Fact-checked by Grok 2 weeks ago

Drug development

Drug development is the integrated process of advancing potential therapeutic compounds from initial identification through rigorous testing and regulatory approval to market availability for treating human diseases. This multidisciplinary endeavor typically spans 10 to 15 years and incurs average costs exceeding $2.6 billion per successful drug, accounting for the expenses of numerous failed candidates. Key stages include discovery and preclinical research to identify and refine lead compounds, followed by phased clinical trials assessing safety and efficacy in human volunteers, and culminating in regulatory scrutiny by agencies like the U.S. Food and Drug Administration (FDA). Despite these structured phases, approximately 90% of drug candidates fail during development, primarily due to insufficient efficacy, unexpected toxicity, or strategic misalignments, underscoring the inherent risks and high attrition rates that define the field's challenges. Landmark successes, such as antibiotics and vaccines, highlight the process's potential to eradicate scourges like infectious diseases, yet persistent controversies revolve around escalating costs, regulatory delays, and debates over whether failure rates reflect overly stringent safety standards or inefficiencies in early-stage prediction models. Post-approval monitoring further ensures ongoing safety, though it reveals rare but critical adverse effects that necessitate withdrawals or label updates.

Historical Context

Pre-Modern Foundations

Early pharmacological practices originated in ancient civilizations through empirical observation and trial-and-error application of natural substances for therapeutic effects. In , around 2400 BC, the earliest known written prescriptions were inscribed on clay tablets, detailing remedies derived from plants, minerals, and animal products to treat ailments such as infections and gastrointestinal issues. Similarly, ancient Egyptian texts like the (circa 1550 BC) documented over 700 herbal formulations, including for pain relief and as a purgative, reflecting systematic recording of observed despite lacking mechanistic understanding. In parallel, and traditions, as recorded in texts like the Shennong Bencao Jing (circa 200-250 AD) and the (circa 300-200 BC), emphasized plant-based medicines—such as for vitality and for inflammation—integrated with holistic philosophies but grounded in generations of accumulated experiential data. These foundations evolved through Greco-Roman and medieval periods, where apothecaries and healers refined herbal preparations via distillation and compounding, often blending empirical remedies with rudimentary chemical processes inherited from . Greek physicians like (circa 460-370 BC) advocated rational observation over superstition, classifying drugs by effects such as emetics or diuretics, while Dioscorides' (circa 50-70 AD) cataloged over 600 plant-derived substances, influencing pharmacopeias for centuries. In the (8th-13th centuries), scholars like advanced by standardizing drug preparation and testing purity, drawing on translated ancient knowledge to produce refined extracts like those from and senna, which were traded globally. apothecaries in the period (15th-17th centuries) further formalized these practices, establishing guilds and early pharmacopeias, such as the 1498 edition, which listed standardized herbal recipes amid the shift from mystical to proto-chemistry. The pre-modern era culminated in the late 18th and early 19th centuries with the chemical isolation of pure active principles from natural sources, enabling more precise dosing and laying groundwork for synthetic drug development. In 1804, isolated from , confirming its role as the primary component and introducing chemistry to . This was followed by quinine's extraction from in 1820 by Pelletier and Caventou, revolutionizing treatment by providing a concentrated antimalarial agent over crude bark infusions. Other key isolations included (1819), (1818), and (1817), primarily by European chemists applying emerging analytical techniques to traditional remedies, thus bridging empirical herbalism with scientific despite high failure rates in early purity assessments. These advancements, driven by from bioassays on animals and humans, underscored the limitations of pre-modern methods—reliant on and lacking controlled validation—but established the evidentiary core of drug through verifiable active compounds.

20th-Century Milestones and Regulatory Foundations

The marked a transition from empirical remedies to systematic drug development, catalyzed by pivotal discoveries and regulatory reforms that prioritized and . In 1906, the enacted the , the first federal legislation prohibiting interstate commerce of misbranded or adulterated drugs and foods, though it lacked pre-market approval requirements. This act, signed by President amid public outcry over unsafe products exposed by Upton Sinclair's , established the Bureau of Chemistry—precursor to the FDA—to enforce labeling standards but did little to prevent untested drugs from reaching markets. Major therapeutic breakthroughs underscored the need for oversight. Insulin's isolation in 1921 by and Charles Best revolutionized treatment, with clinical use beginning in 1922 after rapid testing in humans. The 1928 discovery of penicillin by laid groundwork for antibiotics, though scalable production occurred during via and Ernst Chain's efforts, saving countless lives from bacterial infections. Sulfonamides, introduced in the 1930s starting with in 1935, represented the first synthetic antibacterials effective against streptococcal infections, reducing mortality from diseases like puerperal fever. Tragedies prompted stricter regulations. The 1937 Elixir Sulfanilamide disaster, where diethylene glycol solvent killed over 100 consumers due to untested toxicity, led to the 1938 Federal Food, Drug, and Cosmetic Act, mandating manufacturers prove drug safety via animal and limited human studies before marketing and granting FDA authority over new drugs. Post-World War II ethical concerns from Nazi experiments birthed the 1947 Nuremberg Code, establishing voluntary consent and risk minimization in human trials as international standards. The 1962 Kefauver-Harris Amendments, enacted after thalidomide's association with thousands of birth defects in Europe (averted in the US by FDA reviewer Frances Kelsey), required substantial evidence of efficacy from adequate, well-controlled clinical investigations, formalized informed consent, and mandated adverse event reporting. These reforms entrenched the phased clinical trial structure—Phase I for safety in small groups, Phase II for efficacy in patients, and Phase III for confirmation in larger populations—alongside New Drug Application reviews, balancing innovation with causal accountability for harms. By century's end, these foundations enabled antibiotics proliferation, chemotherapy agents from wartime mustard gas research in the 1940s, and vaccines like Jonas Salk's polio vaccine in 1955, which eradicated epidemics through rigorous testing.

Core Development Pipeline

Target Identification and Lead Discovery

Target identification in drug development entails the systematic selection of biomolecules, primarily proteins such as enzymes, receptors, or ion channels, whose modulation is hypothesized to alleviate disease symptoms or halt progression. This phase draws on empirical evidence from disease biology, prioritizing targets with demonstrated causal roles via genetic, biochemical, or phenotypic data to enhance downstream success probabilities. Drugs targeting proteins supported by human genetic evidence, such as those identified through genome-wide association studies (GWAS), exhibit a 2.6-fold higher likelihood of clinical approval compared to those lacking such validation. Computational approaches, including and models trained on multi-omic datasets, further refine candidate selection by predicting target-disease associations, though experimental confirmation remains essential to mitigate false positives inherent in predictive algorithms.00137-2) Key methodologies for target identification encompass experimental strategies like affinity-based proteomics and phenotypic screening, where compounds induce observable cellular changes traced back to specific proteins via pull-down assays or CRISPR interference. Multi-omic integration—combining genomics, transcriptomics, and proteomics—uncovers dysregulated pathways, as exemplified by the identification of PCSK9 as a target for hypercholesterolemia through GWAS linking rare variants to low LDL levels. Validation involves orthogonal assays, such as knockout models or small-molecule inhibition, to confirm target's necessity in disease models without introducing confounding off-target effects. Despite advances, challenges persist, including incomplete disease modeling in preclinical systems, which contributes to the overall low success rates in later stages, with fewer than 10% of clinical candidates ultimately approved. Lead discovery follows target validation, focusing on identifying initial compounds (leads) that bind and modulate the target with sufficient potency and selectivity. (HTS) of combinatorial libraries, often exceeding 1 million compounds, remains a , utilizing automated assays to measure via fluorescence or enzymatic readouts. Fragment-based lead generation (FBLG) screens smaller, low-molecular-weight fragments (typically <300 Da) for weak , which are then elaborated into higher- leads through structure-activity relationship (SAR) studies informed by or NMR. DNA-encoded libraries (DELs) enable screening of billions of compounds in a single pool by attaching DNA barcodes to molecules, allowing affinity selection and sequencing-based deconvolution, as demonstrated in the discovery of leads for kinases and protein-protein interactions. Virtual screening and rational design complement empirical methods by docking computational libraries against target structures derived from Protein Data Bank entries, prioritizing candidates with favorable pharmacokinetics predicted by quantitative structure-activity relationship (QSAR) models. Success in lead discovery hinges on hit validation to exclude artifacts like assay interference, with confirmed hits advanced to hit-to-lead optimization emphasizing drug-like properties per (molecular weight <500 Da, logP <5, hydrogen bond donors <5, acceptors <10). Attrition at this stage arises from poor selectivity or metabolic instability, underscoring the need for early (absorption, distribution, metabolism, excretion) profiling to align leads with therapeutic indices.

Preclinical Evaluation

Preclinical evaluation encompasses laboratory and conducted to assess a candidate's , pharmacological activity, , and potential prior to human testing. These studies aim to establish proof-of-concept for , determine dosing regimens, and identify adverse effects that could preclude clinical advancement. assays using cell cultures and biochemical tests evaluate mechanisms of action, while experiments in animal models provide data on systemic effects. Key components include , , , and (ADME) profiling to understand the drug's fate in biological systems. Pharmacokinetic studies measure plasma concentrations over time to inform and , often using for initial screening and larger species like dogs or non-human primates for confirmatory data. assessments, conducted under (GLP) standards, encompass acute and repeated-dose toxicity, , and safety to detect target organ toxicities and dose-response relationships. For instance, single-dose studies identify immediate hazards, while subchronic tests spanning weeks reveal cumulative effects. Regulatory agencies such as the FDA require comprehensive preclinical data to support an application, including studies in two species (typically one rodent and one non-rodent) for pivotal . However, animal models exhibit limitations in predicting human outcomes due to interspecies physiological differences, resulting in low concordance for —positive predictive value around 65% and negative predictive value 50% in drugs. Over 90% of candidates succeeding in preclinical phases fail in clinical trials, underscoring translational gaps that prompt initiatives like the FDA's 2025 roadmap to integrate non-animal alternatives such as organ-on-chip and computational modeling.

Clinical Testing Phases

Clinical trials evaluate investigational drugs in human subjects following preclinical testing, progressing through sequential phases that escalate in participant numbers, duration, and evidential rigor to establish , dosing, , and long-term effects. These phases are mandated by regulatory bodies like the U.S. (FDA) under (IND) applications, with protocols designed to minimize risks while generating data for approval. Phase transitions require interim analyses and regulatory oversight, often involving Institutional Review Boards (IRBs) for ethical compliance and Data Safety Monitoring Boards (DSMBs) for ongoing safety reviews. Attrition is high, with historical data indicating only about 10-15% of drugs entering Phase I ultimately reach market approval, driven by failures in or unexpected toxicities. Phase I trials primarily assess safety, tolerability, and in small cohorts, typically 20-100 healthy volunteers or, for certain therapies like drugs, patients with the target condition. Conducted over weeks to months under close monitoring, these studies determine maximum tolerated doses through dose-escalation designs, measuring , , , , and initial pharmacodynamic effects. Adverse events are tracked meticulously, with emphasis on dose-limiting toxicities; success rates from Phase I to II average around 60-70%, reflecting early weeding out of unsafe candidates. Phase II trials expand to 100-300 patients with the disease, focusing on preliminary alongside refined safety profiling over several months to two years. Randomized and often - or active-controlled designs test therapeutic doses identified in Phase I, evaluating endpoints like symptom reduction or changes while monitoring side effects in the target population. These studies provide dose-response data and inform Phase III protocols; however, Phase II has the lowest transition success rate, approximately 30-35%, due to efficacy shortfalls against disease heterogeneity or responses. Phase III trials involve large-scale confirmatory testing in 300-3,000 or more participants, randomized across diverse subgroups to demonstrate statistically significant and risk-benefit profiles compared to standard care or . Spanning one to four years with multicenter, international scope, these pivotal studies generate robust data on clinical outcomes, rare adverse events, and subpopulations, supporting (NDA) submissions. Phase III to approval success hovers at 50-60%, with failures often stemming from underpowered subgroup effects or regulatory thresholds for non-inferiority. Phase IV post-marketing surveillance occurs after FDA approval, monitoring real-world use in thousands to millions via observational studies, registries, or programs. These open-label or comparative effectiveness studies, ongoing indefinitely, detect long-term risks, drug interactions, or off-label applications not evident in trials, potentially leading to label updates, restrictions, or withdrawals—such as the 2011 rofecoxib recall for cardiovascular risks identified post-approval. Compliance is enforced through FDA's Risk Evaluation and Mitigation Strategies (REMS) where warranted.
PhasePrimary ObjectivesTypical ParticipantsKey Metrics AssessedApproximate Success Rate to Next Phase
ISafety, dosing, pharmacokinetics20-100 (healthy or patients)Tolerability, ADME profile60-70%
IIEfficacy signals, side effects100-300 (patients)Dose-response, preliminary endpoints30-35%
IIIConfirmatory efficacy, broad safety300-3,000+ (patients)Clinical outcomes, rare events50-60% (to approval)
IVPost-approval monitoringThousands-millions (general population)Long-term risks, real-world effectivenessN/A (ongoing)
Data derived from FDA guidelines and meta-analyses of trial outcomes across therapeutic areas, noting variability by disease (e.g., higher oncology attrition).

Regulatory Review and Approval

Following successful completion of clinical trials, pharmaceutical sponsors submit comprehensive applications to regulatory authorities seeking market approval, typically in the form of a New Drug Application (NDA) for small-molecule drugs or a Biologics License Application (BLA) for biologics to the U.S. Food and Drug Administration (FDA). These submissions include extensive data on preclinical studies, clinical trial results, manufacturing processes, proposed labeling, and risk management plans, with the FDA requiring demonstration of substantial evidence of safety and efficacy from adequate, well-controlled investigations showing benefits outweigh known risks. Approval decisions hinge on multidisciplinary reviews by pharmacology/toxicology, clinical, biometrics, and chemistry/manufacturing/controls experts, often involving advisory committee consultations for novel or high-risk therapies. The FDA's review process begins with a 60-day filing assessment to determine completeness, followed by substantive evaluation under (PDUFA) performance goals: 10 months for standard reviews and 6 months for priority designations addressing unmet medical needs. Median review times for drugs in recent years have hovered around 10-12 months, with biologics often faster than small molecules due to fewer review cycles. Incomplete or deficient applications may trigger refusals to file or complete response letters necessitating resubmissions, extending timelines; approvals can be full, accelerated (for serious conditions with surrogate endpoints), or conditional, with post-approval commitments for confirmatory studies. Internationally, the () employs a centralized procedure for advanced therapies, orphan drugs, and certain innovative products, where sponsors submit a single Marketing Authorisation Application (MAA) undergoing scientific assessment by and co- member states, culminating in a decision valid across the . This process features 210 active evaluation days, excluding clock-stops for applicant responses, typically spanning 12-18 months overall, with accelerated assessments (150 days) for breakthrough therapies. Other regions, such as Japan's or Canada's , maintain analogous frameworks emphasizing comparable and standards, though harmonization efforts via the International Council for Harmonisation (ICH) guidelines facilitate mutual reliance on data. Regulatory scrutiny extends beyond initial approval through pharmacovigilance systems monitoring real-world adverse events, enabling label updates, restrictions, or withdrawals if emerging risks alter the benefit-risk profile, as evidenced by historical cases like rofecoxib's 2004 voluntary withdrawal following cardiovascular safety signals identified post-approval. Agencies prioritize over theoretical concerns, yet face criticisms for delays impeding access to beneficial therapies or, conversely, approving agents later found inadequate, underscoring the inherent trade-offs in from finite trial data versus population-level outcomes.

Economic Realities

Capital Requirements and Cost Breakdown

Estimates of the total capitalized cost to develop and obtain regulatory approval for a new , for failed candidates in a development portfolio and the of capital, range from $879 million to $2.6 billion per approved compound, with variations arising from differences in data sources, therapeutic areas, inclusion of post-approval studies, and discount rates applied to future costs. The lower estimate derives from an analysis of U.S. data from 2000 to 2018, incorporating public registries and proprietary datasets, while the higher figure stems from a 2016 survey of pharmaceutical firms covering self-originated drugs approved between 1995 and 2007, which emphasizes internal R&D without extensive reliance on partnerships. These figures reflect pre-tax costs and exclude scale-up or expenses, though actual outlays per successful drug are amplified by high attrition rates, where only about 10-12% of candidates entering clinical testing reach approval. Out-of-pocket costs prior to capitalization are substantially lower, averaging $173 million per approved in the 2000-2018 dataset, rising to $516 million when adjusted for expected failures across phases. Preclinical and stages, involving validation, lead optimization, and , account for roughly 7% of out-of-pocket expenses but increase to about 40% under capitalized conditions due to earlier failure risks and lower success probabilities (around 50-70% advancement to clinical phases). Clinical dominates, comprising 68% of out-of-pocket costs, with 3 trials contributing the majority owing to their scale—typically involving hundreds of patients over extended durations—compared to 1 (small safety cohorts) and 2 (preliminary efficacy). Regulatory review adds minimal direct cost (1-2%), while 4 post-approval commitments can extend total outlays by 20-25% for long-term safety monitoring. Costs vary significantly by therapeutic category, with and anesthesia drugs reaching $1.76 billion capitalized (driven by subjective endpoints and high placebo responses requiring larger trials), at $1.21 billion (complex patient stratification and endpoints), and anti-infectives at the lower end of $379 million (simpler trial designs). Recent analyses confirm in distributions, where direct R&D costs per drug are $150 million versus means exceeding $369 million, indicating that a few high-cost outliers inflate averages, particularly in biologics or indications demanding specialized assays and global enrollment. Overall industry R&D expenditures, totaling $83 billion in 2019 across U.S. firms, underscore the , necessitating diversified pipelines and financing strategies to mitigate per-drug risks. Rising complexity, regulatory demands, and trial sizes have driven a 145% increase in capitalized costs since early estimates of $802 million (inflation-adjusted).

Attrition Rates and Probability of Success

The drug development pipeline exhibits exceptionally high attrition rates, with failure predominantly driven by insufficient efficacy, unacceptable toxicity, pharmacokinetic issues, or strategic decisions unrelated to scientific merit. Empirical analyses of clinical programs indicate that the overall likelihood of approval (LOA) from Phase I to regulatory approval stands at 7.9% for candidates entering development between 2011 and 2020, reflecting persistent challenges in translating preclinical promise into human benefit. Including preclinical stages amplifies attrition, as fewer than 0.01% of screened compounds typically advance to market approval, underscoring the causal bottleneck of biological complexity and incomplete predictive models. Phase-specific success rates reveal stark disparities, with Phase II serving as the most formidable barrier due to the initial robust efficacy signals required amid heterogeneous patient responses. Across all therapeutic areas from 2011 to 2020, transition probabilities were approximately 70% from Phase I to II, 33% from Phase II to III, 56% from Phase III to submission, and 92% from submission to approval. These figures derive from aggregated data on over 12,000 phase transitions, primarily from sponsors, though they may understate risks in smaller biotech firms where resource constraints exacerbate failures. Therapeutic area profoundly influences , with oncology programs facing the lowest LOA at around 4.2% from Phase I, attributable to tumor heterogeneity, adaptive resistance mechanisms, and stringent endpoints like overall survival. In contrast, non-oncology areas such as anti-infectives or exhibit higher rates, often exceeding 15%, reflecting more straightforward and surrogate endpoints. Modal differences also matter: small molecules succeed at rates comparable to biologics in early phases but lag in later ones due to manufacturing scalability issues.
Phase TransitionAll Areas Success Rate (2011-2020)Oncology Success Rate (2011-2020)
Phase I to II70%61%
Phase II to III33%24%
Phase III to Submission56%46%
Submission to Approval92%84%
Overall LOA (Phase I to Approval)7.9%4.2%
Data compiled from industry-wide phase transitions; rates vary by sponsor size and indication subtype. Recent trends show modest improvements in LOA for certain modalities, such as gene therapies (reaching 20-30% in some cohorts), driven by refined selection via biomarkers, yet systemic factors like regulatory stringency and economic pressures sustain overall low probabilities. These metrics, drawn from proprietary databases like those of BIO and Amplion, highlight the need for causal interventions in validation to mitigate , rather than relying on volume-based screening alone.

Investment Models and Market Valuation

Drug development relies on diverse investment models to finance the high-risk, capital-intensive process, primarily through venture capital for early-stage biotech firms, corporate partnerships with large pharmaceutical companies, and public market offerings such as initial public offerings (IPOs). Venture capital funding supports target identification and preclinical stages, where investors provide equity in exchange for potential high returns from successful commercialization or acquisitions, though recent trends show a contraction: global biotech venture funding dropped 45% from 2021 peaks into 2023-2024, with H1 2025 financings down over 20% year-over-year amid selective larger rounds favoring derisked assets. Large pharma firms often invest via internal R&D budgets—totaling $276 billion globally in 2021 across biopharma—or milestone-based alliances that mitigate risk by sharing costs and revenues post-proof-of-concept. Public markets enable later-stage funding through IPOs, but volatility has led to fewer listings, with only six biotech IPOs raising $860 million in Q1 2025. Market valuation of drug assets and companies predominantly employs risk-adjusted net present value (rNPV), which discounts projected cash flows from peak sales forecasts by phase-specific probabilities of technical and regulatory success, reflecting attrition rates averaging 85-90% from preclinical to approval. Unlike standard (NPV), rNPV incorporates empirical success probabilities—e.g., 14.3% likelihood of first approval across leading pharma pipelines—to yield realistic enterprise values for pipelines, often comprising 70-90% of a biotech firm's worth. Complementary methods include (DCF) for approved products and comparable transactions for M&A, where multiples vary by therapeutic area; for instance, assets command higher valuations due to larger addressable markets. These models underpin licensing deals, with upfront payments and royalties calibrated to rNPV estimates, though over-optimism in sales projections can inflate valuations, as critiqued in analyses of historical biotech returns. Empirical returns on pharmaceutical R&D investments remain modest despite innovation outputs, with the forecast (IRR) for top 20 biopharma firms rising to 5.9% in 2024 from prior lows, driven by late-stage assets but pressured by escalating costs exceeding $2 billion per approved . Preclinical investments in multi-indication leads have yielded annualized returns of 21% until FDA approval in historical cohorts, underscoring the value of diversified pipelines, yet overall ROI declines highlight sustainability challenges amid regulatory hurdles and pricing pressures. Valuation multiples for biotech revenue in 2025 average 4-6x for clinical-stage firms, influenced by interest rates and , with and AI-enabled platforms attracting premiums due to perceived efficiency gains. These metrics inform investor decisions, balancing high failure risks against blockbuster potential, where successful drugs can generate 10-20x returns on invested capital.

Technological and Methodological Advances

Computational and AI-Driven Approaches

Computational approaches in drug development encompass techniques such as , , and (vHTS), which enable the rapid evaluation of compound libraries against biological targets without extensive physical experimentation. These methods model ligand-protein interactions to predict binding affinities and optimize , significantly reducing the time and cost associated with traditional screening. For instance, vHTS can assess millions of virtual compounds, prioritizing those with favorable for synthesis and testing, thereby streamlining hit identification in the early . simulations further refine these predictions by accounting for conformational flexibility and over timescales of nanoseconds to microseconds. The integration of artificial intelligence (AI) and machine learning (ML) has accelerated these processes, particularly since 2020, by leveraging large datasets for predictive modeling across target identification, lead optimization, and de novo molecule generation. AI-driven tools employ deep learning algorithms, such as convolutional neural networks and generative adversarial networks, to forecast drug-target interactions and design novel scaffolds with desired properties like solubility and selectivity. A landmark advancement is DeepMind's AlphaFold series; AlphaFold2, released in 2021, achieved near-experimental accuracy in protein structure prediction, enabling structure-based drug design for previously intractable targets, while AlphaFold3 in 2024 extended predictions to protein-ligand complexes, outperforming prior tools in binding pose accuracy. This has facilitated applications in over 2 million research instances globally, including accelerated inhibitor design for enzymes like SARS-CoV-2 main protease. Generative AI models exemplify de novo drug design, producing chemically feasible molecules from scratch by learning patterns from chemical libraries. Examples include REINVENT 4 (2024), which uses recurrent neural networks and transformers to generate drug-like candidates optimized for multi-objective criteria such as potency and ADMET profiles, and earlier frameworks like MolGAN for graph-based molecule synthesis. These approaches have demonstrated success in prospective studies, yielding hits with micromolar affinities validated experimentally, though success rates remain below 10% without hybrid experimental validation due to limitations in capturing and synthetic feasibility. Despite biases in —often skewed toward approved drugs—AI has shortened discovery timelines from years to months in select cases, as seen in collaborations compounds for rare diseases. Empirical validation persists as essential, with computational predictions informing but not supplanting preclinical assays to mitigate false positives.

Novel Modalities and Delivery Systems

Novel modalities in drug development encompass therapeutic approaches extending beyond traditional small-molecule drugs and monoclonal antibodies, including RNA-based therapeutics, editing technologies, and therapies, which target underlying mechanisms at the genetic or cellular level. These modalities have proliferated due to advances in , with nearly 3,500 , , and RNA therapies in preclinical or clinical development as of 2023, reflecting accelerated investment amid successes like mRNA vaccines for COVID-19. RNA therapeutics, such as (mRNA) and (siRNA), enable transient protein expression or without altering the , offering reversibility and rapid production scalability compared to DNA-based methods. For instance, mRNA platforms have expanded into and rare diseases, with clinical trials demonstrating efficacy in protein replacement for conditions like . Gene editing modalities, primarily utilizing CRISPR-Cas9 systems, allow precise DNA modifications to correct mutations, with applications in monogenic disorders such as , where editing of hematopoietic stem cells has led to FDA-approved therapies like Casgevy in December 2023. In vivo delivery remains a hurdle, often relying on nanoparticles (LNPs) or adeno-associated viruses (AAVs), but off-target effects and immune responses necessitate ongoing refinements, as evidenced by preclinical data showing improved specificity in editing variants. Cell therapies, including CAR-T cells for hematologic malignancies, involve engineering patient-derived cells to target tumor antigens, achieving remission rates exceeding 80% in refractory B-cell lymphomas, though manufacturing scalability and limit broader adoption. These modalities collectively address unmet needs in , , and genetic diseases, but face higher development costs—often 2-3 times those of small molecules—due to complex and potency assays. Advanced delivery systems are integral to realizing the potential of novel modalities, overcoming barriers like poor , tissue specificity, and stability. Nanoparticle-based carriers, such as LNPs for mRNA, facilitate endosomal escape and cytosolic release, as demonstrated in the Pfizer-BioNTech vaccine's 95% efficacy against symptomatic in 2020 trials, where ionizable lipids enabled hepatic targeting. Targeted nanoparticles conjugated with ligands like aptamers or antibodies enhance selectivity, reducing off-target toxicity; for example, polymeric nanoparticles have improved delivery to solid tumors, achieving 2-5 fold higher intratumoral concentrations in models. Liposomes and micelles provide controlled release, with pH-sensitive variants releasing payloads in acidic tumor microenvironments, supported by clinical data from Doxil, approved in but refined in recent PEGylated iterations for prolonged circulation half-lives up to 100 hours. Emerging delivery innovations include nanofibers for localized sustained release and hydrogels for implantable depots, which degrade predictably to maintain therapeutic levels over weeks, as in insulin-loaded alginate systems extending glycemic in diabetic models. Viral vectors like AAVs dominate delivery, with 9 variants crossing the blood-brain barrier for neurologic indications, though affects 30-50% of patients, prompting non-viral alternatives like electroporation-enhanced LNPs. Smart systems responsive to stimuli—such as glucose-sensitive micelles for —integrate diagnostics and , minimizing dosing frequency and side effects, with preclinical in reducing HbA1c by 1-2% in animal studies. Despite these advances, challenges persist in and regulatory hurdles, with only 10-15% of novel modality pipelines reaching approval, underscoring the need for standardized under cGMP to mitigate variability.

Regulatory and Global Frameworks

Key Agencies and Approval Processes

The Food and Drug Administration () serves as the primary regulatory agency for drug approvals in the U.S., overseeing the process through its Center for Drug Evaluation and Research (CDER) for drugs and Center for Biologics Evaluation and Research (CBER) for biologics. Manufacturers must submit an () application after preclinical testing to initiate clinical trials, followed by phased trials (Phase 1 for safety, Phase 2 for efficacy and dosing, Phase 3 for confirmatory data in larger populations). Upon completion, a () or Biologics License Application () is filed, with the FDA conducting a standard review in approximately 10 months or a in 6 months for drugs addressing unmet needs. Approval requires demonstration that benefits outweigh risks based on substantial evidence from adequate, well-controlled studies. In the European Union, the European Medicines Agency (EMA) manages centralized authorizations for novel, biotech, and advanced therapy medicinal products via a single marketing authorization application submitted to EMA, which coordinates scientific assessment by the Committee for Medicinal Products for Human Use (CHMP). The centralized procedure involves an initial 210 active days of review, excluding clock-stops for additional data requests, leading to a binding recommendation for EU-wide approval if positive. For non-centralized products, national procedures or decentralized/mutual recognition apply, but the centralized route ensures unified market access across member states. Other major agencies include Japan's (PMDA), which reviews applications under the Ministry of Health, Labour and Welfare, emphasizing early consultations and a standard review timeline of 12 months; Australia's (TGA), aligning closely with ICH standards for efficient approvals; and Brazil's National Health Surveillance Agency (ANVISA), handling local requirements amid efforts toward international alignment. The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) facilitates global consistency by developing guidelines on quality, safety, efficacy, and multidisciplinary topics, adopted by agencies like FDA, , and PMDA to reduce duplicative testing and streamline multinational development. ICH guidelines, such as those for stability testing (Q1) and (E6), underpin approval processes worldwide, though implementation varies by jurisdiction. This harmonization aims to ensure safe, effective medicines reach markets efficiently without compromising standards.

International Variations and Harmonization Efforts

Drug approval processes exhibit significant variations across jurisdictions, influenced by differing priorities in safety evaluation, clinical data requirements, and timelines. In the United States, the requires a or Biologics License Application (BLA), emphasizing comprehensive phase III trials and often granting accelerated approvals for unmet needs, with median review times around 10 months for standard NDAs as of 2023. By contrast, the employs a centralized authorization procedure for novel drugs, harmonizing across EU member states but imposing stricter requirements, resulting in approval timelines averaging 210 days for standard procedures in 2022. Japan's mandates bridging studies or local clinical data to account for ethnic differences, extending timelines by 12-18 months compared to FDA pathways, particularly for non-oncology drugs. Emerging markets like Brazil's ANVISA have adopted reliance models on ICH guidelines but retain local manufacturing and stability testing mandates, leading to approval delays of up to two years for complex biologics. These disparities create challenges for multinational developers, including duplicated clinical trials, varying standards, and inconsistent post-approval commitments; for instance, requires risk management plans (RMPs) earlier than FDA's Risk Evaluation and Mitigation Strategies (REMS). In , agencies like ANVISA prioritize local data generation over full reliance on FDA/ approvals, contrasting with more flexible approaches in or that often defer to ICH-aligned assessments. Such variations stem from differences, resource constraints, and historical precedents, with peer-reviewed analyses indicating that FDA approves more drugs annually (about 15-20) than (10-15) due to broader acceptance. Harmonization efforts, led by the International for Harmonisation (ICH), aim to align technical requirements for quality, safety, efficacy, and multidisciplinary aspects of drug development. Founded in 1990 as the International Conference on Harmonisation by regulators and industry from the , , and , ICH has issued over 30 guidelines, such as Q8-Q10 on pharmaceutical quality systems and S9 on nonclinical evaluation for , reducing redundant testing and facilitating global dossiers. Transitioning to a formal in 2015, ICH expanded membership to include , , and , while WHO acts as an observer to promote adoption in low- and middle-income countries. These guidelines, implemented via the (CTD) format, have streamlined submissions, with studies showing a 20-30% reduction in development timelines for ICH-compliant products across regions. Recent developments include ICH's E8(R1) revision in 2017 emphasizing patient-centered trials and ongoing work on integration, alongside WHO's prequalification programs that rely on stringent regulatory assessments for . Bilateral mutual recognition agreements, such as FDA-EMA confidentiality arrangements since 2003, further mitigate variations by enabling without full re-reviews. Despite progress, full convergence remains elusive due to persistent national , as evidenced by Japan's occasional on ethnic-specific and emerging markets' adaptation lags; however, reliance pathways adopted by over 30 countries since 2020 have accelerated access to and biologics during pandemics. Ongoing ICH assemblies continue to address digital submissions and advanced therapies, fostering incremental global alignment while preserving regulatory independence.

Challenges, Criticisms, and Controversies

Safety, Efficacy, and Ethical Concerns

Safety assessments in drug development reveal substantial risks persisting beyond initial approvals, with approximately one-third of new drugs approved by the U.S. Food and Drug Administration (FDA) between 2001 and 2010 encountering major safety issues during post-market surveillance. Attrition rates underscore these vulnerabilities: across clinical phases, safety concerns contribute to 24% of Phase III failures, while overall drug development sees over 90% of candidates fail, often due to undetected toxicities like hepatotoxicity or cardiotoxicity that emerge post-approval. Post-market withdrawals, such as those for severe adverse events, affect a notable fraction of launched products, with 38% of certain post-1950 drugs pulled within one year of the first reported death linked to their use. Efficacy challenges compound safety hurdles, as 40-50% of clinical failures stem from inadequate therapeutic benefits, particularly in late-stage trials where 44% of Phase III discontinuations occur due to insufficient . Roughly half of investigational drugs reaching pivotal trials fail primarily from efficacy shortfalls, exacerbated by that favors positive outcomes and skews meta-analyses toward overstated benefits. Industry-sponsored trials report psychiatric drugs as 50% more effective than independent ones, highlighting how funding sources can inflate perceived efficacy through selective reporting. Ethical concerns permeate both preclinical and clinical stages, including reliance on animal testing where over 90% of drugs deemed safe and effective in animals fail to translate to human approvals, raising questions about the validity and welfare implications of such models. Human subject protections face strains from conflicts of interest, as pharmaceutical payments to FDA advisers post-approval have sparked scrutiny over potential influences on and efficacy evaluations. These dynamics, including biased trial designs and underreporting of negative results, undermine and equitable risk distribution, particularly when trials occur in vulnerable populations or prioritize speed over comprehensive data.

Pricing, Access, and Profit Incentives

Pharmaceutical companies determine drug prices primarily based on recouping substantial (R&D) costs, which averaged approximately $2.3 billion per approved asset in 2024, alongside anticipated , , and profit margins to justify the high failure rates in clinical trials. , where pricing operates in a relatively free-market environment without direct for most payers, list prices for branded drugs reflect factors such as projected needs to cover R&D portfolios—where only about 1 in 10 compounds entering clinical trials ultimately succeeds—and competition from alternatives. Pharmacy benefit managers (PBMs) and insurers negotiate rebates and discounts, often reducing net prices by 20-50% for high-volume drugs, though gross list prices remain elevated to facilitate these negotiations. In contrast, U.S. prices in 2022 were 2.78 times higher than in 33 other () countries, with branded drugs averaging 3.22 times higher, largely due to international reference pricing and government negotiations in and that cap reimbursements based on cost-effectiveness thresholds or external price referencing. For instance, patented drugs in are priced comparably to or below averages through the Patented Medicine Prices Review Board, which can mandate reductions if prices exceed international medians. This disparity positions the U.S. market as a primary source for global R&D, funding innovations that benefit lower-priced markets abroad, though it contributes to domestic affordability challenges, with out-of-pocket costs straining uninsured or underinsured patients. Profit incentives are central to sustaining drug development, as evidenced by the industry's $185 billion R&D expenditure in fiscal 2023—representing about 20% of revenues for major firms, far exceeding other sectors—and studies showing that a 10% in expected U.S. revenues could decrease by up to 15%, measured by fewer new molecular entities entering pipelines. Patents granting 20-year exclusivity (with effective market of 10-15 years post-approval due to regulatory delays) enable pricing power to achieve internal rates of return around 7-10% on successful drugs, compensating for overall portfolio risks where 90% of investments fail. Economic analyses indicate that diminishing these incentives through aggressive , as in some European systems, correlates with slower approval timelines and reduced investment in high-risk areas like , underscoring profits' causal role in directing capital toward unmet needs rather than guaranteed returns. Access barriers persist globally, particularly in low- and middle-income countries where are unavailable or unaffordable for up to one in four people, exacerbated by high originator prices, limited , and dependencies—79% of pharmaceuticals in are imported, inflating costs. Post-patent entry reduces prices by 80-90% in competitive markets, improving access, but enforcement and data exclusivity in trade agreements can delay this by 3-5 years, prioritizing innovation incentives over immediate affordability in resource-poor settings. Initiatives like voluntary licensing and tiered pricing by manufacturers aim to bridge gaps, yet data from 54 low- and middle-income countries show availability below WHO targets of 80%, with affordability issues persisting for chronic treatments due to income disparities rather than pricing alone. Critics, often from groups, argue profits engender inequities, but empirical reviews reveal that profit-driven models have tripled drug approvals since 2000, suggesting curtailed incentives would diminish future access by stalling pipelines.

Intellectual Property and Innovation Barriers

Intellectual property rights, primarily through patents and regulatory exclusivities, serve as the primary mechanism to incentivize pharmaceutical innovation by granting originators temporary market exclusivity to recoup substantial research and development (R&D) investments. The average cost to bring a new drug to market exceeds $2.6 billion, encompassing preclinical and clinical stages, with development timelines spanning 10 to 15 years from discovery to approval. Over 90% of compounds fail in clinical trials, amplifying financial risks and underscoring the necessity for exclusivity to attract capital. Empirical analyses across countries demonstrate that stronger intellectual property protections correlate with increased domestic pharmaceutical innovation and R&D spending, as firms prioritize jurisdictions with enforceable rights to safeguard returns. Patents typically last 20 years from filing, but effective market exclusivity averages 7 to 12 years post-approval due to the protracted regulatory , during which no revenue is generated. Regulatory exclusivities, such as the U.S. Food and Drug Administration's 5-year protection, further extend this period but do not always align with terms, creating a layered . This compressed exclusivity window poses a barrier to , as the finite period must cover not only initial recoupment but also subsequent investments in scale-up, post-approval studies, and lifecycle . In countries with weaker , multinational firms reduce R&D commitments, leading to lower output compared to strong-IP environments like the U.S. or EU. Critics contend that practices like secondary patenting—often termed ""—extend monopolies through minor formulations or delivery modifications, potentially blocking entry and follow-on research. However, analyses indicate that such patents frequently reflect genuine incremental innovations, such as improved stability or , rather than systematic abuse, with U.S. government reviews finding no widespread evidence of undermining the system's innovation mandate. thickets, involving overlapping claims across compounds, processes, and uses, can deter smaller entrants by raising litigation costs, yet cross-country evidence links robust regimes to higher overall R&D investment and novel drug approvals, suggesting net benefits outweigh barriers. Weakening to address access concerns risks diminished future innovation, as observed in jurisdictions with lax protections experiencing reduced pharmaceutical FDI and product launches.

References

  1. [1]
    The Drug Development Process | FDA
    Jan 4, 2018 · Step 1: Discovery and Development · Step 2: Preclinical Research · Step 3: Clinical Research · Step 4: FDA Drug Review · Step 5: FDA Post-Market ...Step 3: Clinical Research · Step 1: Discovery and... · Step 4: FDA Drug Review
  2. [2]
    Drug Development 101: A Primer - PubMed
    Drug development is a term used to define the entire process of bringing a new drug or device to market. It is an integrated, multidisciplinary endeavor ...
  3. [3]
    Research & Development Policy Framework - PhRMA
    On average, it takes 10-15 years and costs $2.6 billion to develop one new medicine, including the cost of the many failures. Only 12% of new molecular entities ...
  4. [4]
    Costs of Drug Development and Research and ... - JAMA Network
    Jun 28, 2024 · This economic evaluation estimates the cost of drug development by therapeutic class and trends in pharmaceutical research and development ...
  5. [5]
    Why 90% of clinical drug development fails and how to improve it?
    Despite this validated effort, the overall success rate of clinical drug development remains low at 10%–15%5, 6, 7. Such persistent high failure rate raises ...
  6. [6]
    90% of drugs fail clinical trials
    Mar 12, 2022 · Lastly, 10% of failures were attributed to lack of commercial interest and poor strategic planning. This high failure rate raises the question ...
  7. [7]
    Development & Approval Process | Drugs - FDA
    Aug 8, 2022 · Get to know FDA's drug development and approval process -- ensuring that drugs work and that the benefits outweigh their known risks.How Drugs are Developed · Drug Development Tools | DDTs
  8. [8]
    Factors associated with clinical trials that fail and opportunities ... - NIH
    Aug 7, 2018 · Clinical trials for pharmaceuticals and medical devices offer many opportunities for failure. Failures can arise from a lack of efficacy, issues ...
  9. [9]
    The History of Pharmacy | Texas Tech University Health Sciences ...
    The world's oldest known prescriptions were recorded on a clay tablet in Mesopotamia (modern-day Iraq), ca. 2400 BC.
  10. [10]
    A Historical Overview of Pharmacology | Carrington College
    Sep 3, 2025 · Documented evidence of early drug use became more abundant around 2,600 BCE, when early medicines were detailed by pressing cuneiform signs into ...
  11. [11]
    Historical review of medicinal plants' usage - PMC - NIH
    Healing with medicinal plants is as old as mankind itself. The connection between man and his search for drugs in nature dates from the far past.
  12. [12]
    The Air of History: Early Medicine to Galen (Part I) - PMC - NIH
    Galen is a giant in the history of medicine and casts a long shadow. His medical theories dominated European medicine for 1500 years.
  13. [13]
    A fascinating look at primitive and ancient medicine by medical ...
    May 25, 2015 · Plinio Prioreschi, MD, PhD (1930–2014), the author of this monumental series of tomes on the history of medicine was an accomplished scholar.
  14. [14]
    The history of pharmacy | Express Scripts
    Like all healthcare professions, the origins of pharmacy can be traced back thousands of years. Take a quick look at the past few centuries to present day.<|separator|>
  15. [15]
    Early drug discovery and the rise of pharmaceutical chemistry
    Early drugs came from natural sources. The first synthetic drug was chloral hydrate. Aspirin was the first blockbuster drug. Barbiturates were introduced at ...
  16. [16]
    Natural Products That Changed Society - PMC - PubMed Central
    Apr 26, 2021 · During the 19th century chemists succeeded in isolating pure natural products such as quinine, morphine, codeine and other compounds with beneficial effects.
  17. [17]
    Drugs and their Manufacture in the Nineteenth Century - OnView
    Drugs that were isolated (or “discovered”) during the first half of the century included alkaloids such as strychnine, emetine, morphine, quinine, and caffeine.
  18. [18]
    Early drug discovery and the rise of pharmaceutical chemistry - Jones
    Jun 23, 2011 · During the first decades of the nineteenth century, French, Italian, and German chemists attempted to isolate the active principle contained ...Missing: ingredients 19th
  19. [19]
    The History of Drug Regulation | FDA
    Feb 1, 2018 · Drug regulation in FDA has evolved considerably since President Theodore Roosevelt signed the 1906 Pure Food and Drugs Act.
  20. [20]
    Milestones in US Food and Drug Law - FDA
    Jan 30, 2023 · 1800-1899: FDA Milestones ; 1820, Eleven physicians meet in Washington, D.C., to establish the U.S. Pharmacopeia, the first compendium of ...
  21. [21]
    Timeline of Discovery | Harvard Medical School
    Harvard Medical School's timeline includes the smallpox vaccine (1799), anesthesia (1846), insulin (1922), the first heart valve surgery (1923), and the ...
  22. [22]
    1900 - 1999: The 20th century - ABPI schools
    Nov 22, 2021 · The 20th century saw the development of insulin, penicillin, and modern medicines, increased life expectancy, and the first heart transplant ...
  23. [23]
    The history and contemporary challenges of the US Food and Drug ...
    The Food, Drug, and Cosmetic Act that replaced it in 1938, and subsequent food and drug laws and amendments, expanded the FDA's responsibilities to cosmetics, ...
  24. [24]
    Major Milestones in Medicine, Drug Development in Recorded History
    Apr 20, 2022 · Chemotherapy was accidentally developed at the beginning of the 20th century when mustard gas was used as a weapon in both World Wars.
  25. [25]
    Refining the impact of genetic evidence on clinical success - Nature
    Apr 17, 2024 · We estimate the probability of success for drug mechanisms with genetic support is 2.6 times greater than those without. This relative success ...
  26. [26]
    Target identification and mechanism of action in chemical biology ...
    In this review, we will cover three distinct and complementary approaches for discovering the protein target of a small molecule: direct biochemical methods, ...
  27. [27]
    Target identification of small molecules: an overview of the current ...
    Oct 10, 2023 · Target identification is a crucial stage in the discovery and development of new drugs since it enables researchers to understand the mode of ...
  28. [28]
  29. [29]
    Advances in Lead Generation - ScienceDirect.com
    Feb 15, 2019 · Lead Generation is the discipline in the drug discovery cascade where chemistry assets are first created and their potential value is assessed ...Abstract · High Throughput Screening · Fragment Based Lead...<|separator|>
  30. [30]
    Efficient Drug Lead Discovery and Optimization - PMC
    Lead generation and optimization can be pursued through joint computational and experimental studies.Lead Generation · Lead Optimization · Heterocycle Scans
  31. [31]
    Step 2: Preclinical Research - FDA
    Jan 4, 2018 · After preclinical testing, researchers review their findings and decide whether the drug should be tested in people. Content current as of: 01/ ...
  32. [32]
    Drug Development and Review Definitions - FDA
    Aug 20, 2015 · Pre-Clinical Research​​ During preclinical drug development, a sponsor evaluates the drug's toxic and pharmacologic effects through in vitro and ...
  33. [33]
    Preclinical Pharmacokinetics in Drug Development - Allucent
    Understanding these key pharmacokinetic elements are essentially captured in the classical areas of Absorption, Distribution, Metabolism and Excretion (ADME).
  34. [34]
    6 Types of Toxicology Studies for IND & NDA Programs - WuXi AppTec
    May 2, 2024 · Like general toxicity testing, both single-dose and repeated-dose toxicology studies are utilized. Conclusion. Selecting the correct toxicology ...
  35. [35]
    The evolving role of investigative toxicology in the pharmaceutical ...
    Feb 13, 2023 · Investigative toxicology tools and strategies are used in pharmaceutical companies to reduce safety-related attrition in drug development.
  36. [36]
    [PDF] Guidance for Industry - S6 Preclinical Safety Evaluation of ... - FDA
    The comparability of the test material during a development program should be demonstrated when a new or modified manufacturing process is developed or other ...
  37. [37]
    Pre-clinical animal models are poor predictors of human toxicities in ...
    Sep 1, 2020 · Overall, animal toxicity did not show strong correlation with human toxicity, with a median PPV of 0.65 and NPV of 0.50.
  38. [38]
    [PDF] Roadmap to Reducing Animal Testing in Preclinical Safety Studies
    Apr 10, 2025 · This scientific roadmap lays out an initial strategy for FDA to reduce and replace animal testing in preclinical safety assessment of drugs and ...<|separator|>
  39. [39]
    Step 3: Clinical Research - FDA
    Jan 4, 2018 · Researchers design Phase 3 studies to demonstrate whether or not a product offers a treatment benefit to a specific population.
  40. [40]
    Factors Affecting Success of New Drug Clinical Trials - PMC - NIH
    May 11, 2023 · In spite of time-consuming processes and high costs, the overall success rate of clinical trials is only 7.9%, which is a high risk for ...
  41. [41]
    FDA's Drug Review Process: Continued
    Aug 24, 2015 · Phase 2 studies (typically involve a few dozen to about 300 people). Phase 3 studies (typically involve several hundred to about 3,000 people).
  42. [42]
    Estimation of clinical trial success rates and related parameters - NIH
    Jan 31, 2018 · Comparing these results against those for all drug development, we see that, while the Phase 1 POS increases from 66.4% to 75.9%, the Phase 2 ...
  43. [43]
    The FDA's Drug Review Process: Ensuring Drugs Are Safe and ...
    Nov 24, 2017 · The FDA uses postmarket requirement and commitment studies to gather additional information about a product's safety, efficacy, or optimal use.
  44. [44]
    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.
  45. [45]
    Step 4: FDA Drug Review
    Jan 4, 2018 · The FDA review team thoroughly examines all submitted data on the drug and makes a decision to approve or not to approve it.
  46. [46]
    US FDA's Median Review Time Remains Less Than One Year ...
    Jan 16, 2025 · Novel agents from the biologics center had a median review time of 10.6 months, beating the drugs center's median of 11.8 months thanks to fewer multicycle and ...
  47. [47]
    How Drugs are Developed and Approved - FDA
    Oct 24, 2022 · For example, OTC drug products previously available only by prescription are first approved through the NDA process and their "switch" to OTC ...Missing: steps | Show results with:steps
  48. [48]
    Authorisation of medicines | European Medicines Agency (EMA)
    Under the centralised authorisation procedure, pharmaceutical companies submit a single marketing-authorisation application to EMA.
  49. [49]
    FDA vs. EMA: Key Differences in Drug Approval You Need to Know
    Oct 16, 2025 · A standard review under the centralized procedure takes roughly 210 days, but this does not include “clock stops,” periods when the applicant ...<|separator|>
  50. [50]
    Regulatory pathways in the European Union - PMC - PubMed Central
    The process of the centralized procedure is triggered when the applicant sends the letter announcing the intent to submit a MAA (letter of intent), which is ...
  51. [51]
    Access and Safety—The FDA Drug Approval Conundrum
    Jul 18, 2024 · The approval process and subsequent safety requirements for new medications introduced to the market is a delicate balance for FDA regulators.
  52. [52]
    Innovation in the pharmaceutical industry: New estimates of R&D costs
    The estimated average out-of-pocket cost per approved new compound is $1395 million (2013 dollars). Capitalizing out-of-pocket costs to the point of marketing ...Missing: breakdown phase
  53. [53]
    Typical Cost of Developing a New Drug Is Skewed by Few High ...
    Jan 7, 2025 · Researchers estimated a median direct research and development cost of $150 million compared to a mean of $369 million.
  54. [54]
    Research and Development in the Pharmaceutical Industry
    Apr 8, 2021 · The pharmaceutical industry devoted $83 billion to R&D expenditures in 2019. Those expenditures covered a variety of activities, including ...
  55. [55]
    [PDF] Clinical Development Success Rates and Contributing Factors 2011 ...
    Oncology drug development program transitions in the 2011–2020 period accounted for 33% of the 12,728 total transitions. With the third-lowest LOA from Phase I ...
  56. [56]
    What Are the Chances of Getting a Cancer Drug Approved?
    We found that only 3.4 percent of all cancer drug development programs from 2000 to 2015 moved from phase 1 to regulatory approval.
  57. [57]
    Large vs. Small Molecule Success Rates - Applied Clinical Trials
    In Preclinical small molecules have a 63% rate of success vs. 79% for large molecules; the difference in Phase I is 41% for small molecules vs. 52% for large.
  58. [58]
    2025 Biotech Fundraising Trends: A Two-Speed Capital Market
    Sep 24, 2025 · Private biotech financings in the first half of 2025 are down more than 20% compared to the same period in 2024, according to PitchBook, with ...
  59. [59]
    [PDF] Comprehensive measurement of biopharmaceutical R&D investment
    Aug 6, 2024 · Global biopharmaceutical R&D investment in 2021 was $276 billion across 4,191 companies, with total net revenue of $1,022 billion across 583 ...
  60. [60]
    EY Biotech Beyond Borders Report 2025
    Venture funding in 2022 and 2023 fell below the pre-pandemic historical norm of US$47.5b. In Q1 2025, only six companies raised a total of. $860 million.
  61. [61]
    Valuing Pharmaceutical Assets: When to Use NPV vs rNPV - Alacrita
    In summary, the NPV method simply and easily determines the current value of an in-process research and development asset based on its expected revenues/costs ...
  62. [62]
    Benchmarking R&D success rates of leading pharmaceutical ...
    Our study reveals an average likelihood of first approval rate of 14.3% across leading research-based pharmaceutical companies, broadly ranging from 8% to 23%.
  63. [63]
  64. [64]
    Risk-Adjusted NPV in Biotech Valuation - Financial Models Hub
    Jun 3, 2025 · Risk-adjusted NPV (rNPV) is a variation of the classic Net Present Value (NPV) method. Like traditional NPV, it projects a stream of future cash ...
  65. [65]
    Measuring the return from pharmaceutical innovation 2024 - Deloitte
    Scaling investment in technologies like gene editing, AI-powered drug development platforms, and automating the lab of the future will be crucial for ...
  66. [66]
    Valuation and Returns of Drug Development Companies
    Jan 11, 2022 · Similarly, we calculated that investment in pre-clinical companies with multi-indication lead products yield annual returns of 21% until FDA ...
  67. [67]
    Biotech Valuation Multiples: 2025 Insights & Trends
    Mar 12, 2025 · This article breaks down how biotech valuation multiples work, what's driving them in 2025, and what to watch for across different segments of the industry.
  68. [68]
    Global Trends in R&D 2025 - IQVIA
    Mar 26, 2025 · R&D funding has increased: Biopharma funding increased for the second consecutive year; Total large pharma R&D spending continued to increase.
  69. [69]
    A Review on Applications of Computational Methods in Drug ...
    Various computational methods have dramatically reduced the time and cost of drug discovery. In this review, we firstly discussed roles of multiscale ...
  70. [70]
    Computational approaches streamlining drug discovery - Nature
    Apr 26, 2023 · Here we review recent advances in ligand discovery technologies, their potential for reshaping the whole process of drug discovery and development.
  71. [71]
    Review High-throughput and virtual screening: core lead discovery ...
    In addition to high-throughput screening (HTS), the main lead discovery technology employed by most pharmaceutical companies today is virtual screening (VS).
  72. [72]
    A review of the current trends in computational approaches in drug ...
    Sep 27, 2024 · Computational tools and methods for drug design and development such as molecular dynamics (MD), molecular docking, quantum mechanics (QM), ...
  73. [73]
    AI-Driven Drug Discovery: A Comprehensive Review | ACS Omega
    Jun 6, 2025 · This comprehensive review critically analyzes recent advancements (2019–2024) in AI/ML methodologies across the entire drug discovery pipeline.
  74. [74]
    Advances in machine learning for optimizing pharmaceutical drug ...
    The integration of advanced machine learning methodologies has revolutionized the field of pharmaceutical drug discovery by addressing critical challenges ...
  75. [75]
    Major AlphaFold upgrade offers boost for drug discovery - Nature
    May 8, 2024 · AlphaFold3, the researchers found, substantially outperforms existing software tools at predicting the structure of proteins and their partners.
  76. [76]
    Review of AlphaFold 3: Transformative Advances in Drug Design ...
    Jul 2, 2024 · AlphaFold 3 has achieved unprecedented accuracy in predicting the three-dimensional structures of proteins, a cornerstone in understanding biomolecular ...
  77. [77]
    AlphaFold impact stories - Google DeepMind
    Scientists are using AlphaFold to accelerate research in nearly every field of biology. Over 2 million researchers in over 190 countries are using the AlphaFold ...<|separator|>
  78. [78]
    Reinvent 4: Modern AI–driven generative molecule design
    Feb 21, 2024 · REINVENT 4 is a modern open-source generative AI framework for the design of small molecules. The software utilizes recurrent neural networks and transformer ...<|control11|><|separator|>
  79. [79]
    survey of generative AI for de novo drug design - Oxford Academic
    In this paper, we explore the impacts and developments of ML-driven de novo drug design in two primary areas of research: molecule and protein generation.Introduction · Preliminary: generative AI... · Applications · Conclusion
  80. [80]
    Prospective de novo drug design with deep interactome learning
    Apr 22, 2024 · We present a computational approach utilizing interactome-based deep learning for ligand- and structure-based generation of drug-like molecules.
  81. [81]
    Transformative Role of Artificial Intelligence in Drug Discovery ... - NIH
    Aug 29, 2025 · This systematic review seeks to discuss how AI is being utilized in an aspect of drug development; from target identification, compound ...
  82. [82]
    The applications and advances of artificial intelligence in drug ...
    Artificial intelligence (AI) has emerged as a transformative force in healthcare, with applications spanning diagnostics to drug development.Review · 3.1. Policies And... · 3.2. Ai Applications In Drug...
  83. [83]
    Quarterly Industry Landscape Report - ASGCT
    The data show unequivocally that gene, cell, and RNA therapies continue to grow rapidly with nearly 3,500 therapies in preclinical and clinical development ...
  84. [84]
    RNA editing: Expanding the potential of RNA therapeutics - PMC
    RNA therapeutics have had a tremendous impact on medicine, recently exemplified by the rapid development and deployment of mRNA vaccines to combat the ...
  85. [85]
    The Limitless Future of RNA Therapeutics - Frontiers
    RNA therapeutics comprise a rapidly expanding category of drugs that will change the standard of care for many diseases and actualize personalized medicine.
  86. [86]
    Clinical advances in gene, cell, and RNA therapies
    May 20, 2025 · This special issue of Molecular Therapy features current advances and exciting developments as well as challenges in clinical gene, cell, and ...
  87. [87]
    Gene editing therapeutics based on mRNA delivery - ScienceDirect
    This review aims to summarize the therapeutic applications of gene editing based on mRNA delivery. We introduce gene editing therapeutics using mRNA.
  88. [88]
    Emerging New Drug Modalities in 2025 | BCG
    Oct 9, 2025 · BCG's third annual report assesses the growth of new modalities in 2025 and potential implications for the biopharmaceutical industry and ...
  89. [89]
    Advances in drug delivery systems, challenges and future directions
    Recent drug delivery systems (DDS) are formulated with improved properties such as smaller particle size, increased permeability, increased solubility, efficacy ...
  90. [90]
    Advances in nanoparticles in targeted drug delivery–A review
    This review examines emerging frontiers, medical applications, and developmental trends of nanoparticles in drug delivery.
  91. [91]
    Current approaches in smart nano‐inspired drug delivery - NIH
    Apr 24, 2024 · Current smart drug delivery approaches include nanoparticles, liposomes, micelles, and hydrogels, each with its own advantages and limitations.
  92. [92]
    Nanofiber Drug Delivery Systems: Recent Advances in ...
    May 6, 2025 · Nanofiber-based drug delivery systems show strong potential due to their high surface area-to-volume ratio and adjustable structure.
  93. [93]
    Global Regulatory Agencies Directory - Asphalion
    Explore Global Regulatory Agencies: Find Key Health Authorities Worldwide - A Comprehensive Interactive Map and Directory.
  94. [94]
    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 ...ICH Guideline page. · Quality Guidelines · History · Mission
  95. [95]
    ICH Guidelines
    ICH has produced a comprehensive set of safety Guidelines to uncover potential risks like carcinogenicity, genotoxicity and reprotoxicity.Quality Guidelines · Safety Guidelines · Index of Guidelines
  96. [96]
    International Regulatory Harmonization - FDA
    Jan 31, 2025 · ICH's mission is to achieve greater harmonization to ensure that safe, effective, and high-quality medicines are developed and registered in the ...
  97. [97]
    Drugs and Devices: Comparison of European and U.S. Approval ...
    This study explores some of the similarities and differences in European and US regulation of drugs and devices, and discusses challenges facing each.
  98. [98]
    Comparison of the Latin America Regulation Landscape and ...
    Sep 8, 2023 · LATAM regulations must adapt to new regulatory standards (FDA/EMA/ICH) through implementation of frequent updates, reliance/expedited ...
  99. [99]
    Comparison of drug approvals of the FDA and EMA between 2013 ...
    Jul 3, 2025 · The study examines differences in approval timelines, therapeutic areas, mechanisms of action, and the marketing authorisation holders for the period between ...Missing: variations | Show results with:variations
  100. [100]
    History - ICH
    A 25-year track record of successful delivery of harmonised guidelines for global pharmaceutical development as well as their regulation.
  101. [101]
    [PDF] ICH Overview - FDA
    Launched in 1990 by the US, EU, and Japan. Canada, Swissmedic and WHO as observers. • Well-defined objectives: – To improve efficiency of new drug development ...
  102. [102]
    Regulatory Harmonization Initiative - Regulation and Prequalification
    Progress towards enhancing regulatory practices related to medical products around the world is supported by capacity building, promoting regulatory convergence ...
  103. [103]
    Global Regulatory Harmonization Efforts in 2025 - Freyr Digital
    Apr 11, 2025 · This blog delves into the recent developments in global regulatory harmonization, highlighting key initiatives and their implications for life sciences ...
  104. [104]
    Editorial: The changing focus of regulatory frameworks around ... - NIH
    Jun 30, 2025 · Medicines regulatory harmonization provides several benefits including improving public health through faster availability of safe, high- ...
  105. [105]
    One-Third Of New Drugs Had Safety Problems After FDA Approval
    May 9, 2017 · Researchers at the Yale School of Medicine found that nearly a third of those approved from 2001 through 2010 had major safety issues.
  106. [106]
    Why Do Drugs in Development “Fail” in Clinical Trials?
    Dec 12, 2016 · In a high level analysis, most drugs were discontinued in Phase III due to inadequate efficacy (44 percent) or safety issues (24 percent). These ...
  107. [107]
    DICTrank: The largest reference list of 1318 human drugs ranked by ...
    Among 133 drugs withdrawn from the market between 1990 and 2010, 25 withdrawals (18.8%) were caused by cardiotoxicity, following hepatotoxicity (36 withdrawals, ...
  108. [108]
    Delays in the post-marketing withdrawal of drugs to which deaths ...
    Feb 5, 2015 · Of 81 products launched after 1950, 31 (38%) were withdrawn within 1 year of the first reported death, 12 (15%) within the second year, and 38 ( ...
  109. [109]
    Failure of Investigational Drugs in Late-Stage Clinical Development ...
    Oct 10, 2016 · Roughly half of investigational drugs entering late-stage clinical development fail during or after pivotal clinical trials, primarily because of concerns ...
  110. [110]
    Bias found when drug manufacturers fund clinical trials
    Oct 7, 2024 · Psychiatric drugs are reported to be about 50% more effective in clinical trials funded by the drug's manufacturer than when trials of the same drug are ...
  111. [111]
    Hidden conflicts? Pharma payments to FDA advisers after drug ...
    Jul 5, 2018 · Science investigation of journal disclosures and pharmaceutical funding records shows potential influence on physician gatekeepers.
  112. [112]
    Reporting bias in clinical trials: Progress toward transparency ... - NIH
    Jan 19, 2022 · Reporting bias can skew the perceived risk–benefit ratio of treatments, mislead medical professionals and policymakers, and ultimately result in ...
  113. [113]
    How Pharmaceutical Companies Price Their Drugs in the U.S.
    Ultimately, the main objective of pharmaceutical companies when pricing drugs is to generate the most revenue. This often means facing competition, which ...<|separator|>
  114. [114]
    US Drug Pricing and Reimbursement: Players, Payers, PBMs, and ...
    They negotiate discounts in the form of rebates with drug manufacturers, negotiate dispensing fees with pharmacies, and process prescription drug claims on ...
  115. [115]
    Comparing Prescription Drugs in the U.S. and Other Countries
    Jan 31, 2024 · In 2022, U.S. drug prices were nearly 2.78 times higher than other countries, with brand drugs at least 3.22 times higher. Most new drugs were ...
  116. [116]
  117. [117]
    Comparing U.S. and International Market Size and Average Pricing ...
    Research using 2022 data found that prescription drug prices in the U.S. were 2.78 times as high as prices in 33 other Organisation for Economic Co-operation ...
  118. [118]
    [PDF] 2023 r&d spending and intensity benchmarks - SAI Books
    The pharmaceutical industry continues to be the biggest R&D investor in fiscal 2023 with budgets of drug companies increasing by over 17% to $185 billion. ...
  119. [119]
    Analysis Finds Meaningful Impact on Pharmaceutical Innovation ...
    Feb 18, 2025 · For every 10% reduction in expected U.S. revenues, pharmaceutical innovation is expected to decline by up to 15%, finds a new Schaeffer ...
  120. [120]
    Striking a balance: Drug prices, profits and incentives for innovation
    Jun 4, 2019 · Higher prices increase the incentive for investment in new drug development, but also create affordability problems, resulting in less access to ...
  121. [121]
    Full article: Incentives and disincentives to drug innovation
    Drug innovation is strongly driven by economic incentives. How these incentives work in determining or changing the level of activity of innovators.
  122. [122]
    The Hidden Toll of Drug Price Controls: Fewer New Treatments and ...
    Jul 17, 2023 · Pharmaceutical price controls in 32 OECD countries, not including the United States, reduced manufacturer sales revenue by in 2018 77 percent, ...
  123. [123]
    Prices and Affordability of Essential Medicines in 72 Low-, Middle ...
    Aug 15, 2025 · However, it is estimated that essential medicines are unaffordable or unavailable to 1 in 4 people worldwide. As governments strive to achieve ...
  124. [124]
    Challenges to the Availability and Affordability of Essential ...
    Jun 13, 2023 · It is assumed that approximately 79% of all pharmaceuticals consumed in Africa are imported. This significantly raises healthcare costs and puts ...
  125. [125]
    Raising the Barriers to Access to Medicines in the Developing World
    This article illustrates how the pharmaceutical industry has convinced the US and the EU to impose data exclusivity on their trade partners, many of them ...
  126. [126]
    Pharma companies are taking steps to address access in low ...
    Nov 19, 2024 · Several pharmaceutical companies are prioritising low-income and least developed countries within their inclusive business models, ...
  127. [127]
    Prices, availability, and affordability of adult medicines in 54 low ...
    This paper aims to present an updated analysis of medicine affordability, availability, and pricing across 54 countries using the WHO–HAI method
  128. [128]
    [PDF] Incentives for Pharmaceutical Innovation: What's working ... - HAL
    Feb 10, 2023 · Much of the increased life expectancy realized during the 20th and 21st centuries can be credited to pharmaceutical interventions.<|separator|>
  129. [129]
    [PDF] Patent protection as a key driver for pharmaceutical innovation | IFPMA
    For pharmaceuticals, however, there is strong empirical evidence that patents have led to the socially desired result of higher R&D spending on developing new.
  130. [130]
    The Cost of Drug Development: How Much Does It Take to Bring a ...
    Sep 28, 2025 · 1. The average cost of developing a new prescription drug is approximately $2.6 billion. Developing a new drug is one of the most expensive ...
  131. [131]
    [PDF] The Effect of Intellectual Property Rights on Domestic Innovation in ...
    This paper analyses the causal impact of Intellectual Property Rights (IPR) on pharmaceutical innovation in a panel of 74 countries.
  132. [132]
    [PDF] Intellectual Property Rights and Pharmaceuticals - WIPO
    Not surprisingly, therefore, R&D activity in pharmaceuticals has historically been concentrated in countries with strong and enforceable intellectual property ...
  133. [133]
    Drug Patents: Essential Guide to Pharmaceutical Patent Protection
    Rating 4.9 (1,739) Jan 14, 2025 · Patents last 20 years from filing, but actual market exclusivity usually spans 7-12 years after factoring in development and FDA approval time.Extending Drug Patents: Smart... · Current Developments in Drug...
  134. [134]
    Frequently Asked Questions on Patents and Exclusivity - FDA
    Feb 5, 2020 · Periods of exclusivity and patent terms may or may not run concurrently. Exclusivity was designed to promote a balance between new drug ...Why does the exclusivity... · Where can I find patent and...
  135. [135]
    Unique Challenges for Patents in the Pharmaceutical Industry
    Feb 20, 2025 · Pharmaceutical patents face the challenge of limited timeframes, particularly given the lengthy development process. From discovery to FDA ...
  136. [136]
    [PDF] Doing R&D in Countries with Weak IPR Protection
    Multinationals are increasingly conducting R&D in countries such as India and. China, where intellectual property rights (IPR) protection is still very weak.
  137. [137]
    Drug patents: the evergreening problem - PMC - NIH
    When brand-name companies patent “new inventions” that are really just slight modifications of old drugs, it's called “evergreening.”
  138. [138]
    The Evergreening Myth - Cato Institute
    When offering descriptions and examples of evergreening, they focus on drug companies continuing to innovate after first introducing a new molecule, and on the ...
  139. [139]
    Biden Administration report debunks myths around patent ... - PhRMA
    Sep 3, 2024 · The study debunks claims that allege biopharmaceutical companies engage in “evergreening” when obtaining patents at different points in a product's life cycle.Missing: evidence | Show results with:evidence
  140. [140]
  141. [141]
    [PDF] IPRS, PHARMACEUTICALS AND FOREIGN DIRECT INVESTMENT
    The strength of IPR protection appears to be one important factor – among others – influencing pharmaceutical sector trade and investment decisions. As IPR ...