Fact-checked by Grok 2 weeks ago

PDUFA date

The PDUFA date refers to the target deadline established under the (PDUFA) for the U.S. (FDA) to complete its substantive review of a (NDA) or biologics license application (BLA). Enacted by in 1992 amid concerns over prolonged drug review backlogs exacerbated by the AIDS epidemic and under-resourced FDA staffing, PDUFA enables the agency to collect user fees from pharmaceutical manufacturers to fund additional personnel and processes for expedited evaluations, without compromising statutory standards for and . Under PDUFA performance goals, the FDA commits to reviewing 90% of new molecular NDAs or BLAs within 10 months of filing and 90% of applications—reserved for drugs addressing unmet medical needs or serious conditions—within 6 months. These timelines represent a marked reduction from pre-PDUFA eras, when median review times often exceeded 30 months, enabling faster patient access to therapies while maintaining post-approval monitoring mechanisms. Reauthorized every five years through negotiations with industry and stakeholders, the program has evolved to incorporate commitments for enhanced communication, such as formal meetings between sponsors and reviewers, and has funded over half of the FDA's human drug review budget in recent cycles. PDUFA dates hold particular significance in the pharmaceutical sector, serving as milestones that influence investment decisions and market expectations for potential approvals, though the FDA retains flexibility to issue complete response letters or extensions if additional data is required, with historical rates exceeding goals in most reauthorization periods. Critics have raised questions about whether fee-driven incentives might prioritize speed over , yet empirical analyses indicate no substantial decline in post-market signals attributable to accelerated reviews, underscoring PDUFA's role in fostering amid resource constraints. The program's ongoing refinements, including PDUFA VII (fiscal years 2023–2027), emphasize patient-focused and to address evolving public health priorities.

Definition and Purpose

Core Concept and Objectives

The PDUFA date designates the target deadline for the (FDA) to complete its substantive review of a () or biologics license application (), following acceptance for filing. Enacted under the () in 1992, this statutory benchmark is generally 10 months from the filing date for standard reviews of non- applications and 6 months for reviews, which apply to therapies offering significant improvements in treating serious conditions. PDUFA enables the FDA to levy user fees on pharmaceutical sponsors submitting these applications, generating revenue—approximately 65% of the agency's human drugs program budget—to hire additional scientific reviewers and support staff, thereby addressing chronic resource constraints that previously protracted evaluations. The core objectives of PDUFA center on accelerating drug approvals to enhance timely access to safe and effective treatments, without compromising the agency's mandate for thorough safety and efficacy assessments. Prior to 1992, FDA review times averaged around 30 months, contributing to backlogs that delayed therapies for life-threatening diseases and underscored a causal imbalance where prolonged waits increased morbidity and mortality from untreated or undertreated conditions far beyond the infrequent risks of expedited approvals. By funding performance-driven processes, PDUFA aims to optimize this trade-off, prioritizing empirical outcomes such as reduced overall patient harm through earlier interventions over marginal gains from extended pre-market scrutiny of rare adverse events. PDUFA dates function as aspirational yet enforceable goals, not rigid guarantees, allowing flexibility for complex applications while committing the FDA to structured timelines. Historical data indicate the agency has met these goals in approximately 90% of cases since PDUFA's , as documented in reports, reflecting sustained improvements in efficiency.

User Fee Structure and Funding

Under PDUFA VII, the fee structure consists of application fees and annual program fees, replacing earlier categories like establishment fees from prior iterations. Application fees apply to new drug applications (NDAs) and biologics license applications (BLAs), set at $4,310,002 for submissions requiring clinical data and $2,155,001 for those not requiring such data in (FY) 2025. Program fees, charged per eligible approved product as of October 1 each year, amount to $403,889 annually in FY2025. These fees generate a target revenue of approximately $1.48 billion for FY2025, with program fees comprising the majority (about 80%) and application fees the remainder. The collected fees fund roughly 65% of the FDA's human drugs program budget, supplementing congressional appropriations that constitute the minimum required non-fee funding level of $287 million for FY2025. This allocation supports hiring specialized staff (including 352 new positions over the five-year reauthorization), upgrades, and operational enhancements for pre-market processes, reducing dependence on variable taxpayer appropriations. By linking fee revenue to statutory goals—such as timeliness and resource commitments—the imposes on the FDA, incentivizing efficient resource use while directing funds exclusively to human drug activities as mandated by law. This -based funding mechanism aligns payments with the costs of expedited reviews, fostering incentives for both efficiency and prioritization of high-impact submissions. Empirical analyses of post-PDUFA approvals show no significant increase in risks, as evidenced by stable rates of market withdrawals for serious adverse events, with residual concerns addressed through enhanced post-market surveillance provisions introduced in PDUFA IV and maintained thereafter.

Historical Development

Initial Enactment (PDUFA I, 1992)

The (PDUFA I) was signed into law on October 29, , as Title I of the Omnibus Budget Reconciliation Act of , amid intense advocacy from AIDS patient groups and pharmaceutical stakeholders confronting FDA review delays that averaged 26 to 31 months for new drug applications in the early 1990s. These delays exacerbated the crisis by postponing access to emerging therapies, prompting congressional action to fund expanded FDA capacity without relying solely on taxpayer appropriations. PDUFA I authorized the FDA to collect application, establishment, and product fees from to support hiring additional reviewers and clearing the existing of applications within one year of enactment. The set specific performance targets, mandating that the agency review and act on 60% of new drug and biologics applications within 10 months during fiscal year 1993, with the threshold rising annually to 90% by fiscal year 1997; standard reviews were targeted at 12 months, though the emphasis on cases addressed urgent unmet needs. These goals were enforceable through , including requirements for annual performance reports to . Implementation of PDUFA I yielded an immediate causal reduction in durations, with times dropping from over 26 months pre-enactment to approximately 13 months by the mid-1990s, enabling faster market entry for therapies while maintaining statutory safety standards grounded in . This shift underscored a policy pivot toward balancing regulatory caution with evidence-based prioritization of patient access to innovations, particularly for life-threatening conditions like .

Key Reauthorizations and Evolutions (1997–2022)

PDUFA II, enacted on November 21, 1997, through the FDA Modernization Act, extended the user fee program for five years while introducing incentives for , including a six-month extension of market exclusivity for manufacturers conducting requested pediatric studies requested by the FDA. This provision addressed the historical understudied status of pediatric applications, which comprised less than 10% of FDA submissions prior to incentives. Additionally, PDUFA II mandated the establishment of scientific advisory committees to provide independent expert input on complex , enhancing without altering core performance goals of 90% attainment for and review timelines. User fees were adjusted upward to account for inflation and rising application volumes, maintaining FDA's operational funding stability over appropriations-dependent models. PDUFA III, reauthorized in October 2002 via the Public Health Security and Bioterrorism Preparedness and Response Act, sustained the pediatric exclusivity incentives and extended commitments to advisory committee utilization, while setting refined performance goals such as 90% review of standard new drug applications within 10 months and priority ones within six months. It incorporated extensions of exclusivity periods tied to supplemental applications, supporting post-approval modifications amid growing workload from biologics and complex submissions. Fee structures continued inflation-linked adjustments, with total revenues calibrated to workload increases, achieving consistent goal attainment rates around 90% and preserving the user-fee mechanism's responsiveness over variable ional funding. Following the 2004 Vioxx withdrawal, which highlighted post-market safety gaps, PDUFA IV under the FDA Amendments Act of September 2007 expanded user fees by approximately 11% initially (with subsequent adjustments) to fund enhanced safety surveillance, including mandatory Risk Evaluation and Mitigation Strategies (REMS) for drugs with significant risks like opioids. This reauthorization broadened fee-eligible activities to post-approval monitoring and , responding empirically to safety signals without diluting review speed goals, which held at 90% compliance. Fees were tied to and workload formulas, ensuring sustained agility in review processes compared to appropriation delays. PDUFA V, via the FDA Safety and Innovation Act of July 2012, formalized the Breakthrough Therapy designation for drugs showing substantial improvement over existing therapies in serious conditions, enabling intensive FDA guidance and rolling reviews to accelerate access based on preliminary clinical evidence. It maintained 90% performance targets while adjusting fees for inflation and expanded submissions, including priorities, reinforcing the user-fee model's data-driven evolution for innovation without safety trade-offs. PDUFA VI, enacted in August 2017 through the FDA Reauthorization Act, heightened focus on complex manufacturing reviews and real-time communication for chemistry, manufacturing, and controls () issues, alongside enhanced post-market surveillance to address evolving risks like those in the opioid context via REMS refinements. Performance goals remained at 90% for timelines, with fees escalated per inflation and workload metrics—reaching $879 million in base revenue for FY2018—to support these increments, empirically validating the program's superiority for timely, evidence-based adjustments over uncertainties.

FDA Review Process and PDUFA Goals

Application Submission and Date Assignment

Drug sponsors submit New Drug Applications (NDAs) or Biologics License Applications (BLAs) to the U.S. Food and Drug Administration (FDA) electronically using the (eCTD) format, which standardizes the structure for regulatory submissions. Upon receipt by the FDA's Center for Drug Evaluation and Research (CDER) or Center for Biologics Evaluation and Research (CBER), the agency conducts an initial 60-day review to determine if the application contains sufficient information for substantive evaluation. If deemed acceptable for filing, the FDA classifies the submission as either standard or based on predefined criteria and assigns a PDUFA goal date accordingly: 10 months from the filing date for standard reviews and 6 months for priority reviews, with these timelines applying specifically to new molecular entity NDAs and original BLAs. Priority review designation is granted when the proposed or biologic, if approved, would offer a significant improvement in safety or effectiveness for treating, preventing, or diagnosing a serious relative to existing , or when no adequate exists. The FDA communicates this classification to the sponsor within 60 days of the original submission receipt. According to FDA records, approximately 56% of approvals in 2024 received status, reflecting the agency's targeted application of expedited timelines to address high unmet medical needs. During the review, submission of a major amendment—such as substantial new —triggers a review clock stop, after which the FDA may extend the PDUFA goal date by up to three months to evaluate the added material. This extension mechanism ensures thorough assessment without compromising the overall performance commitments. The PDUFA date thus serves as a mutually recognized target, fostering alignment between sponsors and the FDA while mitigating the unpredictability inherent in pre-PDUFA indefinite review durations.

Performance Goals and Timelines

The (PDUFA) sets specific performance goals for the (FDA) to review and act on new drug applications (NDAs) and biologics license applications (BLAs), targeting 90% of submissions for timely completion. These goals encompass initial filing decisions, mid-review communications, and final actions, such as approval or issuance of a Complete Response Letter (CRL) detailing unresolved deficiencies. For original applications, the FDA commits to assessing completeness within 60 days of receipt; if deemed sufficiently complete, the application is filed, and the review clock initiates from that filing date for new molecular entity () submissions, or from receipt for non-NME types. Incomplete submissions may result in a Refuse-to-File determination, halting substantive review until resubmission. Standard review goals require the FDA to and act on 90% of original NDAs and BLAs within 10 months of the 60-day filing date, while reviews—reserved for drugs offering significant improvements in or —target completion within 6 months of the same filing date. Non- original NDAs follow analogous timelines measured from : 10 months for standard and 6 months for . During the , the FDA conducts an internal mid-cycle assessment, followed by communication to the within two weeks of the meeting, outlining review status, identified issues, and anticipated milestones to facilitate resolution of potential deficiencies. Final actions prioritize comprehensive data evaluation, with extensions possible for major amendments—adding up to 3 months for originals or 2 months for supplements—ensuring reviews address evolving evidence without undue prolongation. Adaptations for complex or expedited applications shorten effective timelines; fast-track, , or accelerated approval designations prompt the FDA to aim for actions at least one month earlier than standard goals, emphasizing iterative sponsor interactions and targeted data requirements. Under PDUFA VII (fiscal years 2023–2027), these goals integrate efficiency measures, including the Split Real-Time Application Review (STAR) pilot for modular efficacy supplement submissions and expanded use of technologies alongside to streamline assessments without compromising rigor. Such provisions enforce structured discipline in the review process while allowing flexibility for intricate cases, directing resources toward substantive scientific evaluation over mechanical adherence to deadlines.

Empirical Impacts and Benefits

Acceleration of Drug Review Times

Prior to the enactment of PDUFA in , the median FDA review time for new drug applications averaged 29 months in the late . Implementation of PDUFA I markedly accelerated this process, with median review times declining to approximately 12.9 months by the PDUFA era and further stabilizing at 10–12 months in subsequent decades. Empirical analyses, including those from the , quantify the impact of PDUFA I and II as accelerating the pre-existing downward trend in review times, achieving annual reductions of 6–7% during PDUFA I and 3–4% during PDUFA II, cumulatively shortening per-drug approvals by 1–2 years relative to counterfactual projections. PDUFA performance goals formalized these gains, establishing targets of 10 months for new drug and biologics license applications and 6 months for reviews—effectively halving timelines from pre-PDUFA benchmarks and enabling expedited responses to needs. These reductions have facilitated the timelier approval of thousands of new therapeutics since , with FDA data indicating sustained median review durations below 12 months through 2022 despite rising application volumes. Shorter review periods under PDUFA minimize the temporal opportunity costs borne by patients forgoing effective treatments, as faster approvals equate to reduced untreated progression without evidence of elevated post-market withdrawal rates per unit of review time.

Enhanced Patient Access and Innovation

The enactment of PDUFA has facilitated the annual approval of approximately 40-50 novel drugs by the FDA in recent years, enabling earlier therapeutic availability compared to the pre-PDUFA era when approvals averaged around 23 per year. From 2013 to 2022, the FDA averaged 43 novel drug approvals annually, many of which addressed unmet medical needs in areas such as and rare diseases. This acceleration has positioned the as the global leader in approving new molecular entities, outpacing other regulatory agencies and fostering broader patient access to innovative treatments. PDUFA's performance goals have directly supported timely entry of therapies that improve health outcomes, such as antiretrovirals for , where expedited reviews in the 1990s aligned with the shift to combination therapies that reduced U.S. AIDS mortality by over 70% from peak levels in the mid-1990s. Similarly, cardiovascular agents like statins, benefiting from streamlined processes post-PDUFA, contributed to substantial declines in heart disease mortality through widespread adoption following approvals in the late 1980s and 1990s. These examples illustrate how PDUFA bridges the gap between clinical development and market availability, allowing patients to access therapies years earlier than under prior resource-constrained FDA operations. User fees under PDUFA have funded specialized review capacity for advanced modalities, including biologics and therapies, enabling the FDA to maintain expertise amid rapid scientific progress. This infrastructure supports high-quality submissions by incentivizing sponsors to provide robust data, rather than lowering standards, and has been deemed essential for sustaining U.S. leadership. Analyses indicate that PDUFA counters overly cautious regulation by promoting market-driven innovation, with fees comprising over 65% of the FDA's review budget and allowing scalable hiring of reviewers. Empirical cost-benefit evaluations, such as those from the , quantify PDUFA's net positive impact, estimating that faster approvals saved the equivalent of 180,000 to 310,000 life-years through timely access to effective drugs, far exceeding any bounded risks from accelerated processes. These gains stem from reduced delays in therapy deployment, underscoring PDUFA's role in prioritizing evidence-based access over indefinite pre-market perfectionism.

Criticisms, Controversies, and Evidence-Based Rebuttals

Claims of Rushed Approvals and Safety Risks

Critics have argued that PDUFA-imposed deadlines pressure the FDA to approve drugs prematurely, increasing post-market safety issues. A study in the New England Journal of Medicine analyzed 188 new molecular entities approved from 1992 to 2005 and found that drugs approved immediately before PDUFA deadlines—defined as within two months—were associated with higher rates of safety problems, including withdrawals and black-box warnings; specifically, 7 of 11 safety-based withdrawals occurred for such "deadline drugs." This study suggested that end-of-review-period approvals, potentially rushed to meet timelines, correlated with a 3.9-fold higher likelihood of post-approval safety events compared to earlier approvals in the cycle. Advocates for stricter safety standards, including some groups, have cited this as evidence of PDUFA prioritizing speed over thoroughness, with examples like the 2004 withdrawal of (Vioxx) for cardiovascular risks allegedly linked to accelerated review pressures. Further analyses have claimed broader trends of elevated risks under faster reviews. A 2014 examination of 748 new molecular entities approved from 1975 to 2009 reported that post-1992 PDUFA-era drugs were more likely to receive black-box warnings (15% overall rate) or be withdrawn (4% rate), attributing half of withdrawals to the accelerated period and speculating that reduced review times compromised pre-approval scrutiny. Some researchers have quantified potential adverse event increases, estimating that each 10-month shortening of review time could raise serious post-approval reactions by up to 18%, based on patterns in expedited approvals. These critiques often emphasize that while pre-PDUFA withdrawals were rare, the post-PDUFA landscape saw clusters like cerivastatin (withdrawn 2001) and troglitazone (2000), implying deadlines incentivize overlooking marginal risks. Empirical rebuttals, however, indicate no statistically significant rise in overall safety failures attributable to PDUFA. A analysis of withdrawal rates showed an increase from 3.10% in the 8 years pre-PDUFA to 3.47% post-enactment, deemed non-substantive and consistent with historical variability rather than deadline-driven rushing. Peer-reviewed assessments confirm that proportions and timings of withdrawals for drugs approved before and after PDUFA do not differ meaningfully, with overall rates remaining at 1-2% across eras, undermining claims of systemic safety erosion. Subsequent PDUFA iterations have incorporated enhanced post-market , such as PDUFA IV's (2008-2012) expansion of user fees to support monitoring, enabling faster detection and mitigation of issues without evidence of heightened baseline risks from accelerated reviews. Causal evaluations further highlight opportunity costs of delays: the FDA's 1970s moratorium on beta-blockers—delayed until 1981 over animal carcinogenicity concerns—likely caused tens of thousands of preventable deaths from unaddressed secondary heart attack risks, illustrating how pre-PDUFA caution exacted greater mortality tolls than rare post-approval withdrawals. PDUFA goals include review extensions for complex cases, as affirmed by independent audits, countering narratives of inflexible rushing while metrics prioritize verifiable outcomes over anecdotal deadline associations.

Concerns Over Industry Influence and Fee Dependency

Critics of PDUFA have raised concerns that the program's reliance on industry-paid user fees, which constitute approximately 66% of the FDA's human drugs program budget as of 2022, creates incentives for and biases toward approving applications to sustain fee revenue. Organizations such as the have highlighted hypothetical risks of industry influence, positing that fee dependency could pressure the FDA to prioritize speed over scrutiny, potentially mirroring dynamics in other fee-funded agencies where payers gain leverage. Some left-leaning advocacy groups frame PDUFA as a to pharmaceutical interests, emphasizing the transfer of over $1.3 billion in fees annually from companies to the agency while downplaying empirical gains in patient access. These critiques often invoke a precautionary stance against market-like funding mechanisms, arguing they erode public oversight in favor of private priorities. Empirical data, however, reveals no causal link between user fees and diminished approval standards, with FDA rejection or complete response rates for new drug applications remaining stable at around 10-15% across PDUFA eras, unaffected by funding shifts. Studies analyzing pre- and post-PDUFA approval decisions find that while review timelines shortened, approval thresholds did not laxen, as evidenced by consistent evidentiary requirements and post-market enhancements mandated in reauthorizations. dependency, rather than fostering bias, enables operational agility by insulating drug review staffing from congressional appropriation delays, allowing the FDA to maintain a self-sustaining of approximately 4,000 reviewers without equivalent vulnerabilities in tax-funded models. Safeguards within PDUFA further mitigate influence risks, including mandatory performance goals tied to independent advisory committee reviews, where external experts vote on applications under public transparency protocols, and explicit prohibitions on using fees for enforcement actions against fee payers. These structures promote akin to competitive incentives, countering inaction biases inherent in overly precautionary, budget-constrained systems that historically delayed approvals by years and denied access to therapies later proven beneficial. Overall, the fee model aligns regulator incentives with efficient , outperforming alternatives prone to fiscal politicization, as substantiated by sustained innovation outputs without correlated safety erosions.

Recent Developments and Future Outlook

PDUFA VII Implementation (2023–2027)

PDUFA VII, enacted on December 23, 2022, as part of the , authorizes the FDA to collect user fees totaling a base revenue of $1.15 billion for 2023, with annual adjustments for and other factors through 2027, enabling sustained funding for drug review processes. The program emphasizes enhancements such as standardized electronic submissions to streamline reviews, integration of technologies in clinical trials, and targeted support for complex products including cell and gene therapies. For s, it includes commitments like reviewer training on drug development and a pilot program for advancing endpoints in applications, building on prior incentives under the Orphan Drug Act without introducing new fee waivers but facilitating faster evaluation pathways. Fee structures under PDUFA VII have seen progressive increases to match revenue targets; for 2025, rates were finalized and published on July 31, 2024, with the full application fee for submissions set at approximately $4.3 million, reflecting an adjustment from prior years to account for operational costs and . has focused on performance goals outlined in the PDUFA VII agreement, including improved manufacturing quality assessments through enhanced inspection processes and greater incorporation of in regulatory decisions, as detailed in FDA's quarterly performance reports showing completion of key deliverables such as guidance documents on these topics. FDA has achieved approximately 90-95% with timelines in early s of PDUFA VII, per tracking , despite workforce challenges including targeted hiring goals for specialized reviewers (e.g., 132 for biologics in FY2023) and subsequent disruptions from 2025 layoffs affecting several hundred probationary staff, which were partially reversed through rehiring and contractor supplementation to maintain capacity. These efforts have preserved empirical balances in approval speeds and post-market safety monitoring, with no significant deviations from historical safety metrics reported in FDA analyses up to 2025.

Reauthorization Process for PDUFA VIII

The reauthorization of PDUFA VIII, covering fiscal years 2028 through 2032, commenced with a public meeting held by the FDA on July 14, 2025, to initiate discussions on program enhancements, funding needs, and performance goals. This step aligns with the statutory process, where the FDA engages stakeholders including pharmaceutical industry representatives, patient advocacy groups, and consumer organizations through consultations and written comments to develop recommendations. Negotiations focus on balancing review efficiency commitments with revenue projections from user fees, which historically adjust upward by amounts tied to inflation, workload increases, and new initiatives; for instance, PDUFA fees have escalated from an initial $36 million in the program's early years to billions annually by PDUFA VII to sustain FDA's review capacity. The FDA is required to transmit its negotiated commitment letter to Congress by January 15, 2027, outlining proposed fees and goals, followed by legislative approval before the PDUFA VII expiration on September 30, 2027, to avoid disruptions in fee collections that fund approximately 65% of the Center for Drug Evaluation and Research's budget. Key debates in the PDUFA VIII process include proposals to reduce user fees, as suggested by FDA Commissioner in July 2025, who advocated lowering application and program fees—such as the FY 2025 application fee of $4.3 million and annual program fee exceeding $400,000—to alleviate industry burdens amid rising development costs. Critics, including experts from Yale and industry groups, contend that such reductions risk underfunding the agency, potentially reverting to pre-PDUFA eras of prolonged times exceeding two years for applications, without evidence that funding models would maintain the empirical gains in approval speed and volume seen since 1992. Stakeholder input, such as from the American Society of Gene & Cell Therapy, emphasizes enhancements like bolstering first-cycle rates and resource allocation for emerging therapies, rather than fee cuts that could impair staffing and expertise amid biotech innovations in gene editing and . Failure to reauthorize PDUFA VIII promptly would halt new fee collections post-September 2027, compelling reliance on inconsistent appropriations and likely extending review timelines, as evidenced by historical bottlenecks before user fees enabled consistent funding for over 10,000 annual submissions. Empirical data from prior cycles supports continuity of the core framework, with PDUFA-linked reviews achieving median times under 10 months for drugs without commensurate rises in post-market withdrawals, underscoring the need to prioritize evidence-based adjustments over unproven reforms in sustaining pipelines. The process thus hinges on forging commitments that preserve fiscal stability to address advancing therapeutic modalities, countering proposals lacking demonstrated superiority to the user fee model's track record of accelerating access while upholding rigorous evaluation standards.

References

  1. [1]
    PDUFA Legislation and Background - FDA
    Dec 16, 2015 · The Prescription Drug User Fee Act (PDUFA) was created by Congress in 1992 and authorizes FDA to collect fees from companies that produce certain human drug ...
  2. [2]
    What is PDUFA?And what is a PDUFA date? - Patsnap Synapse
    Jan 9, 2024 · The PDUFA date is a specific target date by which the FDA aims to complete its review of a new drug application (NDA) or a biologics license application (BLA).
  3. [3]
    PDUFA - PhRMA
    The PDUFA user fee program was first created in response to a bottleneck of new medicine approvals that left patients waiting for years for an under-staffed and ...
  4. [4]
    [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 ...
  5. [5]
    Priority Review - FDA
    Jan 4, 2018 · A Priority Review designation means FDA's goal is to take action on an application within 6 months (compared to 10 months under standard review).
  6. [6]
    The Prescription Drug User Fee Act: Cause for Concern? - PMC - NIH
    Feb 12, 2018 · The Prescription Drug User Fee Act (PDUFA) was originally enacted into law in 1992. PDUFA provides the Food and Drug Administration (FDA) ...
  7. [7]
    Understanding the FDA Approval Process and PDUFA Dates
    Mar 20, 2025 · The Prescription Drug User Fee Act (PDUFA) was created by Congress in 1992 and collects user fees on human drug applications for review, ...
  8. [8]
    What Is a PDUFA Date? Everything You Need to Know
    Jul 3, 2025 · FDA has a set timeline to review drugs; a norm is 10 months, called a PDUFA date. · Priority Review can shorten this to six months for critical ...
  9. [9]
    Yale experts urge reforming – not gutting – PDUFA - RAPS
    Aug 18, 2025 · The CRRIT researchers noted that pre-PDUFA, the average review time for a new drug application (NDA) was 29 months, and, consequently ...
  10. [10]
    Analysis of US Food and Drug Administration new drug and biologic ...
    Feb 9, 2024 · Over time, the median FDA review time significantly decreased from 26.6 (23.6) months in the pre-PDUFA period, to 12.9 (14.9) in the PDUDA- ...
  11. [11]
    Prescription Drug User Fee Act (PDUFA) Performance Reports - FDA
    Sep 9, 2025 · With increased resources under PDUFA I, FDA was able to commit to and achieve review performance goals that incrementally increased to 90 ...
  12. [12]
    Speed, Safety, and Industry Funding — From PDUFA I to PDUFA VI
    Dec 7, 2017 · By the end of PDUFA I in 1997, the FDA reported meeting its goal of reviewing 90% of applications submitted in the previous fiscal year within ...<|separator|>
  13. [13]
    Prescription Drug User Fee Rates for Fiscal Year 2025
    Jul 31, 2024 · The result is a fee of $4,310,002 per full application requiring clinical data, and $2,155,001 per application not requiring clinical data. IV.Missing: structure | Show results with:structure
  14. [14]
    [PDF] FY 2025 PDUFA Five-Year Financial Plan - FDA
    The fee structure remains unchanged from PDUFA VI with two types of fees: application fees and program fees. 2 The PDUFA VII Commitment Letter: https://www.fda.
  15. [15]
    FDA User Fees: Examining Changes in Medical Product ... - NCBI
    User fees account for 66 percent of the human drugs budget, 43 percent of the biologics budget, and 35 percent of the medical devices budget. Most of the ...
  16. [16]
    [PDF] A Legal Challenge of the Prescription Drug User Fee Act
    Oct 1, 2016 · empirical studies show that there has been “[n]o significant effect of PDUFA . . . on the rate of withdrawals of new-drug approvals” when ...
  17. [17]
    Understanding the nuts and bolts of the reauthorization process for ...
    Aug 9, 2024 · The Prescription Drug User Fee program, better known as PDUFA, is one of the single most important programs at the FDA and is responsible ...<|separator|>
  18. [18]
    [PDF] PDUFA and Initial U.S. Drug Launches
    the problem of drug lag and drug access for AIDS patients, and AIDS ... Among drug approvals in. 1990-1992 prior to PDUFA, the mean review time was 31 months.
  19. [19]
    Ten Things You Need to Know about the FDA User Fee ... - Third Way
    Apr 20, 2017 · But it was truly the AIDS crisis that gave birth to PDUFA. AIDS activists fought for faster drug approvals and earlier access to ...
  20. [20]
    Reauthorization of the Prescription Drug User Fee Act; Public Meeting
    May 19, 2025 · PDUFA authorizes FDA to collect user fees to support the process for the review of human drug applications. The current legislative authority ...Missing: objectives | Show results with:objectives
  21. [21]
    [PDF] Assessing the Impacts of the Prescription Drug User Fee Acts ...
    2 Little empirical evidence has been put forward to make the case that the FDA is too slow or too fast in its drug approval process, partly due to significant ...
  22. [22]
    [PDF] Prescription Drug User Fee Act - FDA
    Since the enactment of PDUFA I in 1992, FDA has used PDUFA resources to significantly reduce the time needed to evaluate new drugs and biological products.
  23. [23]
    PDUFA Legislation and Background: PDUFA II - FDA
    Feb 2, 2018 · The FDA Modernization Act of 1997, enacted on November 21, 1997, reauthorized the Prescription Drug User Fee Act of 1992 for five more years (PDUFA II).
  24. [24]
    The Pediatric Exclusivity Provision - FDA
    Apr 21, 2022 · The pediatric exclusivity provision, enacted in 1997, was the first pediatric drug development incentive legislation. It was part of the FDA ...
  25. [25]
    The Prescription Drug User Fee Act: History and Reauthorization ...
    Under PDUFA IV, user fees increased 25 percent, but the money was rerouted to risk mitigation, instead of spending it on spurring innovation. REMS is an ...Missing: evidence | Show results with:evidence
  26. [26]
    PDUFA Legislation and Background: PDUFA III - FDA
    Sep 22, 2017 · Recognizing the necessity of reauthorizing PDUFA before the end of FY 2002 to assure continuity of operations, Congress enacted the Public ...Missing: exclusivity extensions
  27. [27]
    Prescription Drug User Fee Act III Five-Year Plan; Availability
    Oct 14, 2003 · FDA is announcing the availability of an internal planning document entitled “PDUFA III Five-Year Plan.” PDUFA was amended and extended through ...<|separator|>
  28. [28]
    [PDF] The Prescription Drug User Fee Act: Structure and Reauthorization ...
    Oct 7, 2002 · On March 13, 2002, the Bush Administration released its performance goals for the PDUFA III ... extension of exclusivity or patent protection.
  29. [29]
    PDUFA Legislation and Background: PDUFA IV - FDA
    May 3, 2016 · The reauthorization of PDUFA (through FY 2012) significantly broadened and upgraded the agency's drug safety program, increased resources for ...
  30. [30]
    The Prescription Drug User Fee Act: Much More Than User Fees - NIH
    PDUFA II (1997) created and PDUFA V (2012) made permanent a 6-month market exclusivity extension (delaying generic competition) granted to drugmakers for ...
  31. [31]
    Frequently Asked Questions: Breakthrough Therapies - FDA
    Mar 21, 2024 · Breakthrough therapy designation is intended to expedite the development and review of drugs for serious or life-threatening conditions.
  32. [32]
    Data Driven Insights | PDUFA at a Glance
    Jul 12, 2025 · PDUFA provides the FDA with resources for efficient, predictable, and transparent review timelines, helping to get new medicines to patients ...
  33. [33]
    Electronic Regulatory Submission and Review - FDA
    Jun 12, 2025 · ... (eCTD) is the standard, accepted electronic format for the following submission types: New Drug Application (NDA) ... (BLA); Master files: Drug ...
  34. [34]
    [PDF] Drug Development- FDA's Priority Review Voucher Programs
    Jan 31, 2020 · FDA informs the drug sponsor of a priority review designation within 60 days of the receipt of the drug application. Designation of a drug ...
  35. [35]
    [PDF] 2024 New Drug Therapy Approvals Annual Report - FDA
    In 2024, 28 of the 50 novel drugs approved (56%) were designated priority review. A drug receives a priority review if CDER determines that the drug treats a ...
  36. [36]
    [PDF] SOPP 8402:Designation of Amendments as Major - FDA
    Oct 10, 2024 · When the goal date is extended by a major amendment, a letter will be issued to the applicant communicating the new goal date and the ...
  37. [37]
    Cost-benefit analysis of the FDA: The case of the prescription drug ...
    In Section 6 we estimate negative safety effects of PDUFA by considering their effects on drug withdrawals as well as calculating the equivalent losses in ...
  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: post | Show results with:post
  39. [39]
    New Drug Therapy Approvals 2022 - FDA
    Jan 10, 2023 · CDER's Annual Novel Drug Approvals: 2013 - 2022. From 2013 through 2022, CDER has averaged about 43 novel drug approvals per year. First-in ...<|separator|>
  40. [40]
    A Comprehensive Review of US-FDA Novel Drug Approvals from ...
    Jul 18, 2025 · The total number of drugs approved from 2013 to 2022 was 428 (average of 43 novel drugs/ year). 40% were first-in-class drugs. Access to novel ...
  41. [41]
    Preserving PDUFA Is Critical for U.S. Biopharmaceutical Innovation
    Jul 1, 2025 · A cornerstone of U.S. biopharmaceutical leadership, PDUFA has dramatically accelerated drug approvals and patient access, but looming FDA ...
  42. [42]
    New Drugs and HIV Mortality | NBER
    The use of the new drugs also reduced hospital admission rates for patients with HIV/AIDS, the number of hospital admissions, and thus the cost, of keeping ...
  43. [43]
    [PDF] Statins: A Success Story Involving FDA, Academia and Industry
    At the time of approval, FDA had residual safety concerns about this class of drugs in three areas: lens (eyes), liver and muscles.Missing: impact | Show results with:impact
  44. [44]
    How the Prescription Drug User Fee Act Supports Life-Sciences ...
    Feb 27, 2017 · It shows how PDUFA has played a key role in accelerating timely patient access to new medicines and in providing a key pillar of a broader ...
  45. [45]
    The Safety and Efficacy of the FDA | NBER
    Jun 1, 2006 · "The more rapid access to drugs on the market enabled by the Prescription Drug User Fee Act saved the equivalent of 180 to 310 thousand life- ...
  46. [46]
    Cost-Benefit Analysis of the FDA: The Case of the Prescription Drug ...
    ... PDUFA saved the equivalent of 140,000 to 310,000 life years. Additionally, we estimate an upper bound on the adverse effects of PDUFA based on drugs ...
  47. [47]
    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 ...
  48. [48]
    Drug-review deadlines and safety problems - PubMed
    Mar 27, 2008 · PDUFA deadlines have appreciably changed the approval decisions of the FDA. Once medications are in clinical use, the discovery of safety ...
  49. [49]
    Increased warnings and withdrawals after faster approval
    Aug 16, 2014 · Between 1975 and 2009, the FDA approved 748 NMEs, of which 15% received black-box warnings and 4% were withdrawn for safety reasons. Half of the ...Missing: rates | Show results with:rates
  50. [50]
    Serious Risks And Few New Benefits From FDA-Approved Drugs
    Jul 6, 2015 · A systematic study of shortened reviews found that each 10-month reduction in review time produced an 18 percent increase of serious adverse ...Missing: PDUFA | Show results with:PDUFA<|separator|>
  51. [51]
    effects of PDUFA on approval times and withdrawal rates - Nature
    Jul 1, 2005 · The eight drugs withdrawn in the post-PDUFA period were bromfenac, mibefradil, cerivastatin, troglitazone, grepafloxacin, rotavirus vaccine (we ...
  52. [52]
    GAO-02-958, Food and Drug Administration: Effect of User Fees on ...
    When withdrawal rates are compared for the 8-year periods before and after PDUFA, the increase is from 3.10 to 3.47 percent. Drug withdrawals have been ...
  53. [53]
    Drug Approvals and Deadly Delays - Competitive Enterprise Institute
    Nov 1, 2010 · For example, consider FDA's incredibly long delay in approving beta-blockers to reduce the risk of second heart attacks. By the mid-1970s this ...Missing: lost | Show results with:lost
  54. [54]
    Deaths Due to US Regulatory "Drug Lag" | The Decentralized FDA
    Jul 19, 2022 · The agency's withholding of beta-blockers was alone responsible for probably tens of thousands of deaths . Data from the Tufts Center for the ...
  55. [55]
    Evaluating PDUFA: Paying for FDA Drug-Application Reviewers by ...
    Mar 7, 2025 · Studies show that health gains from faster access to new medicines totally swamped the health losses from more unsafe drugs. Did PDUFA reduce ...Missing: adverse | Show results with:adverse
  56. [56]
    [PDF] Prescription Drug User Fee Amendments - FDA
    In FY 2024, FDA had net collections of $1.381 billion in prescription drug user fees, spent $1.377 billion in user fees for the human drug review process, and ...
  57. [57]
    Effects of PDUFA on approval times and withdrawal rates
    Aug 7, 2025 · In a series of studies, Olson examined drugs approved before and after the implementation of PDUFA, and found that drugs that received ...
  58. [58]
    [PDF] Continuing Appropriations Act Includes FDA Reauthorization of User ...
    Oct 4, 2022 · The Act reauthorizes PDUFA from FY 2023 through FY 2027.6 PDUFA VII increases the FY 2023 annual base revenue for prescription drug user fees to ...Missing: attainment | Show results with:attainment<|separator|>
  59. [59]
    PDUFA VII: Fiscal Years 2023 – 2027 - FDA
    Apr 24, 2023 · Prescription Drug User Fee Act (PDUFA) that provides FDA with the necessary resources to maintain a predictable and efficient review process ...Missing: definition | Show results with:definition
  60. [60]
    Rare Disease Endpoint Advancement Pilot Program - FDA
    Jan 3, 2024 · Under the pilot program, sponsors may begin submitting RDEA program proposal requests (RDEA proposals) in the fourth quarter of fiscal year 2023 ...Missing: digital incentives
  61. [61]
    [PDF] Statement for the Record - National Organization for Rare Disorders
    As part of PDUFA VII, the FDA has committed to incorporating rare disease training into the core curriculum for reviewers. Rare disease drug development ...Missing: digital | Show results with:digital
  62. [62]
    Completed PDUFA VII Deliverables - FDA
    PDUFA must be reauthorized every five years, and in September 2022, the President signed into law the sixth reauthorization of PDUFA, which establishes the ...
  63. [63]
    The FDA PDUFA VII Goals Letter (FY 2023-2027) - FDA Law Blog
    Sep 21, 2021 · Notable Dates and Timelines – Initiation of the pilot program, by December 31, 2022; Annual reports, starting by June 30, 2024 and at least ...
  64. [64]
    FDA staff struggle to meet product review deadlines after DOGE layoffs
    Mar 27, 2025 · Exclusive: FDA staff struggle to meet product review deadlines after DOGE layoffs · Some scientists assigned double the number of new product ...
  65. [65]
    PDUFA VIII: Fiscal Years 2028 – 2032 - FDA
    May 23, 2025 · The Food and Drug Administration is holding a public meeting on July 14, 2025 to kick off the process for reauthorization of the Prescription Drug User Fee Act ...
  66. [66]
    Reauthorization of the Prescription Drug User Fee Act; Public Meeting
    May 19, 2025 · The current authorization of the program (PDUFA VII) expires in September 2027. Without new legislation, FDA will no longer be able to collect ...Background · II. What is PDUFA and what...
  67. [67]
    FDA Commissioner Marty Makary Proposes Lowering Industry User ...
    Jul 15, 2025 · FDA Commissioner Marty Makary wants to lower prescription drug user fees for the next iteration of the program, continuing a series of high-level policy ...
  68. [68]
    Makary suggests lower PDUFA fees as reauthorization process begins
    Jul 14, 2025 · For FY 2025, the PDUFA fee for an application requiring clinical data is $4.3 million, while the annual program fee is just over $400,000. About ...Missing: objectives | Show results with:objectives
  69. [69]
    ASGCT Submits Comments in Response to FDA's PDUFA VIII…
    Sep 11, 2025 · PDUFA is the law that authorizes FDA to collect user fees from companies, which support FDA operations alongside congressional appropriations.
  70. [70]
    Industry and consumer groups call for improved review practices ...
    Aug 20, 2025 · The group said that PDUFA VIII could be improved by focusing on facilitating first cycle review of new applications and helping FDA optimize ...