Fact-checked by Grok 2 weeks ago

Investigational New Drug

An Investigational New Drug (IND) application is a formal request submitted to the (FDA) by a —typically a pharmaceutical company, researcher, or —to obtain authorization for shipping an unapproved drug or biological product across state lines and administering it to human subjects in clinical trials, contingent on preclinical data demonstrating reasonable safety for initial use. The IND serves as the gateway from laboratory and to human evaluation, exempting the from prohibitions under the Federal Food, Drug, and Cosmetic Act against interstate commerce of unapproved substances when used investigatively. The FDA's review focuses primarily on assessing whether the proposed trials pose undue risk to participants, evaluating submitted data on the drug's chemistry, manufacturing controls, , , and clinical protocols; the agency has 30 calendar days to impose a clinical hold if deficiencies are identified, after which the becomes effective and trials may commence unless notified otherwise. Sponsors bear ongoing responsibilities, including reporting adverse events, ensuring investigator qualifications and oversight, and adhering to good clinical practices outlined in 21 CFR Part 312. INDs fall into commercial categories for industry-led development aimed at eventual marketing approval or research categories such as investigator-initiated studies, emergency use for urgent unmet needs, and treatment INDs for to promising therapies in serious conditions prior to full approval. While the IND framework has facilitated the safe advancement of thousands of therapeutics since its codification in the 1960s—driven by lessons from historical tragedies like thalidomide-induced birth defects—it presents procedural challenges, including rigorous preclinical requirements that can delay entry into trials and impose significant resource burdens on smaller sponsors, potentially hindering innovation in resource-constrained settings. Pre-IND consultations with the FDA are available to mitigate such hurdles, though empirical data indicate variable success rates in first-submission clearances due to inconsistencies in data quality and protocol design.

Definition and Purpose

Core Concept and Objectives

An Investigational New Drug (IND) application is a regulatory submission to the U.S. (FDA) that enables a sponsor to conduct clinical trials for a or biological product not yet approved for . It provides an exemption from provisions of the Federal Food, , and Cosmetic Act prohibiting interstate shipment and use of unapproved in humans, synonymous with a "Notice of Claimed Investigational Exemption for a New Drug." The core concept centers on bridging —where sponsors assess if a product is reasonably safe for initial human testing—with controlled human studies to evaluate safety, dosing, and preliminary efficacy. The primary objectives of an IND are to safeguard trial participants by assuring the FDA of adequate protections for their rights, safety, and welfare, while facilitating to support eventual approval. Sponsors must submit comprehensive preclinical data from animal and studies, manufacturing details on composition, stability, and controls, as well as clinical protocols, investigator qualifications, (IRB) approvals, and procedures. The FDA reviews submissions within 30 days, allowing trials to proceed unless a clinical hold is imposed for unresolved safety concerns. In Phases 2 and 3, additional focus shifts to verifying the drug's effectiveness through rigorous trial designs. Governed by 21 CFR Part 312, the IND framework promotes phased progression: Phase 1 emphasizes safety and in small groups, Phase 2 explores and side effects in patients, and Phase 3 confirms benefits versus risks in larger populations. This structure minimizes risks by requiring iterative evidence accumulation, with ongoing amendments for protocol changes or new safety data. Ultimately, IND-enabled trials generate the pivotal evidence for a (NDA), ensuring only drugs demonstrating a favorable benefit-risk profile reach the market.

Distinction from Approved Drugs

An application enables the sponsor to ship an unapproved drug across state lines for use in clinical trials, providing an exemption from certain provisions of the Federal Food, Drug, and Cosmetic Act under 21 CFR Part 312, whereas approved drugs have undergone a review demonstrating substantial evidence of and for marketing. The IND focuses on preclinical data sufficient to justify human testing, including animal and studies to assess potential risks, but does not require proof of efficacy or the comprehensive needed for approval. In contrast, approved drugs must show benefits outweigh risks through typically two adequate and well-controlled Phase 3 trials, along with manufacturing and labeling details in the NDA. Use of an is strictly limited to controlled clinical investigations under an FDA-reviewed , with distribution confined to qualified investigators and a mandatory 30-day waiting period after submission to allow FDA review for safety holds. Approved drugs, however, may be legally marketed and prescribed for their indicated uses without such restrictions, subject to post-market surveillance. Sponsors of INDs are prohibited from commercial promotion or marketing the drug as safe or effective, as this would violate regulations reserving such claims for approved products; violations can lead to enforcement actions under the Act. These distinctions ensure that investigational drugs remain experimental, protecting participants from unverified risks while allowing innovation to proceed under oversight, unlike approved drugs which enter the market with established benefit-risk profiles verified by the FDA's Center for Drug Evaluation and Research (CDER). For instance, while Phase 1-3 trials under an gather escalating evidence of safety and dosage, only successful submission transitions the drug to approved status, often requiring additional risk evaluation and mitigation strategies (REMS) for certain products.

Historical Development

Origins in Early Drug Regulation

The Pure Food and Drugs Act of 1906 represented the initial federal intervention in drug regulation, prohibiting the interstate shipment of adulterated or misbranded drugs but imposing no requirements for pre-market demonstration of safety or efficacy. This legislation, enacted amid public outcry over patent medicines containing dangerous substances like opium and cocaine, focused primarily on truthful labeling and purity rather than testing protocols, allowing manufacturers to introduce new drugs without regulatory review as long as claims were not demonstrably false. Clinical investigations during this era operated with minimal oversight, often relying on voluntary standards from bodies like the American Medical Association's Council on Pharmacy and Chemistry, established in 1905 to assess drug quality through laboratory analysis and expert review. The catalyst for more structured regulation came with the disaster in 1937, where a firm reformulated the using toxic as a , resulting in over 100 deaths, predominantly among children, due to untested toxicity. This tragedy exposed the inadequacies of the 1906 Act, as the product was neither adulterated nor misbranded under existing definitions but caused widespread harm from lack of data. In response, passed the Federal Food, Drug, and Cosmetic Act (FD&C Act) on June 25, 1938, which mandated that manufacturers file New Drug Applications (NDAs) demonstrating a drug's through adequate testing before interstate . The Act defined a "new drug" broadly to include unapproved substances or those with new uses, shifting the burden to prove onto sponsors while authorizing FDA factory inspections and extending oversight to and devices. Although the 1938 FD&C Act did not establish a formal application process, it introduced an investigational exemption permitting the interstate shipment of unapproved drugs for purposes, provided they were not sold commercially and bore a cautionary label: "Caution—New Drug—Limited by Federal (or ) law to investigational use." This provision implicitly recognized the need for a of pre-marketing testing separate from commercial distribution, allowing physicians and researchers to conduct clinical studies under loose FDA notification via the NDA process, without standardized protocols for trial design, , or reporting. Such exemptions facilitated early clinical evaluations but lacked rigorous safeguards, as evidenced by ongoing incidents of unsafe investigational practices until subsequent reforms.

Evolution Post-Thalidomide and 1962 Amendments

The tragedy, involving the sedative's distribution in from 1957 and subsequent recognition of its severe teratogenic effects causing an estimated 10,000 to 12,000 birth defects by 1961, highlighted deficiencies in pre-market drug testing and prompted urgent reforms in the United States. Although was not approved for marketing in the U.S. due to objections raised by FDA reviewer regarding inadequate safety data, reports of and emerging fetal malformations intensified scrutiny of the existing regulatory framework under the 1938 Federal Food, Drug, and Cosmetic Act, which emphasized safety but lacked robust efficacy requirements or standardized oversight. Enacted on October 10, 1962, as the Kefauver-Harris Amendments, this legislation fundamentally altered investigational drug processes by mandating that manufacturers provide "substantial evidence" of a drug's effectiveness, in addition to safety, through "adequate and well-controlled investigations" prior to approval. For investigational new drugs (s), the amendments required sponsors to obtain FDA permission via formal IND submissions before interstate shipment or human testing, shifting from a pre-1962 system of largely voluntary compliance and minimal federal intervention to one enforcing detailed protocols, preclinical animal data, and manufacturing controls to mitigate risks identified in events like . from trial participants became obligatory, with regulations prohibiting promotional claims about unproven investigational agents, thereby establishing ethical and scientific guardrails for early-phase studies. In response, the FDA issued implementing regulations in May 1963, formalizing the IND application under 21 CFR Part 312 and introducing requirements for protocols that included investigator qualifications, subject selection criteria, and mechanisms for reporting. This evolution institutionalized a phased approach to testing—implicitly laying groundwork for later explicit 1, 2, and 3 designations—while empowering the agency to suspend or terminate trials if safety concerns arose, as evidenced by heightened scrutiny of congenital malformation risks in subsequent submissions. These changes directly addressed thalidomide's causal pathway of insufficient long-term and uncontrolled distribution, fostering a precautionary yet evidence-based paradigm that prioritized from controlled data over anecdotal or manufacturer-driven observations.

Regulatory Framework

The legal foundation for the process derives from Section 505(i) of the Federal Food, Drug, and Cosmetic Act (FD&C Act), codified at 21 U.S.C. § 355(i), which empowers of Health and Human Services—delegated to the —to authorize the interstate shipment of unapproved new drugs or biological products solely for qualified investigational use by experts. This provision exempts such shipments from the general on distributing unapproved drugs across state lines under Section 301(d) of the FD&C Act (21 U.S.C. § 331(d)), provided the IND application demonstrates that the investigation will be conducted in accordance with regulatory safeguards to protect human subjects. The statute specifies that authorizations under Section 505(i) are limited to quantities sufficient for the intended investigations and require conditions such as , recordkeeping, and progress reports to the FDA. The FD&C Act, originally enacted on June 25, 1938, established the core framework for drug regulation, but the IND mechanism was formalized and strengthened by the Kefauver-Harris Amendments of October 10, 1962 (Public Law 87-781), which amended Section 505 to mandate preclinical and clinical data supporting for initial human testing and for eventual approval. These amendments responded to public health crises like thalidomide-induced birth defects, imposing requirements for IND submissions to include protocols ensuring investigations are scientifically sound and ethically conducted, while allowing FDA imposition of clinical holds if risks outweigh benefits. Subsequent statutes, such as the Safe Medical Devices Act of 1990 and the Food and Drug Administration Modernization Act of 1997, have refined IND authorities without altering the foundational statutory basis in Section 505(i). Implementing regulations in 21 CFR Part 312 operationalize this statutory authority, outlining IND content requirements (e.g., animal data, manufacturing information, and qualifications under §§ 312.20–312.38), FDA procedures, and obligations for monitoring and reporting adverse events. These regulations, promulgated under the broad rulemaking powers in Sections 701 and 505 of the FD&C Act (21 U.S.C. §§ 371, 355), ensure compliance with constitutional by requiring FDA notifications of holds with specific grounds, such as insufficient safety data or protocol deficiencies (§ 312.42). Authority for biological products under INDs extends via the (42 U.S.C. § 262), cross-referenced in 21 CFR Part 312, harmonizing oversight for drugs and biologics. Violations of IND conditions can trigger enforcement under FD&C Act Sections 301–303 (21 U.S.C. §§ 331–333), including injunctions, seizures, or criminal penalties for misbranded or adulterated investigational products.

FDA Oversight and Authority

The (FDA) derives its authority over (IND) applications from Section 505(i) of the Federal Food, Drug, and Cosmetic Act (21 U.S.C. § 355(i)), which empowers the agency to promulgate regulations permitting the interstate shipment of unapproved drugs for clinical investigations under controlled conditions, exempting them from premarket approval requirements while mandating safeguards for human subjects. This statutory framework, detailed in 21 CFR Part 312, positions the FDA as the primary regulator ensuring that clinical trials proceed only if they pose no unreasonable risk based on available data. The Kefauver-Harris Amendments of 1962 fundamentally strengthened this authority by requiring manufacturers to submit evidence from adequate and well-controlled investigations to demonstrate both safety and efficacy before marketing, while formalizing FDA oversight of investigational phases through rules on , reporting, and protocol adequacy. In reviewing an , the FDA's core objectives are to protect the safety and rights of trial participants across all phases and, for Phases 2 and 3, to verify the scientific rigor necessary for assessing the drug's potential effectiveness. The agency conducts this review within 30 calendar days of submission; absent notification of issues, the IND becomes effective, allowing trials to commence, though the FDA retains ongoing monitoring powers. FDA enforcement mechanisms include the imposition of clinical holds, which suspend or delay trials if preclinical data indicate unreasonable risks, investigators or protocols are inadequate, or sufficient is lacking to assess —grounds specified under 21 CFR § 312.42 for all phases, with Phase 1 holds additionally prohibiting dosing until resolved. Sponsors must address holds promptly, and the FDA can terminate an IND entirely under § 312.44 if trials present unreasonable risks, the fails to pursue development diligently, or there is systemic noncompliance, providing 30 days for before final action. During active investigations, oversight extends to mandatory reporting: sponsors submit IND reports within 15 calendar days for serious, unexpected adverse events (§ 312.32), annual progress reports within 60 days of each anniversary (§ 312.33), and investigators report deviations or unanticipated problems to sponsors for FDA relay (§ 312.64). Further, the FDA conducts inspections of records, sites, and manufacturing facilities under § 312.58 and may disqualify noncompliant clinical per § 312.70, ensuring adherence to good clinical practices. These powers collectively enable the to mitigate risks causally linked to insufficient preclinical or , as evidenced by historical interventions like holds on in the due to patient deaths from inadequate . While bear primary responsibility for conduct, FDA underscores a precautionary approach, prioritizing empirical signals over expediency in unproven therapies.

Types of IND Applications

Commercial INDs

A commercial Investigational New Drug (IND) application is submitted by a sponsor, typically a pharmaceutical corporation or other commercial entity, with the explicit intent to develop the investigational drug or biologic for eventual commercialization via a New Drug Application (NDA) or Biologics License Application (BLA). The sponsor bears primary responsibility for all aspects of the clinical development program, including protocol design, site selection, adverse event monitoring, data analysis, and regulatory compliance to support marketing approval. This contrasts with investigator-initiated INDs, where a physician-scientist independently drives non-commercial research without plans for product marketing. Commercial INDs enable the conduct of pivotal clinical trials—typically Phases 1 through 3—required to demonstrate safety and efficacy for broad patient populations, adhering to (GCP) standards under 21 CFR Part 312. Sponsors must provide comprehensive preclinical data, manufacturing controls, and environmental assessments, with the FDA reviewing submissions within 30 days to issue a clinical hold if risks outweigh benefits or protocols are inadequate. Electronic submissions in eCTD format are mandatory for commercial INDs to facilitate FDA's structured review process. The FDA may reclassify an initially submitted research IND as commercial if evidence emerges of commercialization intent, such as corporate sponsorship or plans for large-scale trials leading to filing, ensuring appropriate oversight levels. Commercial sponsors often engage contract research organizations (CROs) for operational execution, but retain ultimate accountability for and safety reporting, with annual reports summarizing progress and amendments required for protocol changes. This framework supports efficient progression from to approval, as evidenced by the majority of FDA-approved drugs originating from commercial pathways since the 1962 Kefauver-Harris Amendments strengthened efficacy requirements.

Investigator-Initiated INDs

An investigator-initiated , also referred to as a research , is submitted by a who both initiates and conducts the clinical , under whose immediate direction the investigational or biologic is administered or dispensed to human subjects. This type of enables studies of unapproved or approved products for new indications, routes of administration, dosages, or patient populations, primarily for non-commercial purposes such as advancing scientific knowledge or addressing unmet clinical needs rather than product . The submitting serves as the sponsor-investigator, assuming combined responsibilities defined under 21 CFR Part 312 for both sponsoring the trial—selecting qualified investigators, providing adequate supplies with proper labeling and controls, monitoring study conduct, and reporting safety data to the FDA—and investigating, including protecting subject rights, obtaining , and maintaining accurate records. Unlike commercial INDs, which are filed by pharmaceutical manufacturers or corporate sponsors intending to gather data toward approval and requiring mandatory electronic submission in eCTD format, investigator-initiated INDs are typically shorter-term, academic or institutionally supported efforts where paper submissions remain permissible and electronic formatting is optional. Sponsor-investigators often operate within resource-constrained settings like universities or hospitals, necessitating institutional oversight for sponsor duties such as monitoring and data management, which can be burdensome without dedicated staff. To facilitate submissions, sponsor-investigators may obtain letters of authorization from drug manufacturers to reference existing nonclinical, chemistry, manufacturing, and controls () data, avoiding redundant generation of preclinical information when studying approved drugs for new uses. The application process requires standard IND components under 21 CFR 312.23, including FDA Forms 1571 (statement of investigator), 1572 (statement of investigator commitment), a specifying research intent, , summarizing known drug effects and risks, and sufficient preclinical / data to support the proposed study. Submissions are sent in triplicate to the FDA's Central Document Room, becoming effective 30 days after receipt unless a clinical hold is imposed for deficiencies like inadequate data or risks outweighing benefits. FDA guidance emphasizes complete applications to minimize delays, advising sponsor-investigators to pre-submission consult IRBs and potentially request pre-IND meetings for complex proposals, while ensuring compliance with (GCP) and (IRB) approval prior to dosing. These INDs support a significant portion of early-phase academic research but require rigorous documentation to uphold subject , with the FDA retaining authority to inspect records and terminate investigations for non-compliance.

Emergency Use INDs

Emergency Use INDs permit the administration of an investigational drug or biologic to a single in a life-threatening situation or one involving irreversible debilitation where no satisfactory alternative therapy exists and insufficient time allows for submission of a standard application under 21 CFR 312.23 or 312.20. This mechanism facilitates rapid FDA authorization via telephone or other expedited means, enabling shipment of the product prior to formal IND filing if the agency deems the circumstances warrant it. Such uses typically arise when do not qualify for ongoing clinical trials or no open protocol matches their condition. The submission process begins with the treating contacting the drug's or manufacturer to secure access and a Letter of Authorization, followed by immediate outreach to the FDA for verbal or written approval. For CDER-regulated products, contact the Division of Drug Information at (855) 543-3784 or [email protected]; for CBER-regulated biologics, use 240-402-8020 or 800-835-4709; after-hours requests go to the Office of Emergency Operations at 866-300-4374 or 301-796-8240. FDA aims to respond promptly, often within hours, authorizing upon review of details, proposed , and rationale. Within 15 days post-authorization, the physician must submit an individual IND using Form FDA 3926, including the Letter of Authorization, consent (where feasible), and protocol summary. Emergency Use INDs include exemptions from prospective Institutional Review Board (IRB) approval for a single administration, per 21 CFR 56.104(c), provided the situation meets criteria of immediate threat without alternatives. Retrospective IRB notification is required within five working days after use, with full board review needed for any repeat administrations. Informed consent exceptions apply under 21 CFR 50.23(a) if obtaining it would delay life-saving intervention, but investigators must document consultation attempts with a legally authorized representative and notify the IRB within five days. Post-use obligations encompass safety reporting: unexpected fatal or life-threatening adverse events within seven days, other serious events within 15 days, and annual reports within 60 days of the anniversary date. Amendments for treatment changes and a final results summary via Form FDA 3926 ensure ongoing oversight, distinguishing this from broader pathways like Treatment INDs, which involve more structured protocols for multiple patients. These provisions, outlined in FDA guidance from 1998 and updated procedures, prioritize patient access while maintaining regulatory safeguards against unproven risks.

Application Submission Process

Pre-Submission Preparation and Meetings

Prior to submitting an application to the , sponsors must compile comprehensive preclinical data, including and studies sufficient to support initial testing, as well as chemistry, manufacturing, and controls () information demonstrating drug identity, quality, purity, and strength. This preparation ensures the application addresses safety concerns under 21 CFR Part 312, where inadequate data can lead to clinical holds delaying trials. Guidance documents from the Center for Drug Evaluation and Research (CDER) emphasize early alignment with FDA expectations to avoid rework, recommending review of relevant ICH and FDA-specific standards for nonclinical and manufacturing requirements. Pre-IND meetings, classified as Type B formal meetings under FDA policy, provide an optional but highly recommended opportunity for sponsors to obtain non-binding feedback on development plans before IND submission. These interactions aim to clarify data requirements, refine clinical protocols, and mitigate risks of rejection or holds, particularly for drugs or those targeting serious conditions, by allowing discussion of preclinical adequacy, proposed dosing, and designs. The FDA's Pre-IND Consultation Program facilitates these early communications, organized by therapeutic division, to guide sponsors toward complete submissions that enhance development efficiency and reduce timelines to market. To request a pre-IND meeting, sponsors submit a written request to the relevant CDER or Center for Biologics Evaluation and Research (CBER) division, detailing the product, meeting purpose, proposed agenda, specific questions categorized by discipline (e.g., , clinical), quantitative composition, indication, and dosing regimen; requests should be made without a prior IND if none exists. The FDA responds within 21 days and schedules the meeting within 60 days of receipt, with formats including in-person, , or written response only (WRO), though virtual options predominate post-2020. A briefing package must follow 4 weeks prior (or 30 days for certain programs like cellular therapies), containing a program synopsis, summaries of preclinical/clinical strategies and data (e.g., results, manufacturing details), and contextual support for questions without full raw datasets to enable focused review. FDA preliminary comments are typically provided 24-48 hours before the meeting, emphasizing adherence to good clinical practices and alignment with guidances to maximize utility. Such meetings yield benefits like FDA endorsement of endpoints or study designs, potentially averting costly additional preclinical work, though outcomes remain advisory and do not guarantee IND approval. Sponsors are advised to limit questions to pivotal issues and prepare multidisciplinary teams, as unresolved gaps identified in these sessions inform final IND assembly.

Required Documentation and Forms

The submission of an Investigational New Drug (IND) application to the U.S. Food and Drug Administration (FDA) mandates specific forms and supporting documentation as outlined in 21 CFR Part 312. These elements ensure the FDA receives organized, comprehensive information to evaluate the proposed clinical investigation's safety for human subjects. The core forms include FDA Form 1571 for administrative details, FDA Form 1572 for investigator commitments, and FDA Form 3674 for clinical trial registration compliance. Accompanying documentation encompasses scientific and regulatory data, typically submitted in triplicate or electronically via the FDA's gateway. FDA Form 1571 serves as the IND cover sheet, capturing essential administrative data such as the sponsor's identity, drug name, investigational plan summary, and commitments to comply with FDA regulations, including protocol amendments and reporting. FDA Form 1572, signed by each , certifies their qualifications, agreement to adhere to the , supervision of the study, and personal conduct of or oversight over delegated tasks, while disclosing financial interests. FDA Form 3674 certifies the sponsor's or investigator's compliance with registration and results requirements under the FDA Amendments Act of 2007. In cases involving potential environmental impacts, sponsors may submit FDA Form 3455 to request categorical exclusion from a full environmental assessment under the . Beyond forms, required documentation includes a concise identifying the submission type, drug details, and review division; a detailed with page references; and an introductory statement summarizing the investigational plan, rationale, and foreign studies if applicable. Key scientific sections comprise the , which compiles nonclinical and clinical data on the drug's , , , and risks; clinical protocols detailing study design, eligibility criteria, dosing, and endpoints; , manufacturing, and controls (CMC) information on drug substance and product stability; and / summaries from preclinical studies justifying human testing. Additional elements, such as summaries of prior human experience and pediatric study plans if relevant, must be included to address and gaps. Submissions must be well-organized, with clear labeling and indexing, to facilitate FDA review within the 30-day period before the IND becomes effective absent a clinical hold. Electronic formats are preferred for efficiency, aligning with FDA's Study Data Technical Conformance Guide. Failure to provide complete documentation can result in incomplete application status or holds, emphasizing the need for thorough preparation.

FDA Review Timeline and Clinical Holds

The U.S. (FDA) reviews an () application within 30 calendar days of receipt to assess whether the proposed clinical investigations may proceed. This period allows FDA to evaluate concerns based on preclinical , , and details, without requiring formal approval for the IND to take effect. Sponsors are prohibited from initiating clinical trials until the 30-day review concludes, unless FDA explicitly notifies them otherwise, ensuring a mandatory waiting period to mitigate risks. If FDA identifies no significant issues warranting intervention, the IND automatically becomes effective at the end of the 30 days, permitting the sponsor to commence dosing in human subjects as outlined in the . This "silent approval" mechanism streamlines the process for low-risk applications while prioritizing participant safety, as the review focuses primarily on whether investigations pose unreasonable hazards rather than comprehensive judgments. FDA may impose a clinical hold—an order to delay a proposed investigation or suspend an ongoing one—if deficiencies exist in areas such as inadequate nonclinical safety data, unreasonable risk from the drug's profile, flawed clinical design, or insufficient controls that could compromise product quality or subject safety. Holds can be complete (halting all enrollment and activities), partial (affecting specific protocols or phases), or targeted to certain investigations under the , with grounds delineated in 21 CFR 312.42, including for Phase 1 trials the lack of justification for initial dose escalation or reliance on unreliable animal models. Such holds occur in response to submissions and aim to address unresolved safety questions empirically, rather than deferring to sponsor assertions without verification. Upon issuance, FDA provides written notification specifying hold reasons, typically within the initial 30-day window or later for amendments. Sponsors must submit a complete response addressing all concerns, after which FDA reviews it within 30 days (or 14 days if requested for expedited cases involving serious risks). Lifting the hold restores IND effectiveness, but unresolved holds can extend timelines significantly, with empirical data indicating they affect a minority of INDs—often under 10% annually—primarily due to gaps in or rigor rather than arbitrary regulatory hurdles. No new subjects may be enrolled during a hold, though ongoing trials under suspension must continue enrolled participants until resolved.

Key Requirements for Approval

Preclinical Pharmacology and Toxicology Data

The preclinical pharmacology and toxicology data submitted in an Investigational New Drug (IND) application comprise non-clinical studies conducted to evaluate the investigational drug's mechanism of action, pharmacological effects, pharmacokinetic properties, and potential toxicity, ensuring reasonable safety for initial human exposure. These data, derived primarily from in vitro experiments and in vivo animal models, must demonstrate an adequate understanding of the drug's effects to support the proposed clinical dose, route of administration, and duration of exposure without posing unreasonable risks. Under 21 CFR 312.23(a)(8), sponsors provide integrated summaries of pharmacological and toxicological findings, including rationale for species and strain selection in animal studies, with full reports for pivotal toxicology studies. Pharmacology studies focus on the drug's primary , elucidating mechanisms of action and dose-response relationships in relevant animal models or isolated systems, often complemented by secondary pharmacodynamic effects on non-target systems. These assessments may include efficacy indicators in disease models, though for IND purposes, emphasis lies on safety-relevant rather than definitive proof-of-concept, with data sufficient to justify the starting clinical dose—typically derived from the (NOAEL) in studies divided by safety factors (e.g., 10-fold inter extrapolation). data, such as , , , and profiles across , bridge preclinical to clinical predictions, informing human dosing via allometric scaling or physiologically based pharmacokinetic modeling where applicable. Toxicology evaluations encompass acute, subchronic, and studies tailored to the proposed duration, including (e.g., , chromosomal aberration assays), carcinogenicity if is planned, and reproductive/developmental for drugs affecting or . Safety pharmacology studies assess core organ systems (cardiovascular, respiratory, central nervous) per ICH S7A guidelines, often integrated into repeat-dose studies, with endpoints like histopathological changes, , and mortality tracked to identify target organs of and reversibility. For Phase 1 INDs, at least one ( and non- preferred) with studies supporting the clinical route and duration is required, though exploratory INDs may rely on with limited data if justified. All pivotal toxicology studies must comply with (GLP) regulations under 21 CFR Part 58 to ensure data reliability, excluding exploratory or mechanism-focused studies where GLP may not apply. The FDA assesses adequacy by whether the data permit an inference of human safety margins, potentially issuing a clinical hold if findings indicate excessive , inadequate dosing justification, or incomplete characterization of risks like in biologics. Submission includes , protocols, and analyses, with deficiencies addressable via amendments, reflecting the causal link between observed animal toxicities and potential human outcomes absent direct ethical testing.

Chemistry, Manufacturing, and Controls (CMC)

The section of an application provides detailed information on the drug substance (active pharmaceutical ingredient), drug product (final ), and associated formulations to ensure their identity, strength, quality, purity, and potency for safe use in clinical investigations. This information is mandated under 21 CFR 312.23(a)(7), which requires sufficient data to support the of proposed studies without necessitating full-scale validation typical of applications. The FDA reviews CMC data to assess manufacturing consistency and potential risks to trial participants, potentially issuing a clinical hold if deficiencies exist, such as inadequate characterization or stability data. For the drug substance, sponsors must describe its physical, chemical, or biological properties; process, including source material and synthesis or purification methods; analytical procedures for identity, strength, quality, and purity (e.g., via , , or bioassays); and data or a commitment to ongoing testing under appropriate conditions. Imported substances require details on foreign sites and compliance with current good practices (cGMP). For biological products, additional specifics on source material and handling are needed to mitigate contamination risks. product includes components (active and inactive), and procedures, analytical controls, and profiles to confirm shelf-life under conditions. CMC requirements scale with clinical phase: Phase 1 INDs demand less comprehensive data, focusing on basic characterization and controls sufficient for short-term safety in small cohorts, whereas Phase 2 and 3 submissions require expanded details on , batch consistency, and comparative analyses to support larger, longer . FDA guidance emphasizes risk-based approaches, prioritizing information on impurities, degradation products, and compatibility that could impact or . Sponsors often reference environmental assessments under 21 CFR 25 if changes pose ecological risks, but core CMC focuses on human subject safety. Adequate CMC documentation enables FDA clearance within 30 days, facilitating trial initiation while iterative amendments address evolving insights.

Initial Clinical Protocol and Investigator Brochure

The initial clinical protocol submitted with an Investigational New Drug (IND) application details the design and conduct of the proposed initial human clinical investigation, typically a Phase 1 study focused on safety, tolerability, and pharmacokinetics in healthy volunteers or patients. This protocol must demonstrate that the trial will not expose participants to unreasonable risks, incorporating elements such as study objectives, patient eligibility criteria, dosing regimens, monitoring procedures, and statistical plans to evaluate drug effects and safety signals. For instance, it requires specifications for enrollment (e.g., inclusion/exclusion based on age, health status, and comorbidities), randomization methods (if applicable), duration of treatment and follow-up, laboratory assessments for toxicity, and contingency plans for adverse events, ensuring alignment with Good Clinical Practice standards. The Investigator Brochure (IB), also required in the IND submission, serves as a comprehensive document for clinical investigators, summarizing all available nonclinical and prior on the investigational drug to inform and safe administration. Under 21 CFR 312.23(a)(8), the IB must include a physical, chemical, and pharmaceutical description of the drug; nonclinical and summaries (e.g., on , , , , and dose-limiting toxicities); any data from previous studies; known risks, including potential carcinogenicity or reproductive effects; and guidance on dosing, contraindications, and precautions. For drugs without prior exposure, the IB relies heavily on preclinical data, emphasizing dose-response relationships and no-observed-adverse-effect levels to justify starting doses in s, often derived from scaling factors like . Together, the and IB enable the FDA's 30-day period to evaluate whether the proposed design mitigates identified risks, with the empowered to impose a clinical hold if deficiencies exist, such as inadequate preclinical justification for dosing or insufficient . In investigator-initiated INDs, the sponsor-investigator typically authors both documents, drawing from manufacturer-provided data where applicable, whereas commercial INDs may reference an existing IB from the drug developer. Updates to the IB are required prior to each clinical study or as new information emerges, ensuring ongoing relevance without delaying initiation if the initial versions meet regulatory thresholds.

Clinical Trials Enabled by IND

Phase Integration and Progression

The application enables the sponsor to conduct clinical trials across multiple phases under a single regulatory framework, with progression facilitated through protocol amendments rather than separate IND submissions for each phase. Upon the IND becoming effective—typically 30 days after submission unless placed on clinical hold—the sponsor may initiate Phase 1 studies as outlined in the initial , focusing on safety, tolerability, , and in a small cohort of 20 to 100 healthy volunteers or patients. Data from Phase 1, including profiles and dosing information, must inform subsequent phases to ensure risk mitigation, with the FDA requiring annual reports and immediate safety updates to maintain IND viability. Progression to Phase 2 involves submitting an amendment with the proposed , which expands to 100 to 300 participants to evaluate preliminary , optimal dosing, and further in the target patient population; this amendment must include integrated summaries of prior phase data, manufacturing updates, and rationale for design changes. FDA review of such amendments occurs within 30 days, potentially imposing holds if unreasonable risks are identified, such as inadequate preclinical support or flaws. End-of-Phase 1 meetings with the FDA are recommended to discuss transition data and Phase 2 planning, ensuring alignment on endpoints like proof-of-concept signals before resource-intensive expansion. For Phase 3, amendments similarly detail large-scale, randomized, controlled trials involving hundreds to thousands of participants to confirm , monitor rare adverse events, and compare against standards of , with integrated datasets from Phases 1 and 2 forming the evidentiary backbone. This phase often incorporates adaptive elements, such as interim analyses for futility or , under FDA guidance for efficient designs that integrate confirmatory objectives without restarting the process. Overall, phase integration emphasizes causal linkages—where early safety signals causally predict later risks—and cumulative data synthesis, reducing attrition by allowing iterative refinements; however, traditional sequential models predominate, with seamless Phase 2/3 trials reserved for robust early data to accelerate timelines while upholding nonclinical and consistency. Sponsors must justify any phase overlaps in amendments, balancing innovation against the statutory mandate to avert unreasonable harm.

Safety Reporting and Amendments

Sponsors of an Investigational New Drug (IND) application must promptly report safety information to the U.S. Food and Drug Administration (FDA) under 21 CFR 312.32 to ensure ongoing monitoring of potential risks during clinical investigations. This includes expedited reporting of serious and unexpected suspected adverse reactions (SARs) observed in U.S. or foreign clinical trials, where "unexpected" refers to events not listed in the investigator's brochure or differing in nature, severity, or frequency from anticipated risks. For fatal or life-threatening SARs, sponsors must notify the FDA by telephone or fax within 7 calendar days, followed by a written IND safety report within 15 calendar days; other serious unexpected SARs require written submission within 15 calendar days. Reports from animal studies, literature, or foreign cases that are both serious and unexpected follow the 15-day timeline, submitted as narratives or on FDA Form 3500A, with each report identifying prior similar submissions to avoid duplication. Non-expedited safety data, such as expected adverse events or aggregate summaries, are reported in annual reports under 21 CFR 312.33, which must include a brief summary of all serious adverse events, ongoing or completed investigations, and any safety-related changes to protocols or manufacturing. Investigators contribute by all adverse events to sponsors without interpreting , while sponsors assess based on biological plausibility, temporal relationship, and consistency across cases. Follow-up reports for initially submitted are required if new information materially affects the assessment, submitted as amendments or annual reports rather than standalone safety reports to reduce administrative burden. As of April 1, 2026, IND safety reports for serious unexpected will transition to submission via the FDA Reporting System (FAERS), aligning with post-marketing for streamlined electronic . Safety findings often necessitate IND amendments to maintain the integrity of clinical protocols and ensure participant protection. Protocol amendments under 21 CFR 312.30 are required for any changes significantly affecting subject , the investigation's scope, or data reliability, such as modifications to dosing, eligibility criteria, or procedures in response to emerging patterns. These must be submitted to the FDA before implementation, except in life-threatening situations where immediate action is needed followed by prompt notification. Information amendments (21 CFR 312.31) convey other updates, like revised investigator brochures incorporating cumulative SAR data, and should be batched not more frequently than every 30 days to minimize submissions. For instance, if preclinical or clinical data reveal new risks requiring protocol alterations, the sponsor submits the amended with justifications, qualifications, and references to prior versions. Failure to amend promptly can lead to clinical holds if the FDA determines risks outweigh benefits.

Annual Submission Volumes

The U.S. Food and Drug Administration's Center for Drug Evaluation and Research (CDER) receives approximately 1,500 initial applications annually, encompassing submissions for small-molecule drugs and related therapeutics from both commercial sponsors and investigators. This volume reflects the steady influx of early-stage proposals, with the majority advancing past the initial 30-day period unless placed on clinical hold for safety or data deficiencies. The Center for Biologics Evaluation and Research (CBER), which oversees INDs for biologics including , therapies, and cellular products, handles a smaller but growing number of submissions, with 777 original applications received in 2021. Overall FDA IND volumes, combining CDER and CBER, range between 1,500 and 2,000 new applications per year, a figure that has remained relatively stable over the past decade amid expansions in sectors like advanced therapy medicinal products. These submissions are predominantly electronic, submitted via the Electronic Submissions Gateway, facilitating efficient processing despite the high caseload. Fluctuations in annual volumes are minimal, though upticks occur in response to priorities, such as increased filings for antiviral or candidates during disease outbreaks or therapeutic breakthroughs. Investigator-initiated INDs, often for academic or off-label uses of investigational agents, contribute significantly to the total but are outnumbered by commercial INDs aimed at eventual market approval. The FDA's capacity to review these applications within statutory timelines—typically 30 days for initial assessments—supports a robust pipeline, though resource constraints in specialized areas like rare diseases can influence prioritization.

Success Rates and Attrition

The attrition rate from Investigational New Drug (IND) submission to the initiation of clinical trials is influenced primarily by FDA clinical holds, which occur in approximately 9% of cases during the initial 30-day review period, often due to deficiencies in chemistry, manufacturing, controls (CMC), pharmacology, or toxicology data. Most holds are resolved within a year, with CMC issues being the most common cause, allowing the majority of INDs—around 91%—to proceed without initial suspension, though sponsor decisions or additional data requirements can further delay or prevent trial starts. Once clinical trials commence under an IND, the overall probability of success from Phase I to FDA approval remains low, typically ranging from 8% to 14% across therapeutic areas, driven by escalating failure rates in later phases due to shortfalls, concerns, and commercial viability assessments. A comprehensive of clinical development from 2011 to 2020 reported phase transition success rates of 70% from Phase I to II, 35% from II to III, and 55% from III to approval submission, yielding a cumulative likelihood of approval from Phase I of about 10%. is particularly acute in Phase II, where failures account for over 50% of discontinuations, compared to issues dominating earlier phases. Success rates vary significantly by modality and indication; for instance, large molecule biologics exhibit a Phase I-to-approval probability of 21%, versus 10% for small molecules, while drugs achieve higher rates (up to 25%) due to regulatory incentives and smaller trial requirements. programs face the lowest success, with Phase II exceeding 70%, reflecting biological complexity and heterogeneous patient responses. These figures underscore systemic challenges, including inadequate preclinical predictors of human efficacy and the high cost of advancing candidates, which prompts strategic by sponsors prioritizing higher-potential assets.
Phase TransitionSuccess Rate (%)Primary Failure Reasons
IND to Phase I~91 (no hold)Regulatory holds (CMC, tox)
Phase I to II63-70Safety/toxicity
Phase II to III31-35Lack of efficacy
Phase III to Approval58-85Efficacy, safety, commercial
Overall (Phase I to Approval)8-14Cumulative phase failures

Notable Examples and Case Studies

Successful Transitions to Market

, marketed as Humira, exemplifies a successful IND transition for a biologic therapeutic. Developed by Pharma (later acquired by , now ), an was filed in December 1999 to support phase 3 clinical studies in patients. Pivotal trials demonstrated significant reductions in signs and symptoms, with ACR20 response rates of 53% for versus 39% for at week 24, alongside acceptable safety profiles including injection-site reactions but low serious adverse event rates. The FDA approved the biologic license application on December 31, 2002, enabling market entry after approximately three years from IND submission, accelerated by the drug's targeted TNF-alpha inhibition mechanism and unmet need in . In , inavolisib (Itovebi) represents a recent small-molecule success. submitted an on March 27, 2024, following IND-enabled trials targeting PI3Kα-mutated, hormone receptor-positive . Phase 3 data from the INAVO120 study showed a 31% reduction in progression risk when combined with and , with median of 15 months versus 7.3 months for the . Granted due to breakthrough designation, the FDA approved it on October 10, 2024, highlighting how INDs facilitate adaptive trial designs and biomarker-driven development to expedite for precision therapies. These cases underscore causal factors in success: comprehensive preclinical packages minimizing FDA holds during the 30-day review, iterative amendments based on emerging data, and pivotal trials generating robust endpoints like hazard ratios under 0.7 for survival benefits. Aggregate data indicate that while overall phase success rates hover around 10-20% from to approval, targeted modalities in and achieve higher transition rates through specialized pathways, with median clinical timelines of 5-8 years for such approvals when designations like are invoked.

Failed or Delayed INDs

A clinical hold is an order issued by the U.S. (FDA) to an () sponsor to delay a proposed clinical or suspend an ongoing one due to identified deficiencies. Such holds affect approximately 9% of IND applications, with a 2013 pilot study reporting 8.9% placed on hold within 30 days of submission, most resolving to active status within a year. Chemistry, , and controls (CMC) issues represent the most frequent cause, followed by concerns over , , or protocol design. For Phase 1 INDs, holds typically arise from evidence of unreasonable risk based on nonclinical data, such as inadequate studies or dosing plans lacking scientific rationale. In Phases 2 or 3, common triggers include absence of plans to monitor or minimize risks, reliance on flawed prior human data, or failure to provide sufficient information on drug effects in relevant models. delays or rejections often stem from submission errors, such as non-conforming (eCTD) formats or mismatched dosage declarations, which prevent FDA review. Delays can also result from entering inactive status, initiated by the or FDA due to implementation lags, funding shortfalls, or unresolved inquiries; an inactive for five or more years is automatically terminated. In advanced therapy medicinal products like and therapies, hold rates have risen with innovation volume, with 33 holds announced from January 2020 to December 2022, about 15% remaining active by year-end, often linked to complex data gaps or inexperienced . Recovery requires targeted amendments, such as supplemental nonclinical data or protocol revisions, but unresolved holds can escalate costs and timelines by months or years. Notable patterns include cases where sponsors omitted stability data or genetic toxicology assessments, leading to holds and rework; one example involved a company delaying approval by neglecting product documentation entirely. Inadequate trial designs, such as ambiguous endpoints or undersized cohorts, further contribute, signaling to FDA insufficient risk-benefit justification. These failures underscore the need for pre- meetings to preempt issues, as skipping them correlates with higher hold risks. Overall, while most holds lift upon correction, they highlight attrition at the IND stage driven by evidentiary shortfalls rather than outright rejection.

Controversies and Criticisms

Burdens of Regulatory Compliance

The preparation of an application entails extensive documentation, including preclinical safety and efficacy data, chemistry, manufacturing, and controls () information, protocols, and investigator brochures, which collectively impose substantial administrative and scientific burdens on sponsors. This process typically requires 12 to 14 months to complete the IND package, excluding prior preclinical work, due to the need for multidisciplinary coordination and iterative refinements to meet FDA standards. IND-enabling studies, such as and assessments, often extend preparation timelines by an additional 18 months or more, as sponsors must generate robust datasets to mitigate risks of rejection or holds. Financial costs associated with IND compliance are primarily driven by preclinical development, which accounts for about 30% of total per-compound expenses in drug development and can span 3 to 6 years before submission. While the IND filing itself incurs no direct FDA fee, the underlying data generation—encompassing animal studies, formulation development, and quality assurance—frequently totals millions of dollars, with estimates for early-stage preclinical packages ranging from $1 million to over $10 million depending on the modality (e.g., small molecules versus biologics). These outlays strain cash reserves, particularly for startups, where incomplete or inadequately organized submissions risk clinical holds in approximately 9% of cases, requiring costly amendments and delaying trial initiation beyond the mandatory 30-day FDA review period. Small and mid-sized firms face amplified challenges from these requirements, as limited internal expertise often necessitates to organizations, inflating expenses and extending timelines amid FDA constraints like shortages that prolong loops. Such burdens contribute to higher rates for innovators, fostering where smaller entities partner with or are acquired by larger pharmaceutical companies to share compliance loads. Critics, including analyses, argue that the framework's emphasis on exhaustive pre-human data generation prioritizes over expediency, potentially stifling novel therapies for unmet needs by deterring entry-level innovation and prompting some biotechs to explore less stringent regulatory paths abroad. For instance, recent FDA delays have led firms to reassess U.S.-centric strategies, highlighting how procedural rigidities can undermine competitive advantages in .

Trade-offs Between Safety and Innovation

The Investigational New Drug () process requires sponsors to submit preclinical data demonstrating a drug's for trials, with the FDA conducting a 30-day review to identify potential before allowing clinical studies to proceed. This safeguard prevents exposure of trial participants to unacceptably hazardous agents, as reflected in expedited reporting rules that mandate prompt notification of serious adverse events to enable . However, incomplete submissions or unresolved concerns can trigger clinical holds, delaying trial initiation and extending overall development timelines, which averaged 10-15 years from filing to approval as of 2021. Such delays arise from the need for iterative preclinical and studies, increasing upfront costs and deterring investment in novel, high-uncertainty compounds. Empirical analyses link these safety mandates to reduced innovation incentives, particularly under regulatory uncertainty, where firms allocate fewer resources to technologies facing stringent pre-trial scrutiny. For instance, biotechnology companies have increasingly shifted initial trials overseas due to FDA review bottlenecks, with recent organizational disruptions exacerbating hold rates and review lags beyond statutory deadlines. Development costs, exceeding $2.6 billion per approved drug in 2019 estimates, partly stem from compliance with IND safety protocols, contributing to high attrition rates—only about 12% of drugs entering Phase I reach market approval—and a decline in novel molecular entity approvals relative to rising R&D expenditures. Critics contend this framework favors incremental over breakthrough innovations, as smaller sponsors struggle with the financial burden of extensive safety data generation, leading to consolidated pipelines dominated by large pharmaceutical entities. Efforts to balance these trade-offs include FDA programs like Designation, which prioritizes drugs with preliminary evidence of superior while upholding thresholds, potentially shortening by months. Yet, ongoing obligations during trials—requiring sponsors to analyze and report events across studies—can overwhelm resources without proportionally advancing knowledge, as much data proves non-actionable upon . Post-approval revelations of issues in roughly one-third of new drugs validate pre-market caution but underscore that IND-level rigor may overemphasize Type I error avoidance (preventing unsafe trials) at the expense of Type II errors (missing safe, effective therapies), with estimates suggesting regulatory delays cost thousands of lives annually in unmet needs like . Reforms, such as streamlined for low-risk events, aim to preserve while accelerating , though empirical outcomes remain debated amid persistent critiques of overregulation impeding diverse .

Emergency IND Misuse and Ethical Concerns

The emergency provision under U.S. regulations permits the single-patient use of an unapproved drug or biologic in life-threatening situations where no comparable or satisfactory alternative therapy is available, and there is insufficient time to submit a full application. This mechanism, outlined in 21 CFR 312.36, requires prior consultation with the drug sponsor and FDA concurrence, followed by retrospective submission of an within 15 days. While intended to address genuine exigencies, it has raised concerns about misuse, such as invoking the provision for non-emergent cases or when alternatives exist, potentially driven by patient desperation, advocacy, or sponsor interests without robust preclinical or clinical data. Ethical challenges stem primarily from the inherent uncertainties of deploying investigational agents with limited and , exposing vulnerable patients to unknown risks without the safeguards of phased trials. For instance, use of investigational drugs demands adequate scientific substantiation to justify risks, yet contexts often prioritize over comprehensive , heightening the potential for adverse outcomes like unforeseen toxicities. Cases of abuse, though not systematically quantified, have been documented in scenarios involving patients lacking decision-making capacity, where experimental interventions occur without full ethical oversight or prospective , raising questions of and . Further ethical tensions arise from equity issues and systemic pressures: access to emergency INDs disproportionately favors those with medical connections or resources to navigate regulatory hurdles, while broader invocation could divert investigational supplies from ongoing trials, delaying collective knowledge gains. FDA data on programs, which encompass emergency uses, indicate rare reported adverse events—only two instances from 2005 to 2014 led to regulatory actions—but underreporting remains a concern due to voluntary submissions and incomplete follow-up. Critics argue this pathway, if over-relied upon, incentivizes bypassing rigorous testing, potentially eroding public trust in regulatory processes when harms emerge , as seen in historical compassionate use debates where unproven therapies yielded net harm without advancing approvals. Balancing individual against population-level thus requires stringent criteria, including sponsor accountability and independent review, to mitigate misuse while preserving the provision's humanitarian intent.

International Comparisons

Equivalents in Other Jurisdictions

In the , the primary equivalent to the U.S. application is the , which must be submitted to national competent authorities or, since January 31, 2022, through the centralized Clinical Trials Information System (CTIS) under Regulation (EU) No 536/2014. Unlike the product-specific IND, the CTA is protocol-specific and focuses on authorizing individual protocols, with coordinating scientific advice but approvals handled at the member state level. In Canada, requires a Application (CTA) for investigational drugs not authorized for sale in the country, submitted at least 30 days before trial initiation to allow review of safety and ethical considerations. The CTA process aligns with International Council for Harmonisation (ICH) guidelines but emphasizes protocol amendments for ongoing trials, contrasting with the U.S. IND's broader product oversight. Japan's (PMDA) mandates a notification of plans, often termed an application in regulatory contexts, requiring submission of preclinical data, protocols, and manufacturing information in (CTD) format prior to trial commencement. PMDA reviews submissions for safety and ethical compliance, with consultations available for complex cases, though routine notifications may proceed without full approval if no objections arise within 30 days. In , the () employs the Notification (CTN) scheme for most unapproved therapeutic goods, where sponsors notify the agency at least 28 days before supply without requiring prior approval, provided the trial meets predefined risk criteria. Higher-risk trials may necessitate a Approval (CTA) with TGA evaluation, emphasizing institutional ethics review over exhaustive preclinical scrutiny akin to the IND. Post-Brexit in the , the Medicines and Healthcare products Regulatory Agency (MHRA) grants Authorisations (CTA) for investigational medicinal products, requiring submission of protocol, , and quality data, with decisions typically within 30 days under the UK Medicines for Human Use () Regulations 2004. This process mirrors EU standards but operates independently, incorporating mutual recognition for certain low-risk amendments.

Harmonization Efforts and Divergences

The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH), established in 1990, has driven key efforts to standardize requirements for investigational drug applications across major regions, including the , , and , aiming to reduce redundant testing and facilitate global clinical trials. ICH guidelines, such as those in the (CTD) format (e.g., M4Q for quality, M4S for safety), provide a unified structure for submitting preclinical and manufacturing data relevant to investigational new drug () submissions, which the U.S. Food and Drug Administration (FDA) has adopted to streamline reviews. Efficacy guidelines like E6(R3) on (GCP) and E8(R1) on general clinical study considerations further harmonize protocols for trials initiated under IND equivalents, ensuring consistent ethical and scientific standards for safety and during early-phase development. These efforts, informed by among regulators, , and experts, have lowered barriers to multinational trials by aligning data requirements, though full uniformity remains elusive due to regional legal variances. Despite ICH progress, divergences persist in application scope, review processes, and documentation. The FDA's IND is product-specific, authorizing all phases of clinical investigation for a given drug until marketing approval, with a mandatory 30-day silent review period before trials can commence under 21 CFR Part 312. In contrast, the European Medicines Agency (EMA) requires a Clinical Trial Application (CTA) via the Clinical Trials Information System (CTIS) under the 2014 Clinical Trials Regulation, which is protocol-specific and demands separate submissions for each trial amendment or site, emphasizing risk-based assessments but lacking the IND's broad product umbrella. Japan's Pharmaceuticals and Medical Devices Agency (PMDA) mandates a Clinical Trial Notification (CTN) under the 2018 Clinical Trials Act, featuring expedited reviews (often 30 days) and fewer preclinical mandates than the FDA, though it requires prior ethics committee approval and can involve separate regulatory consultations for novel modalities. Chemistry, manufacturing, and controls (CMC) sections also differ: EU Investigational Medicinal Product Dossiers (IMPDs) integrate nonclinical summaries more modularly than FDA INDs, while language requirements vary, with FDA accepting English-only submissions versus multilingual needs in the EU. These discrepancies can impose operational challenges, such as duplicated reviews or staggered trial starts in multi-regional studies, prompting sponsors to leverage FDA data as a for EU IMPDs while adapting to local nuances. Ongoing ICH initiatives, including reflections on real-world , seek to bridge gaps, but entrenched differences in legal frameworks—e.g., FDA's emphasis on comprehensive preclinical versus Japan's conditional approvals—underscore limits to full .

Broader Impact on Drug Development

Economic Costs and Incentives

The preparation and submission of an application entails substantial economic costs for sponsors, primarily driven by the need for comprehensive preclinical data generation, including , , and manufacturing feasibility studies required under 21 CFR Part 312. Preclinical development costs typically range from $15 million to $100 million per candidate, encompassing laboratory testing, , and initial to meet FDA and standards. These expenditures represent a significant barrier, particularly for small firms, as failure to secure IND clearance results in total loss of investment without revenue recovery, contributing to the high attrition rates observed in early pipelines. Regulatory compliance for IND maintenance adds ongoing costs, such as safety reporting, protocol amendments, and clinical hold resolutions, which can extend timelines and inflate outlays by millions annually per program. A analysis estimated pre-launch costs, including IND-enabling activities, ranging from $161 million to $4.54 billion (2019 USD) across successful drugs, with preclinical phases accounting for a disproportionate share when adjusted for risks. Broader studies, such as the Tufts Center's assessment, highlight that capitalized R&D costs per approved exceed $2.6 billion, with IND-stage hurdles amplifying opportunity costs through delayed market entry and capital tie-up, estimated at millions per day in foregone productivity. To counter these disincentives, the FDA provides mechanisms for partial cost recovery, including guidance permitting sponsors to charge for investigational drugs under to recoup direct and expenses, provided they do not exceed reasonable levels or unduly influence participation. Incentives like designation, applicable early in development, offer protocol assistance and potential seven-year market exclusivity upon approval, encouraging submissions for rare diseases despite high upfront risks. Additional programs, such as designation, expedite review and subsequent phases, reducing time-to-market costs, while tax credits under the Orphan Drug Act subsidize up to 50% of clinical testing expenses post-. These measures aim to align economic incentives with needs, though critics argue they disproportionately benefit large incumbents capable of navigating complex filings.

Contributions to Therapeutic Advances

The Investigational New Drug (IND) application serves as the foundational regulatory mechanism enabling sponsors to conduct human clinical trials, generating the safety and efficacy data required for subsequent (NDA) or biologics license application () approvals by the U.S. (FDA). This process has facilitated the approval of novel therapies addressing unmet medical needs, particularly in and rare diseases, by ensuring preclinical findings transition to controlled human testing that validates therapeutic potential. For instance, all FDA-approved novel molecular entities and therapeutic biologics, numbering approximately 50 per year on average from 2018 to 2024, originate from IND-enabled trials that demonstrate clinical benefit while minimizing risks. In , the IND process has accelerated the development of targeted therapies through expedited pathways like Designation (BTD), which builds on initial IND submissions to prioritize review for drugs showing substantial improvement over existing options. , approved in 2013 for , exemplifies this: its IND-led trials provided evidence of superior compared to prior standards, marking the first BTD graduation and influencing subsequent immunotherapies. Similarly, from 2012 onward, BTD has supported dozens of oncology approvals, reducing median IND-to-approval timelines to 74.8 months for such designations versus longer standard paths, thereby hastening access to treatments that extend survival in serious conditions. The IND framework also underpins advances in rare and infectious diseases, as seen with elexacaftor-based triple (Trikafta) for , where IND submission in December 2016 led to approval in October 2019 after trials confirming improved lung function in patients with specific mutations. During the , Pfizer-BioNTech's underwent IND submission (IND 19736) to enable large-scale phase 3 trials, culminating in and full approval based on data from over 40,000 participants, significantly reducing severe outcomes and hospitalizations. These cases illustrate how IND-mandated drives evidence-based refinements, contributing to therapies that enhance survival and across diverse therapeutic areas.

References

  1. [1]
    Investigational New Drug (IND) Application
    ### Summary of Investigational New Drug (IND) Application
  2. [2]
    Investigational New Drug: Key Milestones & Challenges - Allucent
    The Investigational New Drug (IND) application is the primary pathway through which new drugs and biologics are approved to be tested in humans.
  3. [3]
    Four Top Roadblocks to Successful IND Applications
    One such process is the Investigational New Drug (IND) application with the Food and Drug Administration (FDA). The IND application must include information ...
  4. [4]
    21 CFR 312.3 -- Definitions and interpretations. - eCFR
    IND means an investigational new drug application. For purposes of this part, “IND” is synonymous with “Notice of Claimed Investigational Exemption for a New ...
  5. [5]
    Drug Development and Review Definitions - FDA
    Aug 20, 2015 · Investigational New Drug Application​​ During a new drug's early preclinical development, the sponsor's primary goal is to determine if the ...
  6. [6]
    [PDF] Investigational New Drug Applications (INDs) - FDA
    Human Research Without an Investigational New Drug Application, which is intended to clarify whether research using a radioactive drug must be conducted under ...Missing: controversies | Show results with:controversies
  7. [7]
    21 CFR 312.23 -- IND content and format. - eCFR
    A sponsor who intends to conduct a clinical investigation subject to this part shall submit an “Investigational New Drug Application” (IND) including, in the ...Missing: preclinical | Show results with:preclinical
  8. [8]
    21 CFR Part 312 -- Investigational New Drug Application - eCFR
    This part contains procedures and requirements governing the use of investigational new drugs, including procedures and requirements for the submission to, and ...
  9. [9]
    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.
  10. [10]
    Drug Development & Approval Process
    ### Summary of Drug Development Process
  11. [11]
    Milestones in US Food and Drug Law - FDA
    Jan 30, 2023 · The following chronology describes some of the milestones in the history of food and drug regulation in the United States.
  12. [12]
    The History of Drug Regulation | FDA
    Feb 1, 2018 · Federal regulation of drugs emerged as early as 1848, under a law that addressed only imported drugs.
  13. [13]
    [PDF] FDA and Clinical Drug Trials: A Short History
    The 1962 [Kefauver-Harris] Drug Amendments and the 1963 investigational drug ... The acronym IND (Investigational New Drug) was coined to parallel the acronym.
  14. [14]
    Federal Food, Drug, and Cosmetic Act (FD&C Act) - FDA
    Mar 29, 2018 · The Federal Food, Drug, and Cosmetic Act and subsequent amending statutes are codified into Title 21 Chapter 9 of the United States Code.Drugs and Devices · FD&C Act Chapter IV: Food · Short Title and Definitions
  15. [15]
    21 U.S. Code § 355 - New drugs | LII / Legal Information Institute
    No person shall introduce or deliver for introduction into interstate commerce any new drug, unless an approval of an application filed pursuant to subsection ...Section 355a of this title · Risk evaluation and mitigation... · 21 U.S.C. 355g
  16. [16]
    Promoting Safe & Effective Drugs for 100 Years - FDA
    Apr 23, 2019 · "Also critically, the 1962 amendments required that the FDA specifically approve the marketing application before the drug could be marketed, ...The Kefauver-Harris Drug... · Stricter Labeling... · The Orphan Drug Act
  17. [17]
    Reform, Regulation, and Pharmaceuticals — The Kefauver–Harris ...
    in the form of “adequate and well-controlled investigations”— before ...
  18. [18]
    A Brief History of the Center for Drug Evaluation and Research - FDA
    Jan 31, 2018 · In the wake of the new law, the Division of New Drugs was restructured into five branches in 1962. The Investigational Drug Branch, directed by ...
  19. [19]
    The FDA's Evolving Approach to Approving Drugs
    May 26, 2022 · These new laws mandated that pre-clinical trial notification be given through an Investigational New Drug (IND) application and that phased ...The Standard Development... · Adaptive Trials · Summary
  20. [20]
    Research Investigational New Drug Applications – What You ... - FDA
    Jun 25, 2019 · The IND application goes into effect 30 days after FDA receives the application, unless FDA notifies the sponsor that the investigations ...
  21. [21]
    Subpart D—Responsibilities of Sponsors and Investigators - eCFR
    (a) The sponsor shall monitor the progress of all clinical investigations being conducted under its IND. (b) A sponsor who discovers that an investigator is not ...312.50 – 312.70 · 21 CFR 312.62 · 21 CFR 312.57 · 312.64 Investigator reports.
  22. [22]
    Information for Sponsor-Investigators Submitting INDs - FDA
    Jun 27, 2017 · IND regulations are contained in Title 21, Code of Federal Regulations, Part 312. Copies of the regulations, further guidance regarding IND ...
  23. [23]
    Investigational New Drug Applications Prepared and Submitted by ...
    Aug 24, 2018 · The purpose of this guidance is to assist sponsor-investigators in preparing and submitting complete investigational new drug applications (INDs).
  24. [24]
    Emergency Use of an Investigational Drug or Biologic - FDA
    Mar 1, 2024 · The emergency use provision in the FDA regulations [21 CFR 56.104(c)] is an exemption from prior review and approval by the IRB. The exemption, ...
  25. [25]
    Emergency IND Timeline - FDA
    Feb 20, 2018 · Information is intended to provide an overview of timelines applicable to physicians who plan to submit or have submitted individual patient ...
  26. [26]
    [PDF] Content and Format of INDs for Phase 1 Studies of Drugs, Inclu - FDA
    Although in each phase of the investigation sufficient information should be submitted to assure the proper identification, quality, purity, and strength of the ...
  27. [27]
  28. [28]
    [PDF] Formal Meetings Between the FDA and Sponsors or Applicants
    This guidance provides recommendations to industry on formal meetings between the Food and. Drug Administration (FDA) and sponsors or applicants ...
  29. [29]
    Frequently Asked Questions on the Pre-Investigational New Drug ...
    Feb 22, 2022 · A pre-IND meeting can also provide sponsors information that will assist them in preparing to submit complete investigational new drug ...
  30. [30]
    Beginner's Guide to Pre-IND Meetings - Advarra
    May 12, 2022 · For a pre-IND meeting, there are three meeting formats: face-to-face, teleconference, or written response only (WRO). However, due to the FDA's ...
  31. [31]
    OTP Pre-IND Meetings - FDA
    Aug 22, 2024 · The primary purpose of this meeting is to review and obtain feedback on the design of preclinical studies, the design of the initial IND study, and product ...
  32. [32]
    [PDF] Regulatory Interactions: CBER-CDER Pre-IND Meetings - NIH's Seed
    Pre-IND meetings are for innovators to request feedback from FDA before submitting an IND application, sharing early development work and receiving direct ...
  33. [33]
    IND Applications for Clinical Investigations: Regulatory and Administr
    Mar 7, 2022 · The following table includes explanations of various components of an IND application and links to additional information related to application submission.
  34. [34]
    IND Forms and Instructions - FDA
    Mar 31, 2022 · Form FDA 1571 (PDF - 221KB): Investigational New Drug Application (IND) · Form FDA 1572 (PDF - 208KB): Statement of Investigator · Form FDA 3674 ( ...
  35. [35]
    IND Application Procedures: Overview - FDA
    Oct 9, 2015 · Once an IND application is in effect, a drug manufacturer may ship the investigational new drug to the investigator(s) named in the application.
  36. [36]
  37. [37]
    How the IND 30-day Hold Impacts Clinical Trial Activities - Advarra
    Sep 22, 2022 · When Does an IND go into Effect? · 30 days after the FDA receives the application, unless the FDA notifies the sponsor the investigations ...
  38. [38]
    Timeline of regulatory review process. After FDA IND submission, the...
    After FDA IND submission, the FDA has 30 days to review the application and provide any concerns that must be addressed. The IRB submission process is similar, ...
  39. [39]
    [PDF] Guidance for Industry - FDA
    Clinical Hold: An order issued by FDA to the sponsor of an IND to delay or to suspend a clinical investigation for reasons described in 21 CFR 312.42. A ...
  40. [40]
    21 CFR 312.42 -- Clinical holds and requests for modification. - eCFR
    The clinical hold order may apply to one or more of the investigations covered by an IND. When a proposed study is placed on clinical hold, subjects may not be ...
  41. [41]
    Why Does the FDA Place Clinical Holds on Investigational New ...
    Dec 6, 2016 · INDs are placed on hold to further assess the quality of the investigational product to ensure the safety of trial participants. If an ...
  42. [42]
    Submitting and Reviewing Complete Responses to Clinical Holds ...
    May 5, 2020 · A clinical hold is an order issued by the FDA to the applicant to delay a proposed clinical investigation or to suspend an ongoing investigation ...
  43. [43]
    5 Tips to Avoid an IND Clinical Hold: Regulatory Guidance and ...
    Oct 12, 2020 · A clinical hold from the U.S. Food and Drug Administration can significantly prolong the time and increase the cost of drug development.
  44. [44]
    IND Applications for Clinical Investigations: Pharmacology ... - FDA
    Feb 16, 2016 · This component of an IND application is expected to contain information about pharmacological and toxicological (laboratory animals or in vitro) studies.
  45. [45]
    [PDF] IND content and format for Phase 1 studies - FDA
    The regulations are not specific as to the nature of the report of toxicology data needed in an IND submission and the nature of the study reports upon ...
  46. [46]
    Exploratory IND Studies | FDA
    Aug 24, 2018 · This guidance is intended to clarify what preclinical and clinical approaches, as well as chemistry, manufacturing, and controls information ...
  47. [47]
    Step 2: Preclinical Research - FDA
    Jan 4, 2018 · FDA requires researchers to use good laboratory practices (GLP), defined in medical product development regulations, for preclinical laboratory ...
  48. [48]
    IND Applications for Clinical Investigations: Chemistry, Manufacturing
    Feb 25, 2022 · This component of an IND application includes the Chemistry, Manufacturing, and Control information for: (1) drug substance; (2) drug product; (3) placebo ...
  49. [49]
    [PDF] Chemistry, Manufacturing, and Control Information for Human Gene ...
    FDA may place the IND on clinical hold if the IND does not contain sufficient CMC information to assess the risks to subjects in the proposed studies (21 CFR ...
  50. [50]
    INDs for Phase 2 and Phase 3 Studies CMC Information - FDA
    Feb 22, 2018 · This guidance provides recommendations to sponsors of investigational new drug applications (INDs) on the chemistry, manufacturing, and controls (CMC) ...
  51. [51]
    [PDF] Chemistry, Manufacturing, and Controls; Requirements for Early ...
    IND CMC Regulatory Requirements. 21 CFR 312.23(a)(7). • (7) Chemistry, manufacturing, and control information. (i) As appropriate for the particular ...
  52. [52]
    IND Applications for Clinical Investigations: Clinical Protocols - FDA
    Oct 9, 2015 · Clinical protocols must include safety elements, trial design, patient selection, procedures, lab tests, and measures to monitor drug effects. ...
  53. [53]
    Step 3: Clinical Research - FDA
    Jan 4, 2018 · Drug developers, or sponsors, must submit an Investigational New Drug (IND) application to FDA before beginning clinical research. In the ...
  54. [54]
    [PDF] INDs for Phase 2 and Phase 3 Studies - FDA
    FDA reviews the safety information to determine whether a clinical hold on the IND is warranted. A summary of CMC safety information submitted during a subject- ...<|control11|><|separator|>
  55. [55]
    [PDF] Clinical Review of Investigational New Drug Applications - FDA
    This FDA document covers clinical review of INDs, including pre-IND meetings, Phase 1 trials, end-of-Phase 2/3 planning, controlled trials, and IND safety ...
  56. [56]
    [PDF] Master Protocols: Efficient Clinical Trial Design Strategies to ... - FDA
    Master protocols are efficient clinical trial design strategies to expedite development of oncology drugs and biologics. They include single drug or ...<|separator|>
  57. [57]
    [PDF] Expansion Cohorts: Use in First-in-Human Clinical Trials to Expedite ...
    trials is expected to meet the level of detail appropriate for the stage of clinical development.19. 16 See the guidance for industry Investigational In ...
  58. [58]
    21 CFR 312.30 -- Protocol amendments. - eCFR
    This section sets forth the provisions under which new protocols may be submitted and changes in previously submitted protocols may be made.
  59. [59]
    21 CFR 312.32 -- IND safety reporting. - eCFR
    In each IND safety report, the sponsor must identify all IND safety reports previously submitted to FDA concerning a similar suspected adverse reaction, and ...
  60. [60]
    Safety Reporting Requirements for INDs and BA/BE Studies - FDA
    This document provides guidance to sponsors and investigators on expedited safety reporting requirements for human drug and biological products.
  61. [61]
    FDA shifts IND safety reporting over to FAERS in finalized guidance
    Mar 29, 2024 · After 1 April 2026, sponsors will have to submit investigational new drug (IND) safety reports for serious and unexpected suspected adverse events to FDA's ...
  62. [62]
    IND Application Reporting: Protocol Amendments - FDA
    Dec 23, 2015 · Sponsors are expected to submit protocol amendments for new protocols or changes to existing protocols before implementation of the respective changes.
  63. [63]
    IND Application Reporting: Information Amendments - FDA
    Oct 8, 2015 · Information amendments to an IND application should be submitted as necessary but, to the extent feasible, not more than every 30 days.
  64. [64]
    a 1-year pilot study on rates and reasons for clinical hold - PubMed
    The vast majority of initial INDs moved forward within 30 days after submission, and for those applications placed on hold, most became active within 1 year ...
  65. [65]
    A 1-Year Pilot Study on Rates and Reasons for Clinical Hold
    Mar 13, 2023 · ... 1500 initial Investigational New Drug applications (INDs) per year. In the first 30 days after initial IND submission, FDA conducts a review ...
  66. [66]
    ATMP innovation on the rise, sponsors see increase FDA IND holds
    Data from the Food and Drug Administration (FDA) show that the Center for Biologics Evaluation and Research (CBER) received 777 original IND applications in ...
  67. [67]
    How will FDA changes reshape drug approval in 2025 and beyond?
    May 14, 2025 · ... (IND) for FDA review. The document details what a drug is and ... The agency receives between 1,500 and 2,000 INDs each year. The data ...Missing: per | Show results with:per
  68. [68]
    Submission Statistics - FDA
    Oct 3, 2025 · FDA ESG 2014-2025 Total Submission Statistics. 2014, 2015, 2016, 2017, 2018, 2019, 2020, 2021, 2022, 2023, 2024, 2025, Totals ...Missing: annual volumes
  69. [69]
    Key trends in IND applications - Cardinal Health
    Current trends. FDA receives, on average, 1500 new INDs per year2. For a timely review, IND sponsors should ensure that their applications are ...
  70. [70]
    Parsing clinical success rates | Nature Reviews Drug Discovery
    Jun 30, 2016 · They found that the probability of success was 63% in Phase I trials, 31% in Phase II trials, 58% in Phase III trials and 85% during the ...
  71. [71]
    Measuring the risks and rewards of drug development - MIT Sloan
    Jan 31, 2018 · A new study by researchers at the Massachusetts Institute of Technology found that nearly 14% of all drugs in clinical trials eventually win approval from the ...
  72. [72]
    [PDF] Clinical Development Success Rates and Contributing Factors 2011 ...
    The Phase I transition success rate was 52.0% (n=4,414). As this phase is typically conducted for safety testing and is not dependent on efficacy for ...
  73. [73]
    Why 90% of clinical drug development fails and how to improve it?
    If drug candidates in the preclinical stage are also counted, the failure rate of drug discovery/development is even higher than 90%. Analyses of clinical trial ...Missing: IND | Show results with:IND
  74. [74]
    Estimating the Probability of Regulatory Registration Success - RAPS
    Feb 13, 2020 · Large molecule Phase 1 registration success rate was 21%. Comparatively, small molecule Phase 1 registration success rate was 10%. Rare disease ...
  75. [75]
    Approval success rates of drug candidates based on target, action ...
    The current success rate of a drug candidate, from the beginning of the clinical trial to receiving marketing approval, is about 10%–20%, and it has not ...
  76. [76]
    [PDF] BLA12505710 Humira Approval - accessdata.fda.gov
    Study was filed to BB-IND on December 1999 and enrollment was completed on August 31, 2000. The study will be completed by August 31, 2005. A study report with ...
  77. [77]
    Humira (adalimumab) FDA Approval History - Drugs.com
    Aug 25, 2022 · Development timeline for Humira ; Jul 31, 2006, Approval Humira Receives FDA Approval for Treatment of Ankylosing Spondylitis ; Oct 4, 2005 ...
  78. [78]
    [PDF] 219249Orig1s000 - accessdata.fda.gov
    Application Type NDA. Application Number(s) 219249. Priority or Standard Priority. Submit Date(s) March 27, 2024. Received Date(s) March 27, 2024.
  79. [79]
    FDA approves inavolisib with palbociclib and fulvestrant for endocrine
    Oct 10, 2024 · On October 10, 2024, the Food and Drug Administration approved inavolisib (Itovebi, Genentech, Inc.) with palbociclib and fulvestrant for ...Missing: IND | Show results with:IND
  80. [80]
    Clinical development times for innovative drugs - PMC - NIH
    Drugs with the accelerated approval designation have a reduced clinical development time of 1,100 days (95% CI = 563–1637 days). Breakthrough designation was ...
  81. [81]
    Achieving end-to-end success in the clinic: Pfizer's learnings on ...
    By the end of 2020, Pfizer had achieved an end-to-end [First-in-Human (FIH) to regulatory approval] clinical success rate of 21% (Fig. 1a), with the interim ...
  82. [82]
    IND Application Procedures: Clinical Hold - FDA
    Oct 9, 2015 · A clinical hold is an order issued by FDA to the sponsor of an IND application to delay a proposed clinical investigation or to suspend an ongoing ...
  83. [83]
    Human Drug Common Entry Errors | FDA
    Dec 11, 2024 · Common Human Drug Look-up Errors that Result in a Failed Look-up ; Dosage format error, The dosage strength declared on the entry does not match ...
  84. [84]
    Common Problems to Avoid with IND Applications for New Drugs ...
    Let's look at a number of common problems with IND submissions to help your organization avoid these mistakes and get your product to market on time.
  85. [85]
    Understanding FDA Regulatory Requirements for Investigational ...
    This notification is called an Investigational New Drug (IND) application. For drug trials conducted by the pharmaceutical industry or other commercial sponsors ...Missing: controversies | Show results with:controversies
  86. [86]
    Clinical holds for cell and gene therapy trials - NIH
    A total of 33 clinical holds announced from January 2020 to December 2022 were identified. Of these holds, 5 (∼15%) were still active at the end of 2022, and 26 ...
  87. [87]
    IND Clinical Hold: Avoidance and Recovery – A Nonclinical ... - Freyr.
    May 20, 2025 · An Investigational New Drug (IND) application clinical hold can significantly disrupt drug development, leading to delays and increased costs.<|separator|>
  88. [88]
  89. [89]
    Common Pitfalls in Preparing an IND Application - Advarra
    Jun 28, 2022 · Including unnecessary information, disorganized data, or dense text are common missteps when sponsors submit IND applications.
  90. [90]
    IND-Enabling Studies - Charles River Laboratories
    Early planning is crucial as it can take 18 months to prepare IND-enabling studies and manufacturing. Don't wait for a quote – start by understanding your ...
  91. [91]
    Preclinical Development: The Safety Hurdle Prior to Human Trials
    Apr 30, 2016 · On average, drug discovery and preclinical development take three to six years and account for 30.8% of costs per approved compound, ...
  92. [92]
    Biotechs Report Delays, Denied Meeting Requests Amid Staff Cuts
    leaving biotechs facing costly delays, ...
  93. [93]
    FDA Delays Push Biotech Companies to Rethink U.S. Drug ... - Veristat
    Recent FDA organizational disruption, missed review deadlines, and uncertainty around regulatory processes have lead some drugmakers to seriously weigh options ...
  94. [94]
    [PDF] Safety Reporting Requirements for INDs and BA/BE Studies | FDA
    IND Annual Reports and Labeling. The IND safety reporting requirements did not make any changes to the requirements for IND annual reports (21 CFR 312.33) ...
  95. [95]
    The FDA's Final Rule on Expedited Safety Reporting - NIH
    A Final Rule for expedited reporting of serious adverse events took effect in the United States for studies conducted under an Investigational New Drug (IND) ...
  96. [96]
    Research and Development in the Pharmaceutical Industry
    Apr 8, 2021 · On average, the Food and Drug Administration (FDA) approved 38 new drugs per year from 2010 through 2019 (with a peak of 59 in 2018), which is ...
  97. [97]
    Innovation under Regulatory Uncertainty: Evidence from Medical ...
    This paper explores how the regulatory approval process affects innovation incentives in medical technologies.
  98. [98]
    Innovation crisis in the pharmaceutical industry? A survey
    Nov 11, 2021 · The empirical evidence shows that project success rates decreased over time, while attrition rates, development times, and the costs per new drug increased.
  99. [99]
    [PDF] The FDA's Breakthrough Therapy Designation
    Second, regulators face a key trade-off that require striking a balance between moving a new product to market quickly and gathering additional infor- mation ...
  100. [100]
    Optimizing Expedited Safety Reporting for Drugs and Biologics ...
    The IND safety reporting rule implicitly requires the sponsor to review safety data collected across all completed and ongoing studies in an IND, analyze these ...<|control11|><|separator|>
  101. [101]
    One-Third Of New Drugs Had Safety Problems After FDA Approval
    May 9, 2017 · Taking Shortcuts In Drug Testing Can Put Patients At Risk. Drugs ushered through the FDA's accelerated approval process were among those that ...
  102. [102]
    Burying Sites Under Safety Reporting | Applied Clinical Trials Online
    A look at the vague but time consuming requirements imposed on sites and efforts to ease the burden.
  103. [103]
    Emergency Use and Compassionate Use of Experimental Drugs ...
    The emergency use provision in federal regulations allows physicians restricted access to investigational treatments that would be otherwise off-limits.Missing: misuse concerns
  104. [104]
    [PDF] Ethical Consideratons for Clinical Investigations of Medical Products ...
    Sep 19, 2022 · This guidance describes the FDA's current thinking regarding ethical considerations. 18 for clinical investigations of medical products in ...Missing: misuse | Show results with:misuse
  105. [105]
    Ethics framework for treatment use of investigational drugs
    Nov 18, 2020 · To be ethical, the treatment use of IDs must be based on adequate scientific evidence. Otherwise, the patient is exposed to substantial risks ...Missing: misuse | Show results with:misuse
  106. [106]
    [PDF] INVESTIGATIONAL NEW DRUGS FDA's Expanded Access Program
    Oct 3, 2017 · For example, according to a study using FDA data, there were only two instances from 2005 through 2014 in which adverse events from expanded ...
  107. [107]
    Understanding the challenges and ethical aspects of compassionate ...
    Aug 4, 2020 · Compassionate drug use is an important regulatory provision that is being proposed as an amendment to the New Drugs and Clinical Trial Rules, 2019.
  108. [108]
    Clinical trials in human medicines
    Although clinical trials are authorised at national level in the European Union (EU), EMA plays a key role in further developing the EU as a competitive centre ...Trials Regulation · Clinical Trials Information · ACT EU · Development of CTIS
  109. [109]
    Navigating the Global Clinical Landscape: CTA or IND for Your First ...
    Mar 7, 2024 · One primary distinction to note between a CTA and an IND submission is that the CTA is protocol-specific, while the IND is product-specific.
  110. [110]
    Filing of clinical trials Frequently Asked Questions - Canada.ca
    May 4, 2017 · It is recommended that amendments be filed for trials exempted from IND filing with a marketed drug for a new indication and initiated by a medical ...
  111. [111]
    [PDF] Comparing the Canadian CTA to the U.S. IND Submission Process
    Protocol-by-protocol approval. Principle: One open Investigational New Drug (IND) throughout drug development. New protocols submitted as amendments.
  112. [112]
    IND and NDA Regulatory Submissions in Japan- Decoded - Freyr.
    May 3, 2016 · Japan's regulatory system demands the IND Application documents to be prepared in the Common Technical Document (CTD) format.
  113. [113]
    Consultations | Pharmaceuticals and Medical Devices Agency
    PMDA offers consultations to give guidance and advice on clinical trials of drugs, medical devices, and cellular and tissue-based products as well as on data ...
  114. [114]
    Review of the Clinical Trial Approval (CTA) Scheme
    Sep 1, 2024 · The CTA scheme involves an application to the TGA for approval to supply an 'unapproved' therapeutic good(s) in a clinical trial.Missing: IND | Show results with:IND
  115. [115]
    Regulatory requirements for clinical trials: Australia vs the US
    Feb 21, 2020 · An IND submission is not necessary to initiate first-in-human clinical trials in Australia. The simplicity and efficiency of the Australian and ...
  116. [116]
    Clinical trials for medicines: apply for authorisation in the UK - GOV.UK
    How to apply for a clinical trial including eligibility, phases, model IMPDs, costs and how to make changes to your application.
  117. [117]
    Clinical Research Regulation For United Kingdom and ... - ClinRegs
    The MHRA grants permission for clinical trials to be conducted in the UK in accordance with the MHCTR and the MHCTR2006. MHRA-More states that MHRA regulates ...
  118. [118]
    ICH Official web site : ICH
    ICH's mission is to achieve greater harmonisation worldwide to ensure that safe, effective and high quality medicines are developed, and registered and ...Quality Guidelines · Q8/9/10 Training Material · ESTRI page for eCTD V3.2.2 · CTD
  119. [119]
    ICH Guidance Documents - FDA
    Dec 11, 2024 · This International Conference on Harmonization (ICH) document makes recommendations on information that should be included in a core clinical study report.
  120. [120]
    Efficacy Guidelines - ICH
    The ICH harmonised Guideline was finalised under Step 4 in October 1994. This document gives recommendations on the numbers of patients and duration of exposure ...
  121. [121]
    Comparing Clinical Trial Regulations: USA vs. APAC
    Jul 8, 2024 · Japan has comparatively fewer regulations than the U.S. FDA. The Clinical Trials Act of 2018 in Japan created a streamlined and time-saving ...
  122. [122]
    Regulatory and operational challenges in conducting Asian ...
    Differences in regulatory and operational procedures were identified while coordinating the trial. In Japan, regulatory authority reviews should be performed ...
  123. [123]
    [PDF] The CMC section of an EU IMPD: | PPD
    During the last five years, the EMA has organised workshops focused on quality requirements for medicines containing chemical entities3 and biopharmaceuticals4.Missing: Union | Show results with:Union
  124. [124]
    [PDF] ich-reflection-paper-pursuing-opportunities-harmonisation-using ...
    Jul 25, 2024 · Applying different definitions may complicate efforts among regulators to track such data and evidence, causing potential confusion during ...
  125. [125]
    The Staggering Cost of Drug Development: A Look at the Numbers
    Aug 10, 2023 · Preclinical Phase: Costs here, primarily due to laboratory studies and animal testing, can range from $15 million to $100 million. Clinical ...
  126. [126]
    How Much Does It Cost to Research and Develop a New Drug? A ...
    Nov 17, 2021 · Pre-launch R&D costs for a new drug range from $161 million to $4.54 billion (2019 US$), with no universally correct answer.
  127. [127]
    Tufts Center for the Study of Drug Development - Cost of Developing ...
    Nov 18, 2014 · Cost to Develop and Win Marketing Approval for a New Drug Is $2.6 Billion, According to the Tufts Center for the Study of Drug Development.
  128. [128]
    [PDF] Charging for Investigational Drugs Under IND - FDA
    Does FDA consider the financial resources available to a sponsor when determining whether the cost of providing its investigational drug in a clinical trial is.
  129. [129]
    Economic Assistance and Incentives for Drug Development - FDA
    Feb 4, 2020 · Economic assistance is available in the pre-approval period of drug development for the conduct of clinical trials. Economic incentives are granted post- ...
  130. [130]
    The Pathway from Idea to Regulatory Approval: Examples for Drug ...
    BOX E-1. Case Example of Successful Collaboration in Drug Discovery and Development. In 2002, the biotechnology company Sugen and the Salk Institute published ...
  131. [131]
    Novel Drug Approvals at FDA
    Feb 6, 2025 · Each year, CDER approves a wide range of new drugs and biological products: Some of these products have never been used in clinical practice.
  132. [132]
    The Pharmaceutical Industry in 2023: An Analysis of FDA Drug ...
    The US Food and Drug Administration (FDA) has approved 55 new drugs in 2023, a figure consistent with the number authorized in the last five years (53 per year ...
  133. [133]
    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.
  134. [134]
    Learning from the 2012–2013 class of breakthrough therapies - Nature
    Nov 29, 2013 · Less than a year on, on 1 November, Roche and Genentech's obinutuzumab became the first drug to graduate from the breakthrough therapy programme ...
  135. [135]
    An overview of cancer drugs approved through expedited approval ...
    This review summarizes the various expedited approval programs and orphan drug designations that are used across the globe, as well as the anticancer drugs that ...
  136. [136]
    [PDF] Pfizer COVID-19 Vaccine (2024-2025 Formula) Letter of Authorization
    Aug 22, 2024 · Pfizer Inc. must submit to Investigational New Drug application (IND) number. 19736 periodic safety reports monthly, or at another ...
  137. [137]
    [PDF] Electronic Submission - Regulations.gov
    Jan 19, 2022 · ... covid-19 vaccines submitted an Investigational New. Drug application (IND), thus subject to federal regulation (investigational product.
  138. [138]
    Exploring the Impact of IND in Medical Research - bioaccess
    In conclusion, the Investigational New Drug (IND) process is a crucial step in advancing medical science and improving patient outcomes. IND status allows for ...