Mandate and Operations

Core Responsibilities

The Center for Drug Evaluation and Research (CDER) is responsible for regulating the majority of human marketed , including prescription medications, drugs, over-the-counter (OTC) products, and certain biological therapeutics such as biosimilars, but excluding , products, and therapies which fall under the Center for Biologics Evaluation and Research. This oversight extends to diverse items like fluoride toothpaste, antiperspirants, shampoos, and sunscreens when they meet the definition of a drug under . CDER's mission centers on protecting and promoting by ensuring these drugs are safe, effective for their intended uses, manufactured to quality standards, and accurately labeled, while facilitating access to beneficial therapies. A primary function involves evaluating manufacturer-submitted data on drug safety and efficacy without conducting independent testing; CDER reviews applications such as New Drug Applications (NDAs) for novel , Abbreviated New Drug Applications (ANDAs) for generics—which account for approximately 88% of U.S. prescriptions—and Biologics License Applications (BLAs) for certain biologics, assessing whether benefits outweigh risks based on clinical trials and other evidence. Approvals require demonstration of therapeutic equivalence for generics and rigorous benefit-risk analysis for new , incorporating input from advisory committees of independent experts. CDER also enforces truthful advertising for prescription and oversees labeling to prevent misleading claims. Post-approval, CDER conducts surveillance to monitor real-world performance, analyzing reports submitted via systems like MedWatch and initiating actions such as label updates, restricted distribution, or market withdrawal if new risks emerge. Additional responsibilities include promoting generic competition to enhance affordability—though CDER lacks pricing authority—facilitating pediatric studies, and enabling to investigational s for serious conditions. These activities support ongoing research, development, manufacturing compliance, and international efforts to maintain .

Drug Review and Approval Processes

The drug review and approval processes of the Center for Drug Evaluation and Research (CDER) evaluate applications to determine whether new drugs demonstrate safety and efficacy sufficient to outweigh known risks for their intended uses. CDER primarily oversees approvals for small-molecule pharmaceuticals, over-the-counter drugs, and certain therapeutic biologics transferred from the Center for Biologics Evaluation and Research, excluding and blood products. Sponsors must first conduct preclinical studies, including laboratory and , to generate data on , , and before human trials. To initiate clinical testing, pharmaceutical sponsors submit an application to CDER, providing preclinical , manufacturing information, and proposed clinical protocols. CDER reviews the IND within 30 days to assess whether trials can proceed without unreasonable risk; if no hold is issued, 1 trials begin, typically involving 20-100 healthy volunteers to evaluate , dosage, and side effects. 2 trials expand to 100-300 patients with the target condition, focusing on and further , while 3 trials enroll hundreds to thousands of participants across diverse populations to confirm effectiveness, monitor adverse reactions, and compare against existing treatments or placebos. These phases must adhere to standards, with ongoing IND amendments for protocol changes or new . Following successful Phase 3 trials, sponsors submit a () or, for biologics under CDER jurisdiction, a Biologics License Application (BLA), compiling comprehensive data on chemistry, manufacturing controls, nonclinical studies, clinical results, labeling, and risk management plans. CDER's Office of New Drugs (OND) leads the review, assembling multidisciplinary teams of physicians, statisticians, chemists, pharmacologists, and other specialists to scrutinize the submission for filing within 60 days; incomplete applications may receive a refuse-to-file determination. If filed, standard review targets completion in 10 months, while priority review for drugs addressing unmet needs shortens this to 6 months under (PDUFA) goals, with user fees funding the process since 1992. Reviews include facility inspections, assessments for modified formulations, and evaluations of proposed indications, with approvals specifying conditions of use and often requiring post-approval commitments like additional studies. For generic drugs, CDER processes Abbreviated New Drug Applications (ANDAs), which demonstrate to reference listed drugs without repeating full clinical efficacy trials, relying instead on pharmaceutical equivalence and comparative bioavailability studies. Expedited pathways accelerate reviews for serious conditions: designation offers intensive guidance for preliminary evidence of substantial improvement; accelerated approval permits endpoints or clinical benefits likely to predict outcomes, with confirmatory trials required post-approval; halves standard timelines; and fast track facilitates rolling reviews for frequent meetings and eligibility for others. These mechanisms, expanded under PDUFA reauthorizations, aim to balance innovation with rigor, though accelerated approvals have drawn scrutiny for reliance on surrogates that may not fully correlate with clinical benefits in all cases.

Organizational Structure

Key Offices and Divisions

The Center for Drug Evaluation and Research (CDER) operates through a structure of specialized offices and divisions that handle , review, approval, , and post-market monitoring. These components ensure compliance with statutory requirements under the Federal Food, Drug, and Cosmetic Act, as amended, focusing on , , and standards for prescription, over-the-counter, and drugs. The organizational framework has evolved through periodic restructurings to address workload demands and scientific advancements, with recent updates as of 2024 enhancing efficiency in areas like oversight and surveillance. Office of New Drugs (OND) oversees the review of new drug applications (NDAs) and biologics license applications (BLAs) for innovator products, evaluating clinical data on and across therapeutic areas. It includes eight offices covering specialties such as , , and infectious diseases, with 27 review divisions conducting multidisciplinary assessments. OND's decisions determine market access, often under accelerated timelines via programs like , which targets drugs addressing unmet medical needs within six months of filing. Office of Generic Drugs (OGD) manages abbreviated new drug applications (ANDAs) for generic equivalents, verifying bioequivalence to branded drugs without redundant clinical trials. Divided into sub-offices for research, regulatory operations, and therapeutic equivalence, OGD approved 1,020 ANDAs in fiscal year 2023, facilitating cost savings estimated at $313 billion annually for the U.S. healthcare system through increased generic availability. Office of Pharmaceutical Quality (OPQ) enforces current good manufacturing practices (cGMP) and assesses product quality throughout the lifecycle, including inspections and chemistry, manufacturing, and controls (CMC) reviews. Reorganized in 2016 to centralize expertise, OPQ conducts over 1,500 domestic and foreign inspections yearly to mitigate risks like contamination or adulteration. Office of Surveillance and Epidemiology (OSE) monitors post-approval via the FDA Adverse Event Reporting System (FAERS), which received 1.9 million reports in , enabling signal detection and label updates or withdrawals. It collaborates with OND on risk evaluation and mitigation strategies (REMS) for high-risk drugs. Office of Translational Sciences (OTS) supports early-stage through research in , , and , including tools to predict human responses and reduce reliance. OTS divisions focus on innovative methods like integration for regulatory decisions. Supporting offices include the Office of the Center Director (OCD), which provides overall leadership and policy coordination; Office of Compliance, handling enforcement actions like warning letters and seizures for violations; and Office of Executive Programs, managing strategic initiatives and external partnerships.

Leadership and Decision-Making

The Center for Drug Evaluation and Research (CDER) is led by a director who reports to the and holds ultimate responsibility for the center's operations, including drug safety, efficacy evaluations, and policy implementation. As of July 21, 2025, Tidmarsh, M.D., Ph.D., serves as director, marking his first role in government after a career spanning clinical , , and academia at . Tidmarsh oversees a structure that includes deputy directors and heads of specialized offices, such as Marta Sokolowska, Ph.D. (Deputy Center Director for Substance Use and Behavioral Health), Patrick Raulerson, J.D. (Director, Office of Regulatory Policy), and ShaAvhrée Buckman-Garner, M.D., Ph.D. (Director, Office of Translational Sciences). Decision-making within CDER follows a hierarchical and multidisciplinary framework, with routine operational authority delegated to office and division directors while strategic oversight resides with the center director. Key offices, including for innovative therapies and the Office of Generic Drugs for bioequivalents, conduct primary reviews through teams of clinicians, pharmacologists, statisticians, chemists, and other specialists who assess clinical data, manufacturing quality, and risk-benefit profiles submitted via New Drug Applications (NDAs) or Abbreviated New Drug Applications (ANDAs). Approvals require affirmative determinations that a benefits outweigh its known risks, with final authority vested in CDER rather than external entities. For novel or high-stakes applications, CDER often convenes independent advisory committees composed of external experts to provide non-binding recommendations on scientific and medical matters, ensuring decisions incorporate diverse expertise while maintaining agency autonomy. The Office of Regulatory Policy, under its director, coordinates cross-center guidance and chairs the Strategic Policy Council to align decisions with evolving statutory mandates like the (PDUFA). This process emphasizes evidence-based assessments, with escalation to director for disputes or policy implications, though day-to-day approvals are typically executed at the division level to expedite reviews within statutory timelines.

Historical Development

Origins in Early FDA Regulation (1906–1962)

The Pure Food and Drugs Act of 1906, signed into law by President Theodore Roosevelt on June 30, prohibited the interstate shipment of adulterated or misbranded drugs, marking the federal government's initial foray into drug oversight. Enforcement fell to the U.S. Department of Agriculture's Bureau of Chemistry, which established a dedicated Drug Laboratory under Director Lyman F. Kebler to analyze samples for purity, potency, and compliance with U.S. Pharmacopeia standards. The lab's divisions—including inspection, synthetic products, essential oils, and pharmacology—conducted post-market testing, leading to seizures and prosecutions of non-compliant products like patent medicines containing undeclared narcotics or exaggerated claims. Labels were required to disclose quantities of 11 hazardous substances, such as alcohol, opium, morphine, cocaine, and heroin, but no pre-market approval or safety testing was mandated, relying instead on reactive measures against fraud and contamination. Early enforcement faced judicial hurdles, exemplified by a 1911 ruling in United States v. Johnson that the Act did not prohibit false therapeutic claims absent proof of adulteration or misbranding. This prompted the Sherley Amendment of 1912, which banned labels with false or fraudulent therapeutic statements intended to deceive, though proving intent remained challenging and limited prosecutions. By the , drug seizures increased amid growing scrutiny of proprietary remedies, but the Bureau's reactive approach struggled with the burgeoning pharmaceutical market. In 1927, regulatory functions separated from research, forming the Food, Drug, and Insecticide Administration (renamed FDA in 1930), which continued laboratory-based evaluations without proactive safety requirements. The tragedy of 1937, where over 100 deaths resulted from a solvent in an untested liquid formulation of the , exposed critical gaps in safety oversight. This catalyzed the Federal Food, Drug, and Cosmetic (FD&C) Act of 1938, signed on June 25, which for the first time required manufacturers to submit evidence of a new drug's safety via a () before marketing. The FDA gained authority to demand pharmacological data and halt unsafe products, shifting drug evaluation toward pre-market while retaining labeling and adulteration controls; , however, remained unregulated. Subsequent refinements included the Durham-Humphrey Amendment of 1951, which authorized the FDA to classify as prescription-only if unsafe for self-use, mandating oversight and labeling accordingly. By 1962, amid rising concerns over —fueled by congressional hearings and the birth defects crisis—the process had evolved into a formalized , laying the groundwork for modern functions later centralized in the Center for Drug Evaluation and Research. This era's emphasis on empirical testing and federal intervention established causal links between inadequate regulation and public harm, prioritizing verifiable data over manufacturer self-certification.

Expansion and Reforms Post-Thalidomide (1962–1990)

The disaster, which caused severe birth defects in thousands of children worldwide after the drug's approval in Europe without adequate U.S. testing, prompted swift legislative action in the United States. On October 10, 1962, Congress enacted the Kefauver-Harris Amendments to the Federal Food, Drug, and Cosmetic Act, mandating that drug manufacturers prove both safety and efficacy through "adequate and well-controlled investigations" before marketing, shifting the evidentiary burden from the FDA to sponsors and requiring premarket approval for new drugs. These amendments also imposed requirements for in clinical trials, enhanced FDA authority over drug advertising, and authorized regulations for good manufacturing practices with mandatory facility inspections. In response, the FDA's Division of Pharmacology—predecessor to modern drug review units—expanded its capacity to implement these standards, initiating a retrospective Drug Efficacy Study Implementation (DESI) program; in 1966, the agency contracted the to assess the efficacy of approximately 4,000 drugs approved between 1938 and 1962, resulting in the withdrawal or relabeling of hundreds of ineffective products by the early 1980s. This effort, completed through DESI notices starting in 1968, underscored the amendments' causal impact on removing substandard therapies while necessitating a substantial increase in FDA review personnel and expertise to handle both legacy and new applications. Organizational reforms followed to accommodate the heightened regulatory demands. By the late , FDA drug oversight consolidated under evolving structures, culminating in the establishment of the Bureau of Drugs in the early to centralize evaluation, separating it from broader medical bureau functions. Key expansions included the 1970 requirement for patient package inserts on high-risk drugs like oral contraceptives, enhancing post-approval communication, and the 1972 launch of the Over-the-Counter (OTC) Drug Review to categorize and standardize non-prescription products for safety and labeling, involving public advisory panels and monograph development. The 1970 further integrated FDA input into drug scheduling for abuse potential, expanding surveillance to narcotics and psychotropics. These changes reflected a causal progression from efficacy mandates to proactive categorization, with the Bureau of Drugs growing to manage surging applications amid advancing . The 1980s brought targeted reforms addressing innovation barriers and access issues, driven by critiques of approval delays under stringent post-1962 standards. The incentivized development for rare diseases affecting fewer than 200,000 Americans, offering seven years of market exclusivity, tax credits, and protocol assistance, leading to over 200 orphan designations by 1990. Complementing this, the 1984 Drug Price Competition and Patent Term Restoration Act (Hatch-Waxman) streamlined generic entry via abbreviated new drug applications demonstrating rather than full clinical trials, while restoring up to five years of life for innovators lost to regulatory review; this balanced incentives, spurring generic growth from 19% in 1984 to over 50% by 1990 and paving the way for the Office of Generic Drugs in 1989. In 1987, revised investigational drug regulations expanded compassionate use for patients with serious conditions, influenced by the AIDS epidemic, allowing treatment protocols outside formal trials. These reforms culminated in the FDA's 1987 reorganization, formally creating the Center for Drug Evaluation and Research (CDER) from the Bureau of Drugs to specialize in chemical entity oversight, excluding biologics transferred to a separate center, thereby institutionalizing expanded review infrastructure with dedicated offices for new drugs, compliance, and research. The 1988 Prescription Drug Marketing Act further bolstered integrity by prohibiting resale of samples and mandating pedigree tracking to curb counterfeits.

Modernization and PDUFA Era (1992–Present)

The (PDUFA), enacted on October 27, 1992, authorized the (FDA) to collect fees from pharmaceutical manufacturers to fund the review of new drug applications, addressing chronic backlogs at the Center for Drug Evaluation and Research (CDER). Prior to PDUFA, CDER faced resource constraints that extended standard review times to approximately 30 months, contributing to delays in for approved therapies. By fiscal year 1993, PDUFA revenues enabled CDER to hire additional reviewers and implement performance goals, reducing the median approval time for drugs to 10 months by the late 1990s. This shift marked a foundational modernization, prioritizing while maintaining safety standards through structured commitments negotiated with industry stakeholders. Subsequent reauthorizations every five years expanded PDUFA's scope, integrating enhancements to CDER's processes. PDUFA II, effective in 1997 alongside the FDA Modernization Act (FDAMA), introduced advisory committees for expert input on complex applications and pediatric study incentives, further streamlining reviews while mandating post-market safety monitoring. By PDUFA III in 2002, approval times had declined to under 15 months on average, with CDER benefiting from fees comprising over 60% of its for reviews. Later iterations, such as PDUFA V (2012) and VI (2017), incorporated real-time tools and accelerated pathways for breakthrough therapies, enabling CDER to approve novel treatments like targeted cancer drugs more rapidly without evident increases in withdrawal rates for safety reasons. PDUFA VII, signed into law on September 30, 2022, continues to support CDER's modernization through commitments to enhance review capacity for complex modalities like gene therapies and address vulnerabilities exposed by the . Empirical data indicate that since 1992, PDUFA has halved overall approval timelines, from 24-25 months to 10-12 months, correlating with increased annual new molecular entity approvals—rising from 18 in 1992 to peaks exceeding 50 in recent years—while post-approval withdrawals remained low at about 2-3% of approvals. These reforms have bolstered CDER's role in balancing innovation with oversight, though ongoing negotiations for PDUFA VIII emphasize sustaining independence amid industry funding reliance.

Achievements in Drug Regulation

Landmark Approvals and Safety Enhancements

The Center for Drug Evaluation and Research (CDER) has overseen approvals of transformative drugs that addressed previously untreatable conditions through from clinical trials. In 1945, the Penicillin Amendment established FDA certification requirements for safety and effectiveness, leading to the approval of penicillin for civilian use after wartime production; this enabled widespread treatment of bacterial infections, reducing mortality from diseases like and by leveraging the drug's bactericidal mechanism against susceptible pathogens. On March 19, 1987, CDER approved (AZT), the first antiretroviral agent for , based on survival benefits observed in a 282-patient placebo-controlled trial showing delayed disease progression despite risks in up to 25% of recipients. Similarly, on May 10, 2001, CDER granted accelerated approval to imatinib mesylate (Gleevec) for chronic myeloid leukemia, targeting the in 95% of cases and achieving complete cytogenetic responses in 60-80% of chronic-phase patients in phase II studies. CDER's safety enhancements have evolved to balance innovation with risk mitigation, grounded in post-approval data and causal assessments of adverse events. The 1962 Kefauver-Harris Amendments, prompted by thalidomide-induced birth defects (affecting over 10,000 infants globally), mandated proof of efficacy alongside safety via adequate, well-controlled investigations, fundamentally strengthening pre-market requirements and reducing approvals of ineffective therapies. In 2007, the FDA Amendments Act authorized CDER to mandate Risk Evaluation and Mitigation Strategies (REMS) for high-risk drugs, incorporating elements like patient registries and restricted distribution to minimize harms, as applied to over 50 products by 2010 including to prevent teratogenicity. Further advancements include the 2008 Sentinel Initiative, which CDER utilizes for active post-market surveillance across 200 million lives' data, enabling real-time signal detection for issues like cardiovascular risks with , leading to label updates or withdrawals when benefit-risk ratios shift unfavorably. These measures, informed by rather than unsubstantiated assumptions, have iteratively refined drug safety profiles, though empirical critiques note occasional delays in acting on emerging signals due to reliance on voluntary reporting.

Contributions to Public Health Outcomes

The Center for Drug Evaluation and Research (CDER) has advanced by approving drugs that address high-burden conditions, leading to quantifiable reductions in disease-specific mortality and morbidity. Through rigorous pre-market evaluations, CDER ensures that only therapies demonstrating and acceptable profiles reach the market, enabling widespread adoption that correlates with improved population-level outcomes. For example, the approval of antiretroviral drugs has markedly decreased /AIDS-related deaths; since the initial approval of in 1987 and subsequent combinations enabling highly active antiretroviral therapy in the mid-1990s, these therapies have reduced associated mortality, transforming from a near-uniformly fatal condition to a manageable chronic illness. In , CDER's approvals of therapies, starting with in 1987, have contributed to declines in heart disease mortality. lower and reduce the risk of cardiovascular events, including death, heart attack, and by about 25% in at-risk populations, as evidenced by long-term clinical use and observational data. This class of drugs, alongside post-approval expansions of indications, has underpinned broader reductions in U.S. cardiovascular mortality rates, which fell by over 30% from 1980 to 2010 amid increased utilization. CDER's facilitation of generic drug approvals further amplifies these impacts by promoting affordability and access, allowing therapies to scale across diverse populations without prohibitive costs. In rare diseases, targeted approvals like fosdenopterin (Nulibry) in 2021 have directly lowered mortality risks, such as in molybdenum cofactor deficiency, where the drug reduced death rates in clinical settings. Recent expansions, including (Wegovy) in 2024 for reducing in obese adults, continue this pattern by addressing obesity-related comorbidities that strain public health systems.

Controversies and Criticisms

Involvement in the Opioid Crisis

The Center for Drug Evaluation and Research (CDER) played a central role in approving extended-release formulations that facilitated the initial surge in prescribing during the 1990s and early 2000s. CDER approved Purdue Pharma's OxyContin ( hydrochloride extended-release tablets) on December 21, 1995, for the management of moderate-to-severe when continuous analgesia was needed for an extended period, based on clinical trials demonstrating but with limited data on long-term abuse potential. The approval label included language stating that "delayed absorption...will reduce the abuse liability of a widely abused ," and referenced studies claiming addiction rates below 1% in medically treated patients, which drew from a single-paragraph letter in a rather than comprehensive epidemiological evidence. This framing minimized warnings about misuse, enabling broad marketing to physicians for non-cancer , despite opioids' known risks of dependence established since the 1960s. Critics argue that CDER's review process inadequately scrutinized and diversion risks, approving high-potency, 12-hour dosing schedules without mandating restrictions to opioid-tolerant patients or robust post-approval . For instance, prior to OxyContin, CDER had approved other extended-release opioids like MS Contin ( sulfate) in 1987, but without requirements for tamper-resistant formulations or risk evaluation and mitigation strategies (REMS), which were not implemented until 2007 and expanded in 2011. A 2003 report highlighted that OxyContin's approval and subsequent promotion contributed to rising reports by 2002, with over 7 million prescriptions dispensed annually by then, yet CDER's pre-approval assessments relied heavily on manufacturer-submitted data without independent verification of . Subsequent analyses, including a 2020 review in the AMA Journal of Ethics, contend that stricter labeling—limiting indications to severe, —could have curbed the 300% rise in opioid prescriptions from 1999 to 2010, which correlated with over 500,000 overdose deaths linked to prescription s by 2020. Pharmaceutical influence on CDER decisions has drawn scrutiny, including consulting firms like advising both manufacturers and CDER on approval processes, raising concerns over conflicts that may have prioritized over safety signals. Congressional investigations in revealed McKinsey's dual role in optimizing Purdue's submissions to CDER while later consulting for the agency, potentially undermining impartiality in evaluating abuse-deterrent claims. While CDER later withdrew original OxyContin approval in 2013 after Purdue reformulated it to deter crushing, this followed years of crisis escalation, with prescription -involved deaths rising from 3,442 in 1999 to 17,029 in 2017. These approvals, grounded in efficacy-focused trials but deficient in real-world risk modeling, exemplify how regulatory emphasis on access under the (PDUFA) framework may have inadvertently amplified causal factors in the epidemic, including overprescribing driven by optimistic label interpretations.

Regulatory Capture and Pharmaceutical Influence

The phenomenon of at the Center for Drug Evaluation and Research (CDER) manifests primarily through the "" between agency personnel and the , where former regulators frequently join firms they previously oversaw, potentially prioritizing industry interests in decision-making. A 2023 study found that pharmaceutical companies hiring ex-FDA employees experience increased rates of approvals, elevating firm value by influencing outcomes. Similarly, of 28 approvals revealed that 11 of 16 former FDA medical examiners involved were later employed by or consulting for the sponsoring companies, raising concerns about impartiality in CDER's evaluation processes. CDER's reliance on industry-funded user fees under the (PDUFA), enacted in 1992 and reauthorized periodically, has intensified these dynamics by constituting nearly half of the agency's drug review , enabling faster approvals but fostering on pharmaceutical payers. Critics argue this aligns CDER more closely with timelines over rigorous assessments, as PDUFA deadlines have altered approval decisions, correlating with subsequent safety withdrawals. For instance, from fiscal years 2023-2027, PDUFA commitments include performance goals tied to fee collections, which some analyses link to resource imbalances favoring industry-submitted data. Pharmaceutical lobbying further exacerbates influence, with the industry expending over $372 million on federal in 2022 alone, often targeting FDA policies. indicates that firms increasing lobbying expenditures secure more FDA approvals, with one analysis showing a 67.3% higher approval probability for active lobbyists from 1998-2013. Between 2012 and 2019, lobbying activities correlated with less severe FDA recall classifications for defective products, suggesting softened enforcement. Such patterns underscore causal links between financial influence and regulatory leniency, though proponents of PDUFA and lobbying contend they enhance efficiency without compromising core standards.

Emergency Approvals and Post-Market Surveillance Failures

The Center for Drug Evaluation and Research (CDER) has issued Use Authorizations (EUAs) for drugs during crises, allowing provisional use of unapproved products or off-label applications when no adequate alternatives exist and benefits outweigh known risks. During the , CDER granted an EUA for on May 1, 2020, for hospitalized adults and children with severe disease, citing interim data from the ACTT-1 trial indicating a recovery time reduction of 4 days versus . Full approval followed on October 22, 2020, based on further review of clinical trials, though subsequent observational studies identified risks such as in up to 16.3% of treated patients, prompting calls for enhanced monitoring. Similarly, EUAs for and sulfate, issued March 28, 2020, for hospitalized patients, were revoked on June 15, 2020, after randomized controlled trials, including and ORBITA, showed no mortality benefit and heightened risks of ventricular arrhythmias and prolongation. These revocations highlighted challenges in EUA evidence thresholds, with critics noting reliance on limited, non-peer-reviewed data amid political pressures. CDER's post-market surveillance, primarily through passive systems like the FDA (FAERS), aims to detect signals of rare or delayed but is hampered by underreporting—estimated at less than 10% of serious events—and biases toward consumer-submitted "drug ineffective" reports that often lack clinical utility. A notable failure involved (Vioxx), approved by CDER on May 20, 1999, for pain relief; it was voluntarily withdrawn by Merck on September 30, 2004, after the APPROVe trial revealed a 46% increased of confirmed cardiovascular events (e.g., , ) after 18 months of use compared to . Internal FDA reviews later acknowledged overlooked early signals from trials like VIGOR (2000), which showed a threefold higher rate, attributing delays to inadequate post-approval assessment protocols. analyses have criticized CDER's surveillance as insufficiently proactive, recommending mandatory active monitoring and better integration of advisory committee input to mitigate such gaps. These episodes underscore systemic limitations in CDER's framework, including resource constraints and dependence on manufacturer-reported data, which a 2005 analysis deemed flawed for objective post-marketing evaluations due to incomplete ascertainment. Despite reforms like the 2007 FDA Amendments Act mandating Risk Evaluation and Mitigation Strategies (REMS) for high-risk drugs, persistent under-detection of harms—evident in over 500 post-approval actions since 2000—has fueled debates on balancing expedited access with rigorous oversight.

Impact and Reforms

Broader Effects on Innovation and Access

CDER's regulatory framework, while designed to prioritize and , has imposed substantial time and financial burdens on pharmaceutical development, thereby influencing the pace and direction of . Empirical analyses indicate that FDA approval processes, overseen by CDER for the majority of new s, contribute to average development timelines exceeding 10 years and costs surpassing $2 billion per approved , with accounting for a significant portion of these expenditures. These hurdles disproportionately deter investment in treatments for diseases or small populations, where returns may not justify the fixed costs of navigating CDER's requirements, resulting in fewer therapies entering clinical pipelines. Consequently, studies have documented reduced in established therapeutic categories, as firms shift resources toward incremental modifications of existing drugs rather than groundbreaking first-in-class innovations. Reforms such as the (PDUFA), enacted in 1992 and periodically reauthorized, have mitigated some delays by funding additional CDER review staff through industry fees, reducing standard approval times from over 30 months pre-PDUFA to around 10-12 months by the . This acceleration has facilitated earlier market entry for over 1,500 new drugs and biologics since PDUFA's inception, enhancing patient access to therapies like targeted cancer treatments approved in record time during the . However, PDUFA's reliance on pharmaceutical funding raises concerns of , potentially prioritizing high-volume drugs over niche innovations, while post-approval exclusivity periods granted by CDER enable price premiums that can limit affordability, with U.S. drug prices remaining 2-3 times higher than in comparable markets due to these monopolistic dynamics. Beyond direct approvals, CDER's stringent post-market surveillance and restrictions on off-label uses have broader ripple effects on access, often delaying or denying patients investigational treatments through programs, which comprised less than 1% of serious disease cases annually from 2005-2015. Economic modeling further reveals opportunity costs, including an estimated 560,000 additional deaths from delayed approvals between 1999-2019, as regulatory friction diverts resources and discourages risk-taking in high-uncertainty fields like therapies. Initiatives like the Center for Clinical Trial Innovation (established 2024) aim to foster adaptive trial designs, but persistent approval uncertainties continue to erode U.S. leadership in biopharma R&D relative to less regulated jurisdictions.

Recent Initiatives and Future Challenges

In recent years, CDER has advanced its review processes through the 21st Century Review Initiative, which establishes performance standards for multi-office drug evaluations, emphasizing early identification of issues via structured meetings and timelines to foster integrated, accountable assessments. Complementing this, the Center has expanded quantitative modeling for , , and evaluations, incorporating standards and submission tools to modernize pre-market reviews and post-market . CDER's Emerging Technology Program, ongoing since 2014, continues to facilitate approvals for novel manufacturing technologies by providing early regulatory feedback and site evaluations, addressing technical hurdles in producing high-quality drugs. In parallel, efforts to combat drug shortages have intensified, with CDER maintaining a dedicated for industry notifications and issuing annual reports documenting actions like import authorizations and production incentives; for instance, the report highlighted mitigation for 55 new shortages through interventions. Additionally, CDER has prioritized drug development via the Rare Disease Cures Accelerator, promoting standardized platforms and patient input to accelerate readiness. Looking ahead, CDER faces persistent challenges from escalating drug shortages, which affected 258 unique active ingredients between 2018 and 2023 due to manufacturing disruptions and fragility, necessitating enhanced and international coordination despite limited enforcement tools. The integration of and in and manufacturing poses validation and oversight hurdles, as CDER adopts a risk-based framework but grapples with ensuring model transparency and generalizability amid rapid technological evolution. Resource constraints, including workforce reductions, further complicate timely reviews of complex therapies like cell and products, while balancing accelerated approvals with robust post-market evidence remains critical to sustain innovation without compromising safety.

References

  1. [1]
    Center for Drug Evaluation and Research (CDER) - FDA
    Feb 29, 2024 · The Center for Drug Evaluation and Research (CDER) performs an essential public health task by making sure that safe and effective drugs are available.CDER Offices and Divisions · CDER Organization Chart · CDER Leadership Bios
  2. [2]
    A Brief History of the Center for Drug Evaluation and Research - FDA
    Jan 31, 2018 · The Center for Drug Evaluation and Research (CDER), which assures that safe and effective drugs are available to the American people, ...
  3. [3]
    FDA Center for Drug Evaluation and Research (CDER) - NCI SBIR
    Aug 29, 2022 · The Center for Drug Evaluation and Research (CDER) regulates over-the-counter and prescription drugs, including biological therapeutics and generic drugs.
  4. [4]
    CDER Initiatives - FDA
    Jul 3, 2024 · We actively pursue initiatives that modernize the drug review process, emphasizing efficiency and transparency, with the aim of expediting access to safe and ...Missing: notable achievements controversies
  5. [5]
    Drugs - FDA
    The Center for Drug Evaluation and Research (CDER) ensures that safe and effective drugs are available to improve the health of the people in the United States.Center for Drug Evaluation · Drug Approvals and Databases · Guidances
  6. [6]
    Summary - Challenges for the FDA - NCBI Bookshelf
    The FDA faces challenges including underfunding, organization issues, technical problems, unclear authority, and relies on industry fees, with a lack of ...Missing: notable achievements
  7. [7]
    Frequently Asked Questions about CDER - FDA
    Oct 28, 2019 · The Center oversees the research, development, manufacture and marketing of drugs. CDER ensures truth in advertising for prescription drugs and ...Missing: core | Show results with:core
  8. [8]
    [PDF] Center for Drug Evaluation and Research (CDER) Overview - FDA
    Feb 20, 2018 · is to protect and promote public health by helping to ensure that human drugs are safe and effective for their intended use, that they meet ...
  9. [9]
    Development & Approval Process | Drugs - FDA
    Aug 8, 2022 · FDA approval of a drug means that data on the drug's effects have been reviewed by CDER, and the drug is determined to provide benefits that outweigh its known ...How Drugs are Developed · Drug Approvals and Databases
  10. [10]
    The FDA's Drug Review Process: Ensuring Drugs Are Safe and ...
    Nov 24, 2017 · When an NDA comes in, the FDA has 60 days to decide whether to file it so that it can be reviewed. The FDA can refuse to file an application ...
  11. [11]
    New Drug Application (NDA) - FDA
    Jan 21, 2022 · The NDA application is the vehicle through which drug sponsors formally propose that the FDA approve a new pharmaceutical for sale and marketing in the US.
  12. [12]
    Office of New Drugs - FDA
    Oct 2, 2025 · OND's role is twofold: review applications and make an approval decision; set guidance and policy to ensure an efficient review process. OND ...
  13. [13]
    FDA's Drug Review Process: Continued
    Aug 24, 2015 · The FDA inspects the facilities where the drug will be manufactured as part of the approval process. FDA reviewers will approve the application ...
  14. [14]
    New Drug Development and Review Process | FDA
    Jul 31, 2023 · A team of CDER physicians, statisticians, chemists, pharmacologists, and other scientists reviews the company's data and proposed labeling. If ...
  15. [15]
    Expedited Programs for Serious Conditions | Drugs and Biologics
    Nov 28, 2023 · The four FDA programs are: fast track, breakthrough therapy, accelerated approval, and priority review designation.
  16. [16]
    Statement of Organization, Functions, and Delegations of Authority
    Dec 23, 2024 · The CDER, OMP, Office of Medical Policy Initiatives retitled the Division of Clinical Trial Quality to the Division of Clinical Innovations.
  17. [17]
    Office of Generic Drugs | Offices and Divisions - FDA
    Jul 23, 2025 · The IO provides oversight, leadership, strategic direction, and support for the Office of Generic Drugs (OGD) and its five sub-offices.Missing: key | Show results with:key<|separator|>
  18. [18]
    [PDF] CENTER FOR DRUG EVALUATION AND RESEARCH KEY ... - FDA
    Mar 1, 2025 · Division of Drug Quality and Compliance Services and Solutions (DDQCSS) ... Office of Drug Evaluation Sciences (ODES). Director. Jeffrey Siegel ...
  19. [19]
    CDER Office of Surveillance and Epidemiology - FDA
    Jul 26, 2024 · Office of the Center Director · Office of Communications · Office of Compliance · Office of Executive Programs · Office of Generic Drugs · Office of ...Missing: key | Show results with:key
  20. [20]
    CDER Offices and Divisions - FDA
    Dec 16, 2024 · Office of Cardiology, Hematology, Endocrinology and Nephrology (OCHEN) · Division of Cardiology and Nephrology (DCN) · Division of Diabetes, ...
  21. [21]
    Stanford faculty member George Tidmarsh, M.D., Ph.D ... - FDA
    Jul 21, 2025 · Stanford faculty member George Tidmarsh, M.D., Ph.D. named Director of Center for Drug Evaluation and Research. More Press Announcements.
  22. [22]
    CDER Leadership Bios | FDA
    Jul 23, 2025 · Dr. George Tidmarsh is the Director of the Center for Drug Evaluation and Research. The Center's mission is to ensure that safe, effective, and ...Missing: structure | Show results with:structure
  23. [23]
    Center for Drug Evaluation and Research Organization Chart - FDA
    Mar 20, 2025 · Organization chart for the FDA's Center for Drug Evaluation and Research, including leadership roles.Missing: key | Show results with:key
  24. [24]
    How Drugs are Developed and Approved - FDA
    Oct 24, 2022 · The mission of FDA's Center for Drug Evaluation and Research (CDER) is to ensure that drugs marketed in this country are safe and effective.Missing: functions | Show results with:functions
  25. [25]
    Promoting Safe & Effective Drugs for 100 Years - FDA
    Apr 23, 2019 · An overview of the history of FDA's regulation of drugs, originally published in FDA Consumer magazine.Missing: origins | Show results with:origins
  26. [26]
    Early Work of the Bureau of Chemistry Drug Lab - FDA
    Feb 1, 2018 · Explores the early history of the USDA Bureau of Chemistry's regulation of drugs.
  27. [27]
    Part I: The 1906 Food and Drugs Act and Its Enforcement | FDA
    Apr 24, 2019 · On 30 June 1906 President Roosevelt signed the Food and Drugs Act, known simply as the Wiley Act, a pillar of the Progressive era.
  28. [28]
    Milestones in US Food and Drug Law - FDA
    Jan 30, 2023 · Federal controls over the drug supply began with inspection of imported drugs in 1848, although the first federal biologics law, which addressed ...Missing: CDER | Show results with:CDER
  29. [29]
    Reform, Regulation, and Pharmaceuticals — The Kefauver–Harris ...
    in the form of “adequate and well-controlled investigations”— before ...
  30. [30]
    Trends in FDA drug approvals over last 2 decades - NIH
    Jan 28, 2020 · [2] In 1982, bureau of biologics was merged with it. In 1987, two different entities Center for Drug Evaluation and Research (CDER) and Centre ...Trends In Drug Approval In... · Table 4 · First Cycle Approval
  31. [31]
    [PDF] Prescription Drug User Fee Act of 1992 - FDA
    There are five major FDA components that support the PDUFA program: the Center for Drug Evaluation and Research (CDER), the Center for Biologics Evaluation and.<|separator|>
  32. [32]
    Speed, Safety, and Industry Funding — From PDUFA I to PDUFA VI
    Dec 7, 2017 · PDUFA began in 1992 with the laudable goal of clearing the substantial backlog of investigational drugs that had successfully completed clinical ...
  33. [33]
    effects of PDUFA on approval times and withdrawal rates - Nature
    Jul 1, 2005 · With PDUFA I and II, however, predicted approval times fell more rapidly, from about 25 months in 1992 to slightly less than 15 months in 2002, ...
  34. [34]
    Food and Drug Administration Modernization Act (FDAMA) of 1997
    Mar 29, 2018 · The Food and Drug Administration Modernization Act (FDAMA), enacted Nov. 21, 1997, amended the Federal Food, Drug, and Cosmetic Act relating to the regulation ...Missing: CDER | Show results with:CDER
  35. [35]
    The Prescription Drug User Fee Act: Much More Than User Fees - NIH
    PDUFA legislation has instituted a broad range of changes to FDA policy, including evidentiary standards for drug approval, accelerated pathways for approval.
  36. [36]
    PDUFA VII: Fiscal Years 2023 – 2027 - FDA
    Apr 24, 2023 · On September 30, 2022, the President signed into law the FDA User Fee Reauthorization Act of 2022.
  37. [37]
    Prescription Drug User Fee Act (PDUFA) Performance Reports - FDA
    Sep 9, 2025 · Since the implementation of PDUFA I in 1993, FDA has used PDUFA resources to significantly reduce the time it takes to evaluate new drugs ...
  38. [38]
    effects of PDUFA on approval times and withdrawal rates - PubMed
    Here, we review data on drug approvals and drug-approval times, both as a whole and by therapeutic class, which demonstrate that implementation of the PDUFAs ...Missing: impact | Show results with:impact
  39. [39]
    The History of FDA's Role in Preventing the Spread of HIV/AIDS | FDA
    Mar 14, 2019 · AZT (zidovudine)​​ In March of 1987, FDA approved zidovudine (AZT) as the first antiretroviral drug for the treatment of AIDS.
  40. [40]
    Drug Approval Package: Gleevec (Imatinib Mesylate) NDA #21-335
    Oct 17, 2001 · Gleevec (Imatinib Mesylate) Capsules. Company: Novartis Pharmaceuticals Corporation. Application No.: 21-335. Approval Date: 5/10/2001.
  41. [41]
    [PDF] Enhancement and Modernization of the FDA Drug Safety System
    PDUFA VII modernizes FDA drug safety by improving REMS assessments and optimizing the Sentinel Initiative, including enhancing its analytic capabilities.Missing: major reforms
  42. [42]
    Health-related Quality of Life Assessment after Antiretroviral Therapy
    The development and dissemination of antiretroviral (ARV) therapy (ART) has resulted in significant reductions in mortality associated with HIV infection and ...
  43. [43]
    [PDF] Statins: A Success Story Involving FDA, Academia and Industry
    reduction in the endpoints of heart disease, death or heart attack. Dr. Sobel noted, "Although the results of this study were quite modest by our present ...
  44. [44]
    3 Myths About Cholesterol-Lowering Statin Drugs
    Often the first line of therapy after lifestyle changes, statins can reduce the risk of stroke, heart attack and even death from cardiovascular disease by 25 ...Missing: approval | Show results with:approval
  45. [45]
    FDA use of Real-World Evidence in Regulatory Decision Making
    Sep 26, 2025 · Table 1. CDER drug approvals that used real-world evidence ; Nulibry® (Fosdenopterin). NDA 214018. Sentynl Therapeutics, Medical Records ; Orencia ...
  46. [46]
    [PDF] 2024 New Drug Therapy Approvals Annual Report - FDA
    In 2024, CDER approved 50 novel drugs, either as new molecular entities (NMEs) under. New Drug Applications (NDAs), or as new therapeutic biologics under ...
  47. [47]
    [PDF] Application Number: NDA 20-553/S-002 - accessdata.fda.gov
    A controlled-release form of oxycodone hydrochloride (OxyContin Controlled-Release Tablets, 10, 20 and 40 mg) was approved on 12/21/95. This submission seeks ...Missing: details | Show results with:details
  48. [48]
    [PDF] OxyContin Abuse and Diversion and Efforts to Address the Problem
    Dec 23, 2003 · OxyContin, a schedule II drug manufactured by Purdue Pharma L.P., was approved by FDA in 1995 for the treatment of moderate-to-severe pain.
  49. [49]
    The Promotion and Marketing of OxyContin: Commercial Triumph ...
    When OxyContin entered the market in 1996, the FDA approved its original label, which stated that iatrogenic addiction was “very rare” if opioids were ...Missing: CDER | Show results with:CDER
  50. [50]
    How FDA Failures Contributed to the Opioid Crisis | Journal of Ethics
    ... approved Purdue Pharma's new drug application for extended-release (ER) oxycodone in 1995. Had it done so, ER oxycodone's label would have had a narrow ...Missing: CDER | Show results with:CDER
  51. [51]
    Opioid Approval and Monitoring by the U.S. Food and Drug ... - NCBI
    In the 1960s and 1970s, the FDA approved short-acting combination products such as oxycodone/acetaminophen (Percocet, 1976). In the late 1980s and early 1990s, ...
  52. [52]
    Overdose Prevention Activities Timeline - FDA
    1995. OxyContin (oxycodone controlled-release) approved; first formulation of oxycodone that allowed dosing every 12 hours instead of every 4 to 6 hours. 1998.
  53. [53]
    [PDF] External Review of FDA Regulation of Opioid Analgesics Final Report
    Jan 25, 2023 · Addressing the drug overdose crisis, and its varied root causes, while also ensuring that prescription opioids are available for the evidence- ...Missing: criticisms | Show results with:criticisms
  54. [54]
    Senator Hassan Leads Push for an Investigation into McKinsey's ...
    May 4, 2022 · The firm was deeply involved with the Center for Drug Evaluation and Research (CDER), FDA's principal center for approving new drugs, including ...Missing: criticisms | Show results with:criticisms
  55. [55]
    [PDF] The Firm and the FDA: McKinsey & Company's Conflicts of Interest ...
    Apr 13, 2022 · CDER approves new drugs, including prescription opioids, ... 237 See McKinsey Settles for Nearly $600 Million over Role in Opioid Crisis, New York ...
  56. [56]
    Purdue Pharma L.P.; Withdrawal of Approval of a New Drug ...
    Aug 7, 2013 · The Food and Drug Administration (FDA) is withdrawing approval of a new drug application (NDA) for OXYCONTIN (oxycodone hydrochloride) Extended-Release Tablets.Missing: CDER | Show results with:CDER<|separator|>
  57. [57]
    Unlocking the Revolving Door: How FDA-Firm Relationships Affect ...
    Dec 20, 2023 · I find that when pharmaceutical companies hire former FDA employees, the rate of drug approvals increases which in turn raises firm value.
  58. [58]
    FDA's revolving door: Companies often hire agency staffers who ...
    Jul 5, 2018 · In 2012 and 2013, data expert Joan Buenconsejo led FDA's analysis of medical statistics in drug reviews, including offerings from AstraZeneca.
  59. [59]
    FDA Job Cuts, Key Departures Bring 'Revolving Door' Debate Front ...
    Apr 2, 2025 · In 2018, Science found that 11 of 16 ex-FDA medical examiners who worked on 28 drug approvals were currently employed by or consulting for ...<|separator|>
  60. [60]
    Evaluating PDUFA: Paying for FDA Drug-Application Reviewers by ...
    Mar 7, 2025 · PDUFA has allowed the FDA to hire more drug-application reviewers by charging user fees to drug manufacturers. User fees now account for almost half of FDA ...
  61. [61]
    History, status, and politicization of the FDA - ScienceDirect
    In 1992, Congress passed the Prescription Drug User Fee Act (PDUFA), which granted the FDA the right to collect fees directly from drug manufacturers to fund ...
  62. [62]
    Drug-Review Deadlines and Safety Problems
    Mar 27, 2008 · PDUFA deadlines have appreciably changed the approval decisions of the FDA. Once medications are in clinical use, the discovery of safety ...
  63. [63]
    [PDF] PDUFA REAUTHORIZATION PERFORMANCE GOALS AND ... - FDA
    This document contains the performance goals and procedures for the Prescription Drug User. Fee Act (PDUFA) reauthorization for fiscal years (FYs) 2023-2027 ...<|separator|>
  64. [64]
    Despite record federal lobbying spending, the pharmaceutical and ...
    Feb 2, 2023 · Pharmaceutical and health product companies poured over $372 million into federal lobbying last year, outspending every other industry.
  65. [65]
    Two faces of corporate lobbying: Evidence from the pharmaceutical ...
    We document that increase in lobbying expenditures leads to more number of FDA approvals.
  66. [66]
  67. [67]
    The effects of lobbying on the FDA's recall classification
    Jun 20, 2023 · This study assesses whether the FDA's product recall classifications in recall scenarios are influenced by firms' lobbying activities.
  68. [68]
    Emergency Use Authorization of Medical Products - FDA
    Sep 12, 2022 · This guidance explains FDA's general recommendations and procedures applicable to the authorization of the emergency use of certain medical products.
  69. [69]
    [PDF] Emergency Use Authorization (EUA) for remdesivir, an unapproved ...
    May 1, 2020 · Remdesivir received EUA for treatment of severe COVID-19 in adults and children, due to a public health emergency and lack of alternatives.Missing: criticisms | Show results with:criticisms
  70. [70]
    FDA's approval of Veklury (remdesivir) for the treatment of COVID-19
    Oct 22, 2020 · CDER's detailed review of Veklury clinical trials that supported FDA approval this week reflects our independent clinical evaluation of the ...Missing: criticisms | Show results with:criticisms
  71. [71]
    Identification of Bradycardia Following Remdesivir Administration
    Feb 14, 2023 · This cohort study examines the incidence of bradycardia occurring during or after administration of remdesivir in patients with COVID-19 ...Missing: criticisms | Show results with:criticisms
  72. [72]
    Ethics of Emergency Use Authorization During the Pandemic
    Oct 30, 2020 · FDA revoked this authorization on June 15, after well-controlled randomized trials demonstrated no benefit from the drugs and reports of serious ...
  73. [73]
    From 9/11 to COVID-19: A Brief History of FDA Emergency Use ...
    Jan 28, 2021 · Trump notoriously pressured FDA officials into authorizing chloroquine and hydroxychloroquine, anti-malarial drugs that many believed might pose ...Missing: CDER | Show results with:CDER
  74. [74]
    Enhancing Postmarket Safety Monitoring - Challenges for the FDA
    While this passive surveillance system may be capable of detecting rare serious adverse events, it has several limitations, including profound underreporting, ...
  75. [75]
    An Evaluation of “Drug Ineffective” Postmarketing Reports in Drug ...
    "Drug ineffective" (DI) is the most reported adverse event in FAERS, often by consumers, but only a minority are useful for pharmacovigilance.
  76. [76]
    Vioxx (rofecoxib) Questions and Answers - FDA
    Apr 6, 2016 · Merck's decision to withdraw Vioxx from the market is based on new data from a trial called the APPROVe [ Adenomatous Polyp Prevention on VIOXX] ...Missing: CDER | Show results with:CDER
  77. [77]
    Rofecoxib (Vioxx) voluntarily withdrawn from market - PMC - NIH
    Oct 26, 2004 · Merck & Co. announced Sept. 30 a voluntary worldwide withdrawal of rofecoxib (Vioxx) after a study showed patients taking the drug on a long- ...
  78. [78]
    FDA and Vioxx withdrawal - Medscape
    Nov 23, 2004 · The FDA's Office of New Drugs was charged with missing early warnings about Vioxx cardiovascular risks, discounting bad news even from its ...
  79. [79]
    GAO-06-402, Drug Safety: Improvement Needed in FDA's ...
    An important part of the drug approval and postmarket monitoring process is the advice FDA receives from 16 human-drug-related scientific advisory ...
  80. [80]
    The Flawed Basis for FDA Post-Marketing Safety Decisions - Nature
    Dec 14, 2005 · The failure of the FDA's post-marketing surveillance system is reviewed. The data necessary for objective evaluations of possible post-marketing ...
  81. [81]
    Postmarketing Surveillance Programs - FDA
    Apr 2, 2020 · FDA maintains a system of postmarketing surveillance and risk assessment programs to identify adverse events that did not appear during the drug approval ...
  82. [82]
    [PDF] Approval Costs and Innovation in Medical Technologies
    Sep 29, 2023 · How does FDA regulation impact innovation, market structure, and product safety in es- tablished product categories?
  83. [83]
    Innovation under Regulatory Uncertainty: Evidence from Medical ...
    This paper explores how the regulatory approval process affects innovation incentives in medical technologies.
  84. [84]
    [PDF] Impacts of First-in-Class Drug Approvals on Future in-Class Innovation
    We analyze the potential impacts of FDA approval decisions on future drug development by first examining the economic theory of medical innovation and ...
  85. [85]
    PDUFA: Still a weird word worth knowing - PhRMA
    In the last 20 years, PDUFA has provided patients with timelier access to more than 1,500 new drugs and biologics and decreased review times for these ...Missing: effects | Show results with:effects
  86. [86]
    The Prescription Drug User Fee Act: Good, Bad, or a Lot of Both?
    Mar 11, 2025 · PDUFA has reduced drug approval times and increased application rates, enhancing access to new therapies. Concerns exist about conflicts of ...
  87. [87]
    Timely, Cheap, or Risk-Free? The Effect of Regulation on the Price ...
    Mar 18, 2024 · In this paper, we review and discuss the impact of different pricing rules on the expenditure and availability of new drugs.
  88. [88]
    Impact of Expanded Access on FDA Regulatory Action and Product ...
    The purpose of this study is to address concerns that expanded access may negatively impact the ultimate regulatory action and product labeling for new drugs.
  89. [89]
    Quantifying 21st Century Opportunity Costs of FDA Regulation | NBER
    Dec 16, 2021 · This paper revisits Peltzman (1973) in light of two recent opportunities to quantitatively assess tradeoffs in drug regulation.
  90. [90]
    FDA establishes CDER Center for Clinical Trial Innovation (C3TI)
    Apr 15, 2024 · C3TI will be a central hub within CDER that supports innovative approaches to clinical trials that are designed to improve the efficiency of drug development.
  91. [91]
    Emerging Technology Program (ETP) - FDA
    Feb 24, 2025 · The Emerging Technology Program - or ETP - at FDA helps industry earn regulatory approval for drug applications with innovative technologies and ...How to Participate in ETP · ETP News · Lifecycle of an ETP Technology
  92. [92]
    [PDF] Drug Shortages | FDA
    This annual report to Congress summarizes the major actions taken by the U.S. Food and Drug Administration (FDA or Agency) during calendar year (CY) 2023 to ...Missing: initiatives | Show results with:initiatives
  93. [93]
    [PDF] Analysis of Drug Shortages, 2018-2023 - https: // aspe . hhs . gov.
    Jan 8, 2025 · Between 2018 and 2023, a total of 258 unique active ingredients (i.e., molecules) went into national shortage per the FDA's report to ...Missing: initiatives | Show results with:initiatives
  94. [94]
    Artificial Intelligence for Drug Development - FDA
    Feb 20, 2025 · AI will undoubtedly play a critical role in the drug development life cycle and CDER plans to continue developing and adopting a risk-based ...
  95. [95]
    FDA Regulatory Uncertainty Calls for Strategic Decision-Making by ...
    Apr 4, 2025 · The recent reductions in the US Food and Drug Administration's workforce are poised to have a substantial impact on drug and biologic ...