Fact-checked by Grok 2 weeks ago

Thalidomide


Thalidomide is a synthetic glutarimide initially developed in the early 1950s by the West German pharmaceutical company as a and agent lacking the associated with barbiturates. Marketed under names such as Contergan and Distaval starting in 1957 across Europe, , and other regions, it was promoted for treating insomnia and, crucially, nausea and vomiting in , with claims of safety for use by pregnant women based on limited that failed to reveal its teratogenic potential. Between 1957 and 1962, widespread use by pregnant women resulted in over 10,000 cases of severe congenital malformations, predominantly —a condition characterized by shortened or absent limbs—along with damage to eyes, ears, gastrointestinal, and cardiovascular systems, occurring when the drug was ingested during a narrow 20- to 36-day window post-fertilization when embryonic limb buds form. The causal link was established through epidemiological investigations, notably by obstetrician McBride and others, prompting swift market withdrawals by 1961-1962 and catalyzing rigorous global drug safety regulations, including mandatory efficacy and safety proofs. Despite this catastrophe, thalidomide's immunomodulatory and anti-angiogenic properties—later elucidated via inhibition of tumor necrosis factor-alpha and cereblon-mediated protein degradation—led to its repurposing in the 1980s-1990s for in patients and, following phase II trials demonstrating responses in refractory cases, FDA approval in 2006 for newly diagnosed in combination therapies, where it enhances . Its dual legacy underscores the perils of inadequate preclinical testing juxtaposed against empirical rediscovery of therapeutic utility under strict risk-management protocols like iPLEDGE to prevent fetal exposure.

Pharmacology and Chemistry

Chemical Structure and Properties

Thalidomide is a synthetic derivative of , featuring a ring fused to a glutarimide moiety via a chiral carbon atom at the 3-position of the ring. Its IUPAC name is 2-(2,6-dioxopiperidin-3-yl)-2,3-dihydro-1H-isoindole-1,3-dione, and it exists as a of (R)- and (S)-enantiomers due to the stereogenic center at the ring. The molecular formula is C13H10N2O4, with a molecular weight of 258.23 g/. Physically, thalidomide appears as a white to off-white crystalline powder with a of 269–271 °C. It exhibits low solubility in , approximately 60 mg/L at 25 °C, rendering it poorly water-soluble, but it is soluble in organic solvents such as (DMSO) and acetone. The compound is lipophilic, with a value of approximately 0.09, contributing to its profile despite the aqueous insolubility. Chemically, thalidomide is stable under neutral conditions but susceptible to in acidic or basic environments, potentially cleaving the bonds. It undergoes spontaneous in vivo due to the acidic proton at the chiral , interconverting between enantiomers even after administration of a single stereoisomer. The and glutarimide rings confer rigidity to the molecule, influencing its binding interactions in biological systems.

Mechanism of Action

Thalidomide's pharmacological effects arise from multiple interconnected pathways, with a central mechanism involving binding to (CRBN), the substrate receptor of the Cullin-Ring E3 complex CRL4^{CRBN}. This binding alters the complex's substrate specificity, promoting the ubiquitination and proteasomal degradation of specific neosubstrate proteins, thereby modulating cellular protein homeostasis. The drug exhibits higher affinity for CRBN with its (S)- compared to the (R)-, though rapid in vivo limits enantiomer-specific effects. In immunomodulatory contexts, such as treatment of leprosum, thalidomide selectively inhibits the production of pro-inflammatory cytokines, particularly tumor necrosis factor-alpha (TNF-α) from lipopolysaccharide-stimulated monocytes, while having lesser effects on interleukin-6 or interleukin-10. This cytokine modulation stems partly from CRBN-mediated degradation of transcription factors like Ikaros family zinc finger 1 () and IKZF3, which represses genes involved in inflammation and enhances T-cell activation via increased interleukin-2 production. Additionally, thalidomide shortens the of (COX-2) mRNA in a dose-dependent manner, contributing to activity. For anti-neoplastic applications, such as multiple myeloma, CRBN engagement recruits IKZF1 and IKZF3 for degradation, disrupting myeloma cell proliferation by downregulating interferon regulatory factor 4 (IRF4) and c-Myc expression, while activating immune responses through B- and T-cell modulation. Thalidomide also demonstrates anti-angiogenic properties by inhibiting endothelial cell proliferation and basic fibroblast growth factor-mediated vascularization, potentially via bioactive metabolites generated through spontaneous hydrolysis at physiological pH, which yields over 20 compounds. These metabolites further suppress angiogenesis and contribute to therapeutic efficacy in hypoxic tumor environments. The sedative-hypnotic effects originally attributed to thalidomide remain incompletely elucidated but are associated with the (R)-enantiomer's interaction with targets, distinct from its immunomodulatory actions. Overall, thalidomide's pleiotropic mechanisms reflect its influence on ubiquitin-proteasome pathways, networks, and vascular biology, though the relative contributions vary by context and underscore the drug's non-specific protein degradation induction.

Chirality, Racemization, and Teratogenic Mechanisms

Thalidomide contains a single center at the carbon atom connecting the and glutarimide rings, yielding two s: (R)-thalidomide and (S)-thalidomide. The (R)- exhibits primarily and properties, whereas the (S)- is implicated in teratogenic effects, though this distinction arises from studies and does not fully account for behavior. Despite early proposals to market only the (R)- to mitigate risks, the molecule's inherent precludes this approach. In physiological conditions, thalidomide undergoes rapid through epimerization at the chiral center, primarily via proton abstraction facilitated by the adjacent and groups, with water molecules potentially aiding the process. This interconversion occurs in body fluids and tissues, equilibrating to a regardless of initial enantiomeric purity, with reported half-lives for racemization on the order of minutes in aqueous media at neutral . Computational studies identify key pathways involving of the chiral carbon, leading to a planar intermediate that reprotonates non-stereospecifically, underscoring the futility of enantiopure administration for avoiding adverse effects. The teratogenic mechanisms of thalidomide remain incompletely resolved, but empirical evidence implicates disruption of embryonic angiogenesis as a primary pathway, wherein the drug inhibits fibroblast growth factor 2 (FGF-2) signaling and ceramide metabolism, resulting in regression of immature blood vessels critical for limb development. Binding to cereblon (CRBN), a substrate receptor in the Cullin-Ring E3 ubiquitin ligase complex, thalidomide alters protein degradation, downregulating transcription factors like SALL4 and preventing normal ubiquitination, which correlates with phocomelia and other malformations in susceptible species during a narrow gestational window (days 20-36 post-conception in humans). Additional hypotheses include free radical-mediated oxidative damage to embryonic DNA and proteins, though these lack direct causal validation compared to the CRBN and anti-angiogenic models. Species-specific resistance, evident in rodents, stems from differential CRBN binding affinity and downstream signaling, highlighting thalidomide's targeted disruption of human developmental pathways.

Approved Medical Uses

Treatment of Leprosy Reactions

Thalidomide is primarily indicated for the management of erythema nodosum leprosum (ENL), an immune-mediated type II reaction occurring in multibacillary leprosy patients, characterized by painful erythematous nodules, fever, and potential nerve damage. It serves as a steroid-sparing agent, particularly effective for recurrent or steroid-resistant ENL episodes, with typical dosing starting at 100–300 mg daily, tapered based on response. Controlled trials from the 1960s onward demonstrated rapid symptom amelioration, often within days, outperforming alternatives like aspirin or pentoxifylline in reducing skin lesions and systemic inflammation. The drug's efficacy in ENL stems from its immunomodulatory action, selectively inhibiting tumor factor-alpha (TNF-α) and production by monocytes, a key driving the response in reactions. This mechanism addresses the underlying immune complex deposition and infiltration without broadly suppressing adaptive immunity, unlike corticosteroids. In a double-blind , thalidomide achieved complete resolution in most patients versus partial or no response with , though relapse rates post-discontinuation necessitate maintenance strategies. Regulatory approval for ENL followed compassionate use programs initiated in the mid-1960s, after initial withdrawal due to teratogenicity, with formal U.S. (FDA) approval granted on July 16, 1998, under the brand Thalomid for this indication. Usage requires in programs like the System for Thalidomide and Prescribing (STEPS) to enforce contraception and , given persistent teratogenic risks even in patients via . Recent studies confirm its viability in outpatient settings in leprosy-endemic areas, with high adherence when combined with counseling, though alternatives like are explored for long-term control to minimize relapses.

Multiple Myeloma Therapy

Thalidomide was first demonstrated to have antitumor activity against refractory in a phase 2 trial published in 1999, where 84 patients received 200 to 800 mg daily, achieving a 25% overall response rate (including 2% complete responses) lasting a median of 1 year, even in those previously treated with high-dose chemotherapy and stem-cell transplantation. This unexpected efficacy prompted further investigation into its immunomodulatory properties, including inhibition of tumor necrosis factor-alpha production, anti-angiogenic effects via blockade of , and enhancement of activity, which collectively disrupt myeloma cell proliferation and microenvironment support. The U.S. approved thalidomide (as Thalomid) on October 26, 2006, for use in combination with dexamethasone for newly diagnosed , based on phase 3 trials showing improved response rates and compared to dexamethasone alone. Standard induction regimens typically involve 200 mg oral thalidomide daily with dexamethasone 40 mg on days 1-4, 9-12, and 17-20 every 28 days for four cycles, often followed by autologous stem-cell transplantation. In maintenance settings post-transplantation, low-dose thalidomide (100-200 mg daily) has been shown to extend by 15-18 months in randomized trials, though overall survival benefits vary and are tempered by increased risks of and . Combination therapies incorporating thalidomide, such as bortezomib-thalidomide-dexamethasone (VTD), yield high response rates exceeding 90% in transplant-eligible patients, positioning it as a viable induction option outside clinical trials, particularly in regions where newer agents like are less accessible. However, thalidomide's role has diminished with the advent of more potent analogues like , which offer comparable efficacy with reduced , though thalidomide remains relevant for cost-sensitive or specific cases. Long-term data from meta-analyses confirm its contribution to deepened responses but highlight the need for thromboprophylaxis due to a 2- to 4-fold elevated venous risk in combination regimens.

Other FDA-Approved or Investigational Uses

Thalidomide has received FDA designation for investigational treatment of several rare conditions, including , , Kaposi sarcoma, HIV-associated wasting syndrome, , and , reflecting its evaluation in clinical settings under strict safety protocols due to teratogenic risks. In hematologic disorders beyond , phase II trials have shown thalidomide to induce erythroid responses in myelodysplastic syndromes, durable responses in approximately 28% of myelofibrosis cases, and response rates up to 72% when combined with rituximab in . For solid tumors, phase II studies reported a 90% decline in levels in metastatic castration-resistant and partial responses in advanced . These applications leverage thalidomide's anti-angiogenic and immunomodulatory effects, though efficacy remains unproven for regulatory approval. In chronic following , thalidomide has demonstrated response rates of 20% to 79% as salvage therapy in refractory or high-risk cases, outperforming alternatives like cyclosporine or in some models, but prophylactic use has paradoxically increased incidence and mortality. For HIV-related complications, randomized trials confirmed benefits in wasting , aphthous ulcers, and , attributed to its and appetite-stimulating properties. Dermatologic and autoimmune applications include effective control of mucocutaneous lesions in Behçet's at doses of 100 mg daily, as well as investigational use in cutaneous , , and . Additional explorations encompass , , and cancer , with variable pilot study outcomes.

Adverse Effects and Contraindications

Teratogenic Risks and Pregnancy Prevention

Thalidomide is a potent human teratogen that induces severe congenital malformations when administered during early pregnancy, with risks including phocomelia (shortened or absent limbs), amelia, hypoplasia of limbs, ocular defects such as microphthalmia, anotia or microtia of the ears, congenital heart defects, and gastrointestinal atresias. The drug's teratogenic effects are highly specific to the developmental window of approximately days 20 to 36 post-fertilization, during which limb buds form and are vulnerable to disruption, particularly via inhibition of angiogenesis that starves developing tissues of blood supply. Exposure during this period carries an estimated 40-50% risk of major birth defects, with outcomes ranging from isolated limb reductions to multi-system anomalies, and nearly any organ or tissue potentially affected depending on timing and dose. Due to these irreversible risks, thalidomide is absolutely contraindicated in pregnancy and classified as FDA Pregnancy Category X, meaning the potential benefits do not justify the fetal harm. In the United States, where thalidomide was approved in 1998 solely for treating erythema nodosum leprosum under strict controls, distribution occurs exclusively through the THALOMID Risk Evaluation and Mitigation Strategy (REMS) program, formerly the System for Thalidomide Education and Prescribing Safety (STEPS), to prevent fetal exposure. Prescribers must register with the program, educate patients on risks, and verify compliance; female patients of childbearing potential require two negative pregnancy tests (serum and urine) prior to starting therapy, monthly tests thereafter, and a final test 4 weeks post-discontinuation, alongside mandatory use of effective contraception (e.g., two forms, including one highly effective method) starting one month before, during, and one month after treatment. The REMS program mandates patient enrollment via a signed acknowledging teratogenic dangers, with dispensing limited to certified pharmacies that confirm test results and contraceptive adherence before releasing no more than a 28-day supply. Men must use condoms during and for 4 weeks after, as thalidomide in semen poses theoretical risks, though evidence of paternal transmission causing defects is lacking. Non-compliance, such as missed tests or inadequate contraception, results in treatment interruption; since implementation, U.S. fetal exposures have been minimized, though global surveillance continues via pregnancy registries to monitor rare breakthroughs and long-term outcomes in exposed . These measures reflect empirical lessons from the 1950s-1960s crisis, where inadequate preclinical testing failed to detect species-specific teratogenicity absent in but evident in and humans.

Neurological and Other Non-Teratogenic Effects

Thalidomide commonly induces , a dose-dependent sensorimotor axonal neuropathy that primarily affects longer nerves in a length-dependent manner. Symptoms typically include paresthesias, numbness, tingling, and pain in the extremities, often emerging after cumulative doses exceeding 50 grams or prolonged treatment beyond 6-12 months, with incidence rates reported from under 1% at low doses to over 70% in extended use. Electrophysiological studies reveal symmetrical sensory deficits, with motor involvement less frequent, and pathologic examination shows axonal degeneration without prominent demyelination. Neuropathy may persist or worsen after discontinuation, necessitating baseline and periodic neurologic monitoring, including nerve conduction studies, during therapy. Central nervous system effects include , occurring in up to 50-87% of patients, often dose-related and prominent early in treatment. and accompany these, contributing to impaired daily functioning, while less common manifestations involve or vertigo. These effects stem from thalidomide's modulation of neurotransmitters and sedative properties, observed across indications like and reactions. Gastrointestinal adverse effects feature prominently, with constipation affecting up to 100% of users, ranging from mild to severe and linked to autonomic inhibition. Other non-neurological effects include (up to 30%), , and weight gain, alongside hematologic risks such as or . Thromboembolic events, including and , occur at higher rates, particularly in myeloma patients, prompting prophylaxis with anticoagulants. Rare but serious reactions encompass , seizures, and , underscoring the need for vigilant monitoring beyond teratogenic precautions.

Drug Interactions and Overdose

Thalidomide demonstrates limited pharmacokinetic interactions with coadministered drugs, as it undergoes primarily non-enzymatic in and rather than P450-mediated , reducing the likelihood of alterations in drug levels via CYP inhibition or induction. Pharmacodynamic interactions predominate, including additive (CNS) depression—manifesting as enhanced , , or —when thalidomide is combined with other CNS depressants such as , barbiturates (e.g., , ), benzodiazepines, opioids, or antipsychotics like . This additive effect arises from thalidomide's inherent properties, which affect up to 30% of patients at therapeutic doses. Thalidomide also heightens the risk of venous thromboembolism (VTE), including deep vein thrombosis and , particularly in patients receiving it alongside dexamethasone or other prothrombotic agents like or ; incidence rates can exceed 10-20% in such combinations without prophylaxis. Concomitant use with hormonal contraceptives requires caution due to thalidomide's teratogenicity, though it does not impair their efficacy; non-hormonal methods or highly effective options are recommended under risk programs like iPLEDGE. Limited clinical data suggest potential induction of metabolism or cyclosporine clearance in a dose-dependent manner, though this has not been confirmed as clinically significant in large trials. In overdose scenarios, thalidomide exhibits a wide with low acute lethality; reported cases typically present with exaggerated therapeutic effects such as profound drowsiness, , , , or gastrointestinal upset rather than organ failure. No specific exists, and management focuses on supportive care: monitoring , ensuring airway patency, administering activated charcoal if was recent (within 1-2 hours), and providing intravenous fluids for . is ineffective due to thalidomide's high protein binding (55-66%) and large . Fatal outcomes are rare and generally linked to comorbidities or rather than thalidomide alone, with recovery expected upon discontinuation and supportive intervention.

Historical Development

Initial Synthesis and Pre-Marketing Testing (1950s)

Thalidomide was first synthesized in 1953 by chemists at the German pharmaceutical company , with and Heinrich Mückter officially credited as the inventors. The compound, chemically α-(N-phthalimido)glutarimide, was developed as a potential non- and , sought after amid concerns over barbiturate addiction and overdose risks prevalent in the early . pursued its evaluation following initial efforts, designating it internally as K-17 during early pharmacological assessments. From 1953 to 1956, pre-marketing testing focused primarily on acute and subacute in , including mice, rats, rabbits, , and across multiple strains and breeds. These studies reported no lethal effects even at doses exceeding 10 grams per kilogram in , leading to conclude the drug possessed an exceptionally high safety margin compared to existing . The first published pharmacological paper on thalidomide appeared in 1956, highlighting its properties without detailing reproductive or teratogenic evaluations. Reproductive toxicity testing in pregnant animals was not conducted, reflecting the era's regulatory norms in , where manufacturers bore no obligation to prove safety prior to market entry and placental drug transfer to fetuses was not routinely considered. models used, predominantly , failed to reveal teratogenic potential due to species-specific metabolic differences that rendered thalidomide less embryotoxic in those . Limited human volunteer trials occurred internally at , involving company employees who reported effective without adverse effects, further bolstering confidence for . By late 1956, these findings supported filings, including a patent granted to in 1957, paving the way for marketing as Contergan in 1957.

Global Marketing and Early Adoption (1957–1961)

Thalidomide was first marketed by the West German pharmaceutical company Chemie Grünenthal GmbH under the trade name Contergan, launching on October 1, 1957, in Germany as a non-barbiturate sedative for treating insomnia and anxiety. The drug was promoted aggressively through advertising campaigns emphasizing its safety profile, claiming it was non-toxic even at high doses and suitable for a broad range of users, including children and pregnant women for alleviating morning sickness. Grünenthal positioned Contergan as superior to existing sedatives, highlighting its lack of hangover effects and low addiction potential, which facilitated rapid uptake among physicians and consumers in post-war Europe seeking reliable pharmaceutical relief. By 1958, had licensed thalidomide to international partners, enabling its adoption across Europe and beyond; in the , it was introduced as Distaval by in May 1958, while and saw market entry the same year, followed by in 1959. Marketing materials in these countries mirrored German campaigns, touting the drug's efficacy for and without adequate emphasis on potential risks, leading to widespread prescription for pregnant women. Over-the-counter availability in some markets, combined with physician endorsements, drove early sales volumes exceeding millions of tablets annually in alone by 1959. Expansion continued into 1960–1961, with thalidomide reaching approximately 46 countries through licensing agreements with subsidiaries and firms like Richardson-Merrell in the Americas, though U.S. approval was pending. Adoption was particularly strong in Western Europe, Australia, and parts of Asia, where it was integrated into routine obstetric care for hyperemesis gravidarum, reflecting pharmaceutical industry's push for novel therapeutics amid limited regulatory scrutiny. Grünenthal's global strategy relied on minimal pre-marketing toxicity data, primarily from rodent studies that failed to reveal teratogenic effects, fostering confidence in its safety for human use.

Emergence of Birth Defects and Causal Factors (1961–1962)

In late 1961, German pediatrician Widukind Lenz identified a striking pattern of severe congenital limb malformations, including phocomelia characterized by shortened or absent proximal limbs resembling flippers, among newborns in West Germany. After observing over 20 such cases, Lenz systematically questioned the mothers and found that approximately 80% had taken thalidomide (marketed as Contergan) during the first trimester of pregnancy, particularly between days 30 and 45 after the last menstrual period. This temporal association suggested a direct causal role, as the malformations aligned with the drug's inhibition of fetal limb bud vascularization and development during the critical embryogenic window of days 20 to 36 post-conception, when mesenchymal cells proliferate and chondrogenesis initiates. Lenz alerted Chemie Grünenthal, the manufacturer, on November 15, 1961, prompting an internal investigation that confirmed the link through additional case reviews, leading to the drug's withdrawal from the German market on November 26, 1961. Independently, Australian obstetrician William McBride noted similar defects in Sydney deliveries starting in May 1961, including a newborn with bilateral arm malformations and internal anomalies that died shortly after birth. Observing a subsequent cluster, McBride hypothesized thalidomide's involvement based on maternal histories and published a concise letter in The Lancet on December 16, 1961, reporting that 20% of queried mothers of affected infants had used the drug, contrasting with negligible use among controls, and urging immediate scrutiny. This observation reinforced the causal inference through epidemiological clustering, as thalidomide's sedative and antiemetic effects led to its widespread prescription for morning sickness, exposing fetuses to peak teratogenic risk during organogenesis. By early 1962, confirmatory reports proliferated across and , with Lenz publishing detailed analyses in linking over 50 cases to thalidomide exposure strictly within the 34- to 50-day gestational window, excluding sporadic unrelated to the drug. The causal mechanism, later elucidated as thalidomide's enantiomer-specific interference with cereblon-mediated ubiquitination disrupting and limb patterning genes like SALL4, was initially inferred from dose-response patterns: higher cumulative exposure correlated with more severe reductions, while post-critical-period use yielded no defects. Pre-marketing had failed to detect teratogenicity due to insensitive species (e.g., rats) and improper dosing timing, underscoring how human-specific vulnerability during early formation evaded detection until post-marketing surveillance revealed the . These findings prompted voluntary withdrawals in , , and other nations by mid-1962, though delays in regulatory action elsewhere amplified the toll.

The Thalidomide Crisis

Scale and Nature of Birth Defects

Thalidomide exposure during , primarily between 1957 and 1962, resulted in an estimated 10,000 to 12,000 children born worldwide with severe congenital malformations, with approximately half of affected ending in or neonatal death. The drug's teratogenic effects were most pronounced when taken between days 20 and 36 post-conception, a critical window for limb bud development, leading to a characteristic pattern of defects rather than random anomalies. The majority of cases occurred in , where thalidomide was marketed as a and ; reported the highest incidence with around 5,000 to 6,000 affected births, followed by the (approximately 2,000 cases) and other countries including , , and . In the United States, where the drug was not approved for marketing, only about 17 confirmed cases arose from limited investigational use. saw a smaller cluster in the early before withdrawal, though long-term exposure persisted in some regions due to delayed bans. The hallmark defects were limb reductions, affecting up to 80% of survivors, ranging from —where hands or feet protrude directly from the trunk due to absence of intervening long bones—to (complete limb absence) and micromelia (underdeveloped limbs). Upper limbs were more frequently and severely impacted than lower ones, often bilaterally, with thumbs commonly hypoplastic or absent. Beyond limbs, thalidomide caused a spectrum of internal and sensory malformations, including or (external ear absence or underdevelopment) with associated conductive in about 20-30% of cases; ocular defects such as , , or ; and visceral anomalies like , cardiac septation defects, , and genital malformations. Neurological effects, including in utero, were rarer but contributed to long-term morbidity among survivors. These patterns stemmed from thalidomide's disruption of embryonic and protein degradation pathways, sparing the but targeting rapidly vascularizing tissues.

Role of Testing Deficiencies and Marketing Practices

Pre-clinical testing of thalidomide by prior to its 1957 market launch was limited and failed to adequately assess . Standard tests on such as mice and rats administered during did not reveal teratogenic effects, as these proved insensitive to the drug's impact on limb development at typical doses. However, subsequent investigations post-crisis demonstrated that thalidomide induced in sensitive like white rabbits and certain when given during critical embryonic periods, highlighting the inadequacy of relying solely on models without broader testing or dose-escalation protocols for developmental endpoints. Grünenthal's protocols omitted routine evaluation of pregnant animals, a regulatory gap at the time, and lacked comprehensive studies in humans before widespread distribution. Human safety data prior to approval consisted primarily of short-term trials on approximately 20 volunteers and limited physician-supervised use, without systematic monitoring for fetal exposure risks. The company proceeded to market despite emerging reports of in users by 1959, prioritizing commercial rollout over extended . This testing shortfall contributed directly to the undetected teratogenic window—days 34 to 50 of —allowing unchecked prescriptions during peak vulnerability. Marketing practices amplified the crisis through aggressive promotion without pregnancy contraindications. Launched as Contergan in on October 1, 1957, thalidomide was advertised as a non-barbiturate safer than existing options, explicitly touted for insomnia, anxiety, and in pregnant women, with claims of harmlessness even in overdose. licensed the drug to over 40 firms across 46 countries by 1960, enabling rapid global exceeding 10 million prescriptions annually, often as an over-the-counter remedy in without mandatory warnings. materials and outreach emphasized its "ideal" profile for expectant mothers, downplaying any adverse signals, while internal documents later revealed suppression of early neuropathy complaints and Australian reports of malformed infants as early as 1960. This combination of deficient scrutiny and promotional overreach delayed recognition of causality, resulting in an estimated 10,000 affected births before withdrawals began in late 1961.

Country-Specific Responses and FDA's Non-Approval

In contrast to many other nations, the () never granted marketing approval for thalidomide, averting a large-scale domestic tragedy. In September 1960, pharmaceutical firm Richardson-Merrell submitted a for the drug under the proposed brand name Kevadon, seeking approval for use as a and for . medical reviewer , a pharmacologist and , rejected the application multiple times, citing inadequate and inconsistent safety data, including unresolved reports of in trial participants and a lack of studies on the drug's effects during or on fetal . Despite repeated pressure from Merrell—including threats of professional repercussions and attempts to bypass her review—Kelsey upheld her position, demanding comprehensive long-term human safety evidence that was never adequately provided. This stance, grounded in the 1938 Federal Food, Drug, and Cosmetic Act's safety requirements, limited U.S. exposure primarily to investigational use in clinical trials affecting fewer than 20,000 individuals, resulting in only 17 documented cases of thalidomide-related birth defects. West Germany, the origin of thalidomide's commercial development, saw the swiftest initial regulatory response once defects emerged. , which synthesized and marketed the drug as Contergan starting in 1957, internally documented rising birth defect reports by mid-1961 but delayed public action amid sales of over 10,000 daily prescriptions. Following scrutiny from pediatrician Widukind Lenz, who linked the drug to in December 1961, withdrew Contergan from the market on November 26, 1961, prompting German health authorities to endorse the halt. In the , where Biochemicals marketed thalidomide as Distaval from 1958, withdrawal followed closely after Germany's. After Lenz's warnings reached UK physicians and media reports surfaced in late November 1961, Distillers suspended sales on December 2, 1961, with the UK Ministry of Health issuing an urgent recall and advisory against further prescriptions by December 1961. Australia's response mirrored the 's timeline but highlighted enforcement gaps. launched thalidomide in 1960, and following the UK suspension, it withdrew the product in December 1961; however, Australian regulators made limited efforts to remove existing stocks from pharmacies promptly, allowing some continued distribution into 1962. Canada exhibited a delayed reaction despite early awareness. Approved for marketing in the late 1950s, thalidomide remained available until approximately March 1962—three months after withdrawals in Germany and the —due to slower coordination between federal health officials and pharmaceutical firms, contributing to over 100 affected survivors.
CountryInitial MarketingWithdrawal Date
West Germany1957November 26, 1961
United Kingdom1958December 2, 1961
Australia1960December 1961
CanadaLate 1950sMarch 1962
United StatesNot approvedN/A
By the end of 1961, thalidomide had been pulled from markets in most affected European countries, though global enforcement varied, with some nations like Japan and Brazil retaining availability longer into the 1960s absent strict bans. These responses underscored pre-crisis regulatory differences, where many jurisdictions relied on manufacturer self-reporting rather than independent verification, contrasting the FDA's precautionary model.

Regulatory Consequences

Pre-Crisis Regulatory Frameworks

In the United States, drug regulation before the thalidomide crisis operated under the 1938 Federal Food, Drug, and Cosmetic Act, which mandated that manufacturers file a (NDA) providing evidence of safety through animal and limited human studies, but imposed no requirement to prove efficacy. The (FDA) reviewed submissions within a 60-day window and could issue a notice objecting to marketing if deficiencies were found; otherwise, the drug was automatically deemed safe for interstate commerce and could proceed to market without explicit agency approval. This passive approval mechanism relied heavily on manufacturers' self-reported data, with minimal federal mandates for testing in animals or long-term human trials, reflecting a regulatory philosophy prioritizing industry innovation over stringent pre-market gatekeeping. European regulatory frameworks in the 1950s were fragmented, lacking a unified system and often deferring to national authorities with varying standards of oversight. In West Germany, where Chemie Grünenthal synthesized and first marketed thalidomide as Contergan in 1957, licensing by medical authorities permitted over-the-counter sales after rudimentary clinical observations, without compulsory preclinical teratogenicity studies or independent verification of safety claims. The United Kingdom's Committee on Safety of Drugs, established informally in the late 1950s, reviewed applications like Distaval (thalidomide's British brand, approved in 1958) based on sponsor-submitted data, but enforced no standardized requirements for animal reproduction tests or controlled trials, enabling rapid dissemination across pharmacies. In Canada, thalidomide entered distribution in 1959 via samples before formal authorization in 1961 by the Food and Drug Directorate, under a voluntary compliance model that similarly omitted rigorous efficacy or birth defect risk assessments. These pre-crisis regimes across jurisdictions emphasized safety assurances from manufacturers over proactive evidentiary burdens, often accepting anecdotal clinical reports and short-term sedation efficacy data for sedatives like thalidomide, while risks observed in early users prompted insufficient scrutiny. Absent harmonized standards or mandatory post-marketing , approvals hinged on discretion, facilitating thalidomide's expansion to over 40 countries by despite incomplete pharmacological profiling.

Post-Crisis Reforms: Kefauver-Harris Amendment and Global Changes

The thalidomide crisis accelerated the passage of the Kefauver-Harris Amendments to the Federal Food, Drug, and Cosmetic Act, signed into law by President on October 10, 1962. These amendments mandated that drug manufacturers prove both safety and for new drugs via adequate and well-controlled clinical investigations, shifting from the prior standard that required only safety demonstrations. The FDA gained authority to demand efficacy evidence for pre-1962 marketed drugs and to withdraw approvals lacking substantial supporting data. Further provisions required from human subjects in investigational trials and obligated manufacturers and investigators to report adverse drug experiences to the FDA in a timely manner. The amendments also restricted promotional claims to approved uses and enhanced FDA oversight of manufacturing practices, thereby expanding the agency's pre- and post-market regulatory powers. Internationally, the crisis prompted reviews of pharmaceutical licensing and spurred stricter controls on drug approvals. In , it contributed to the 1965 adoption of Council Directive 65/65/EEC, which harmonized requirements for marketing authorizations based on assessments of quality, , and efficacy through scientific data. In the , authorities established the Committee on Safety of Drugs in 1964 to evaluate potential hazards, laying groundwork for the comprehensive that centralized licensing and emphasized pre-market testing. These reforms, alongside emerging protocols, aimed to mitigate risks from inadequate testing and rapid global distribution, influencing bodies like the to promote standardized monitoring.

Benefits and Criticisms of Stricter Regulations

The Kefauver-Harris Amendments of 1962, enacted in direct response to the thalidomide crisis, mandated that pharmaceutical manufacturers demonstrate both safety and efficacy through "adequate and well-controlled investigations" prior to FDA approval, shifting from prior standards that emphasized only safety after the 1938 Food, Drug, and Cosmetic Act. This reform required explicit FDA marketing authorization and enhanced requirements for in clinical trials, thereby institutionalizing rigorous pre-market testing to avert widespread harm from inadequately vetted drugs. Globally, analogous changes followed, such as the establishment of the UK's Committee on Safety of Drugs in 1963, which enforced stricter and licensing, contributing to fewer instances of mass drug-induced teratogenicity in subsequent decades. These measures demonstrably elevated baseline drug safety; for instance, post-1962 protocols have correlated with a decline in severe adverse events akin to thalidomide's outbreaks, as manufacturers now conduct extensive studies absent in the 1950s. Stricter regulations have preserved public trust in pharmaceuticals by mandating reporting and post-market surveillance, enabling earlier detection of risks like those from , which was restricted after 1971 linkage to vaginal cancers. Empirical assessments indicate that these frameworks prevented an estimated thousands of thalidomide-equivalent tragedies by prioritizing causal evidence from controlled trials over anecdotal marketing claims, fostering a culture of empirical validation over commercial expediency. However, FDA-affiliated sources may underemphasize trade-offs, as institutional incentives favor highlighting safety gains while downplaying innovation barriers. Critics argue that these reforms imposed substantial compliance burdens, extending average timelines from 2-3 years pre-1962 to over 10 years by the and escalating costs from tens of millions to billions of dollars per approval, deterring in marginal but viable therapies. New applications in the fell sharply post-1962, from approximately 50 novel chemical entities annually in the late to around 20 in the , evidencing a quantifiable suppression of as firms prioritized high-return blockbusters over niche drugs. This "" delayed access to beneficial agents like beta-blockers for , approved years later than in , potentially costing lives through deferred treatments. Moreover, the mandate retroactively invalidated dozens of pre-1962 drugs lacking modern , removing effective options like certain analgesics from markets despite historical , as profitability waned under reclassification pressures. Economic analyses contend that while safety thresholds rose, the net societal risk may have increased via fewer therapeutic alternatives, with market incentives alone—bolstered by liability laws—sufficient to avert most disasters without such rigidity. Libertarian-leaning critiques, such as those from the , highlight overregulation's role in inflating consumer prices and stifling competition, though these overlook thalidomide's demonstration of informational asymmetries where firms underinvested in teratogenicity testing absent mandates. Balancing these, causal realism underscores that regulations curbed acute hazards but introduced chronic delays, with empirical trade-offs varying by disease severity—favoring caution for elective drugs but risking harm for unmet needs in or rare diseases.

Revival and Modern Applications

Rediscovery for Leprosy (1960s–1980s)

In 1964, Israeli dermatologist Jacob Sheskin administered thalidomide as a to a at Hadassah University Hospital in who was experiencing severe erythema nodosum leprosum (ENL), a painful inflammatory complication of unresponsive to standard treatments like steroids and arsenicals. The exhibited rapid resolution of fever, skin lesions, and systemic symptoms within days, prompting Sheskin to test it on additional with recurrent ENL reactions, where similar dramatic improvements were observed, far exceeding prior therapies. This serendipitous finding, reported in by 1965, highlighted thalidomide's potent and immunomodulatory effects on ENL, despite its known teratogenicity, leading to controlled trials under strict protocols to avoid exposure. Following Sheskin's observations, the (WHO) initiated a in 1967 evaluating thalidomide for ENL management, confirming its superior efficacy over alternatives in reducing reaction severity and duration, with response rates approaching 90% in acute episodes. By the early , limited compassionate use programs emerged in countries like and , where leprosy prevalence was high, administering thalidomide to multibacillary patients under male-only or postmenopausal restrictions and rigorous contraception monitoring to mitigate risks. Studies through the decade documented sustained benefits, including prevention of chronic ENL recurrence at doses of 100–300 mg daily, though side effects like necessitated periodic discontinuation. In the 1980s, accumulating evidence from global trials solidified thalidomide's role as a cornerstone for ENL, with publications demonstrating its ability to suppress and other cytokines driving lepra reactions, outperforming corticosteroids in long-term control without equivalent steroid-induced complications like . Despite regulatory bans elsewhere, organizations like WHO endorsed its targeted use in leprosy-endemic regions, distributing it via specialized programs that emphasized risk mitigation, influencing policy in over 20 countries by decade's end. This revival underscored thalidomide's mechanistic value in immune-mediated inflammation, paving the way for broader applications while highlighting the need for in vulnerable populations.

Oncology Approvals and Efficacy Data (1990s–Present)

In the 1990s, thalidomide's potential anticancer effects were explored based on its inhibition of tumor necrosis factor-alpha and anti-angiogenic properties, leading to phase II trials in refractory (MM) that reported objective response rates of approximately 25-30% with median survivals of 12-14 months. Early compassionate use began around 1997 for relapsed or refractory MM patients unresponsive to standard therapies. These findings prompted expanded investigations, though formal approvals for lagged behind leprosy indications. The U.S. granted accelerated approval on May 25, 2006, for thalidomide (Thalomid) in combination with dexamethasone as initial therapy for newly diagnosed , marking the first new agent approved for this indication in over a decade. In the , the authorized thalidomide (initially as Thalidomide Celgene, later Thalidomide BMS) on April 16, 2008, for treatment under status, following positive opinions on its efficacy in frontline settings. Approvals were limited primarily to , with no broad endorsements for solid tumors despite exploratory trials in (response rates ~10-20%) and other malignancies showing modest or inconsistent benefits. Efficacy in MM derives from randomized and phase II trials demonstrating improved response rates and (PFS), though overall survival (OS) benefits vary. As monotherapy in relapsed/refractory MM, thalidomide yields overall response rates (ORR) of 25-35%, with median PFS of 6-12 months and OS of 12-18 months in heavily pretreated patients. Combined with dexamethasone (Thal-Dex), ORR rises to 50-72% in newly diagnosed or relapsed cases, with complete response rates of 10-20%; for instance, a phase II study reported 72% ORR and median OS exceeding 38 months in transplant-eligible patients. In induction therapy, Thal-Dex outperformed vincristine-doxorubicin-dexamethasone (VAD), achieving 74% versus 57% ORR and enabling higher collection rates prior to autologous transplant. Maintenance therapy post-autologous transplant with thalidomide extends event-free survival (EFS) by 10-15 months and OS by up to 8 months compared to observation or , as shown in trials like the IFM 99-04 study (3-year OS 87% versus 77%). However, large III trials such as the MRC IX reported PFS benefits (23 versus 17 months) without OS improvement, attributed to toxicity-related discontinuations and subsequent therapies. Neuropathy, occurring in 30-50% of patients, limits long-term use, with diminishing in advanced disease. Post-2010 data confirm thalidomide's role in resource-limited settings but highlight inferiority to analogs like in PFS and tolerability.
Trial/RegimenPatient PopulationORR (%)Median PFS (months)Median OS (months)Key Notes
Thal monotherapy (phase II, 1999-2001)Relapsed/ 25-356-1212-18Initial demonstration of activity; neuropathy in 40%.
Thal-Dex induction (phase III, e.g., IFM)Newly diagnosed, transplant-eligible60-7415-25>38 (not reached in some)Superior to VAD; 12-20% .
Thal (post-ASCT, IFM 99-04)Post-transplant N/A (maintenance)EFS +10-15+8 (3-yr 87%)Improves remission duration; risk increased.
Thal in IX (phase III)Newly diagnosed elderly~50 (with )23 (vs 17)No differencePFS benefit; OS neutral due to .

Risk Management Programs (i.e., iPLEDGE and STEPS)

The System for Thalidomide Education and Prescribing Safety (STEPS) was established by Celgene Corporation in 1998, concurrent with the U.S. Food and Drug Administration's approval of thalidomide (marketed as Thalomid) for the treatment of erythema nodosum leprosum (ENL), a complication of leprosy. This manufacturer-led initiative aimed to mitigate the drug's severe teratogenic risks—known to cause phocomelia and other birth defects—through restricted distribution, mandatory education, and ongoing surveillance. STEPS employed a multifaceted approach: limiting access to registered prescribers and pharmacies, requiring comprehensive counseling for healthcare providers and patients on contraception and fetal risks, and enforcing compliance via periodic surveys and audits. Prescribers were required to document patient understanding via signed agreements, while females of reproductive potential underwent two negative serum pregnancy tests prior to initiation, followed by monthly testing; contraception with two reliable methods was mandatory for women, and men were instructed to use condoms or abstain from intercourse. Dispensing was confined to a single central pharmacy network, with prescriptions limited to 28-day supplies without automatic refills. In 2010, STEPS was integrated into the FDA-mandated THALOMID Risk Evaluation and Mitigation Strategy (REMS), a formalized program under the FDA's post-marketing safety framework to further standardize and oversee thalidomide's distribution for indications including . The REMS maintains core STEPS elements but expands certification requirements: prescribers must enroll via an online portal, complete patient risk acknowledgment forms and annual surveys, and obtain a unique authorization number for each prescription after verifying status. Pharmacies, limited to certified specialty outlets, must confirm prescriber authorization and provide patient counseling with medication guides before release. Patients are enrolled upon their first prescription, committing to ongoing pregnancy prevention; non-compliance triggers treatment interruption. This structure parallels programs like iPLEDGE for , another potent teratogen, though iPLEDGE incorporates a centralized, FDA-overseen database integrating wholesalers and requiring unified monthly attestations, whereas THALOMID REMS emphasizes prescriber-pharmacy coordination without wholesaler mandates. Both prioritize eliminating fetal exposure, but STEPS/REMS reflects thalidomide's narrower therapeutic scope and historical lessons from its 1950s–1960s global crisis. Evaluations indicate high efficacy in averting exposures: since STEPS implementation, no thalidomide-associated fetal malformations have been documented among over 100,000 U.S. recipients, attributable to rigorous (over 90% in surveys) and barriers to casual . Independent assessments confirm that REMS elements enhance patient and provider knowledge of safe-use conditions, with low discontinuation rates due to administrative burden alone (approximately 10% linked to combined toxicities rather than program logistics). Nonetheless, challenges persist, including equitable in underserved populations and the need for vigilant monitoring amid off-label uses or analogs like , which share similar REMS protocols. These programs underscore a causal link between enforced prevention and zero-exposure outcomes, validating stringent controls for drugs with thalidomide's immutable embryotoxic profile.

Research and Derivatives

Ongoing Clinical Trials and Emerging Indications

As of 2025, thalidomide continues to be investigated in phase II and III clinical trials primarily for its immunomodulatory, anti-angiogenic, and properties in hematologic malignancies, gastrointestinal disorders, and radiation-induced complications, often in combination regimens to mitigate toxicity. For instance, a multicenter phase II trial (NCT04453345) evaluates the thalidomide-prednisone-methotrexate (TPM) regimen in (LGLL), reporting a 90% clinical response rate in 20 patients treated since 2013, with a 2025 study confirming efficacy in 52 symptomatic, methotrexate-refractory cases, including 75% overall response and median of 24 months. Similarly, active trials explore thalidomide in transfusion-dependent beta-thalassemia (NCT06146478) to assess impacts on production and in (NCT03318835) combined with R-CHOP chemotherapy for newly diagnosed patients. In gastrointestinal applications, ongoing research targets vascular and inflammatory conditions where thalidomide's inhibition of tumor necrosis factor-alpha and shows promise. A phase III trial demonstrated that 100 mg daily thalidomide reduced recurrent bleeding episodes by 53% compared to in patients with small-intestinal over 12 months, with sustained effects at 18-month follow-up, though limited by in 27% of participants. Complementary studies include phase II evaluations for chronic radiation proctitis (NCT04680195), where low-dose thalidomide addresses refractory bleeding via vascular stabilization, and (NCT02501291), testing 50-100 mg doses for endoscopic remission in steroid-dependent cases. A 2025 review synthesizes evidence for broader digestive system uses, such as in and , attributing efficacy to thalidomide's downregulation of pro-inflammatory cytokines, with response rates up to 60% in open-label cohorts but calling for randomized data to confirm long-term safety. Emerging indications extend to radiation-related toxicities and autoimmune disorders, leveraging thalidomide's established under programs like iPLEDGE to enable controlled access. In radiation-induced , a (NCT05059613) combines thalidomide with to reduce grade 3-4 incidence in head-and-neck cancer patients undergoing radiotherapy, reporting preliminary 40% severity reductions via TNF-alpha suppression. For , phase II data (NCT02201043) indicate improved Bath Ankylosing Spondylitis Disease Activity Index scores with 100 mg thalidomide versus , particularly in TNF-inhibitor failures, though neuropathy risks necessitate monitoring. These pursuits reflect cautious expansion beyond approved uses in and , prioritizing combinations to enhance efficacy while addressing teratogenicity through stringent protocols, with over 20 active trials listed on as of October 2025 focusing on refractory or niche indications.

Thalidomide Analogs: Lenalidomide and Pomalidomide

Lenalidomide and represent second-generation immunomodulatory analogs of thalidomide, engineered with structural modifications to amplify antineoplastic activity and while attempting to diminish certain toxicities, including sedation and observed with the parent compound. These agents primarily target (MM) by binding to (CRBN), a component of the Cullin Ring Ligase 4 (CRL4) E3 complex, which redirects substrate specificity to promote proteasomal degradation of zinc-finger transcription factors such as (IKZF1) and Aiolos (IKZF3). This degradation disrupts myeloma cell proliferation and survival pathways while enhancing T-cell and natural killer cell-mediated against malignant plasma cells. In , cereblon expression correlates with sensitivity to these drugs, underscoring the mechanism's causal role in efficacy. Lenalidomide (CC-5013, marketed as Revlimid) was synthesized in 1999 as the first thalidomide analog with clinical potential, featuring a 4-amino substitution on the ring that enhances potency over thalidomide by approximately 50,000-fold in tumor necrosis factor-alpha inhibition assays. Developed by Celgene Corporation, it received designation from the (FDA) for MM in October 2001 and initial approval on December 27, 2005, for use with dexamethasone in patients with relapsed or refractory MM after at least one prior therapy. Subsequent expansions include approval for newly diagnosed MM in combination with dexamethasone and for myelodysplastic syndromes with deletion 5q in 2006, and in 2013. Clinical trials demonstrated objective response rates of 25-60% in relapsed MM, with median extending 11-18 months when combined with dexamethasone or other agents, outperforming thalidomide in direct comparisons due to superior tolerability and reduced incidence. Pomalidomide (CC-4047, marketed as Pomalyst), a 3-amino analog reported in 2001, emerged as a third-generation with even greater potency, exhibiting 10- to 100-fold higher activity than in preclinical models of inhibition and myeloma cell . Developed through structure-activity optimization by researchers including D'Amato at and licensed to , it binds with higher affinity, inducing more rapid degradation of /3 and additional neo-substrates in resistant cells. The FDA approved on February 8, 2013, in combination with dexamethasone for relapsed or refractory MM patients who had received at least two prior regimens, including and , based on phase II trials showing objective response rates of 29-31% and median duration of response of 8 months in heavily pretreated cohorts. Unlike , retains activity in some lenalidomide-resistant cases, though cross-resistance via CRBN mutations or downregulation limits sequential use. Both analogs retain thalidomide's teratogenic liability, classified as FDA X due to demonstrated embryo-fetal toxicity in animal models, including limb malformations and cardiovascular defects, prompting mandatory Risk Evaluation and Strategies (REMS) programs. The Revlimid REMS and Pomalyst REMS require prescriber , patient registries, monthly testing for females of reproductive potential, and dual contraception for at least 4 weeks before, during, and 4 weeks after , with restricted to certified pharmacies to avert fetal exposure. Preclinical data suggest may exhibit reduced teratogenicity in non-mammalian models like and embryos at therapeutic concentrations, potentially due to species-specific binding differences, but human data are absent and persists. Common adverse effects include (affecting 50-70% of patients), , and , with secondary malignancies observed in 5-10% of long-term users, necessitating . These programs have achieved near-zero rates among enrolled patients, validating their causal in .

Mechanistic Studies and Future Prospects

Thalidomide's primary molecular mechanism involves binding to (CRBN), a substrate receptor in the Cullin Ring 4 (CRL4) E3 ubiquitin ligase complex, which promotes the ubiquitination and proteasomal degradation of specific transcription factors. This "molecular glue" effect recruits CRBN to neo-substrates, altering ; for instance, in teratogenesis, it degrades SALL4 and related factors critical for limb development, explaining observed in exposed embryos as early as 1957–1961. Mechanistic elucidation advanced significantly post-2010, with structural studies confirming CRBN-thalidomide interactions via , revealing how the glutarimide ring binds the thalidomide moiety to induce conformational changes in CRBN's residues, enabling substrate recruitment. These findings, derived from high-resolution cryo-EM and biochemical assays, underscore causal links between CRBN modulation and both toxic and therapeutic outcomes, bypassing earlier hypotheses of nonspecific or metabolic interference. In therapeutic contexts, thalidomide's immunomodulatory effects stem from CRBN-mediated degradation of zinc-finger transcription factors (Ikaros) and IKZF3 (Aiolos), which represses pro-inflammatory production and enhances T-cell and NK-cell activity against myeloma cells. It also inhibits TNF-α by accelerating mRNA degradation, independent of pathway blockade in some models, while exerting anti-angiogenic actions via downregulation of VEGF and basic FGF expression in endothelial cells. Empirical data from studies quantify TNF-α reduction by up to 90% at micromolar concentrations, correlating with clinical responses in leprosum, though pleiotropic effects like PDE4 inhibition contribute variably across cell types. Recent proteomic screens have identified over 100 potential neo-substrates, highlighting thalidomide's polypharmacology, but causal validation remains limited to key targets like IKZF1/3, with off-target degradation risking neuropathy observed in 20–50% of long-term users. Future prospects hinge on leveraging thalidomide's CRBN-binding scaffold for next-generation protein degraders, inspiring designs that selectively ubiquitinate disease-specific proteins beyond . Clinical trials as of 2025 explore combinations in maintenance (e.g., with ), showing extensions of 12–18 months in relapsed patients, though adoption lags in high-income settings due to analogs' superior tolerability. Emerging indications include gastrointestinal disorders via inhibition and autoimmune conditions, with preclinical data indicating efficacy in models at doses below teratogenic thresholds (under 200 mg/day with contraception). However, systemic biases in academic reporting—favoring positive immunomodulatory narratives—necessitate scrutiny; independent meta-analyses confirm modest overall survival benefits (HR 0.74 in myeloma), tempered by neuropathy rates exceeding 30%. Long-term research prioritizes biomarker-driven patient selection, such as CRBN polymorphism screening, to mitigate risks while exploiting degradation pathways for neurodegeneration or , though regulatory hurdles from historical toxicity constrain broad revival.

Societal and Ethical Dimensions

Victim Outcomes and Long-Term Quality of Life

Approximately 10,000 infants were born worldwide with severe congenital malformations due to prenatal thalidomide exposure between 1957 and 1961, with estimates indicating that fewer than 3,000 survivors remain as of the 2020s. among affected cases reached 40-50%, primarily from internal organ failures such as cardiac, gastrointestinal, or respiratory defects, rather than limb anomalies alone. Early projections for survivors suggested limited , with some cases given estimates as low as 19 years, though medical advances and adaptive care have enabled many to reach , often into their 50s or 60s. Survivors predominantly exhibit or related limb reductions, affecting upper extremities more severely than lower ones, alongside malformations of eyes, ears, and internal structures including duplicated thumbs, absent long bones, ocular defects, and auditory nerve damage. As cohorts age, secondary complications emerge at accelerated rates compared to the general population, including progressive musculoskeletal degeneration, , , cardiovascular issues, and gallbladder abnormalities. Surveys of and German survivors indicate that nearly half report multiple concurrent health problems, with 46% experiencing three or more, often necessitating increased reliance on assistive devices, prosthetics, or caregiving despite prior independence. These conditions stem causally from the original teratogenic disruptions to embryonic and nerve development, compounded by mechanical stresses on malformed anatomy over decades. Health-related quality of life (HRQOL) assessments reveal systematically lower scores among thalidomide survivors versus age-matched peers, with longitudinal declines linked to prevalence exceeding 70% in some cohorts and elevated rates of anxiety, , and other disorders. Women with thalidomide embryopathy, in particular, report poorer physical functioning and social participation, though many maintain employment or family roles through adaptations. Middle-aged survivors describe a transition from relative autonomy to escalating , with 3% reporting no issues but the majority facing multimorbidity that outpaces typical aging trajectories. Despite these burdens, resilience is evident in advocacy and creative pursuits, as exemplified by filmmaker , who has documented survivor experiences.

Compensation, Litigation, and Corporate Accountability

In , Chemie , the original manufacturer of thalidomide, faced criminal proceedings initiated in 1963 against nine senior executives for negligent bodily injury and related to the drug's teratogenic effects. The Regional Court terminated the case on September 25, 1970, without convictions, citing insufficient evidence of foreseeability at the time of marketing, though the company had received early reports of by 1960 and withdrawn the drug domestically in November 1961 upon recognizing phocomelia risks. maintained that the birth defects could not have been anticipated through then-standard testing, a position that delayed broader . In the , distributor Distillers Biochemicals (later acquired by ) engaged in protracted litigation starting in the mid-1960s, culminating in a of approximately £28 million to compensate 62 affected families, initially structured at 40% of assessed damages following a 1968 agreement. The funded the Thalidomide Children's Trust (now Thalidomide Trust), which supported around 460 survivors, though some claims were rejected due to strict eligibility criteria linking disabilities directly to documented maternal ingestion between 1958 and 1962. Additional government payments, including £20 million announced in 2010, supplemented private funds as corporate payouts proved inadequate for lifelong care needs. Recent actions, such as a 2014 by eight Britons against and , highlight persistent disputes over excluded cases and escalating medical costs. Australia saw a class-action filed in 2009 on behalf of over 100 victims born between 1958 and 1970, targeting and Australian distributor Adler (part of O'Neil). The Victorian approved an A$89 million in 2014, providing individual payouts averaging around A$650,000 after legal fees, following a 2012 out-of-court award of millions to lead Lynette Rowe, whose case affirmed despite expired statutes of limitations. New Zealand victims were included in the fund. Similar challenges arose in , where lawsuits against manufacturers and regulators persist without a comprehensive national , underscoring difficulties in proving causation decades later amid varying provincial limitation periods. A in 2014 ordered to pay €35 million to 22 victims, rejecting the company's €120,000 annual offer as insufficient and affirming in marketing without adequate warnings. Globally, reported cumulative payments of approximately $680 million by 2010 across various funds, though survivors in multiple jurisdictions criticized allocations as underfunding long-term disabilities, with demands for inflation-adjusted annuities unmet. Corporate accountability remained limited for decades, exemplified by Grünenthal's August 31, 2012, from CEO Harald at a Stolberg unveiling, expressing regret for failing to reach victims "person to person" but denying and attributing silence to "shock" rather than evasion. Victims' groups deemed the insulting absent financial remedies or admission of withheld safety data, such as internal 1950s toxicity concerns. Grünenthal's stance—that pre-1961 testing standards precluded detection of teratogenicity—has been contested by subsequent analyses revealing ignored and lapses, yet no executive faced personal penalties, and the firm continued operations without divestiture of thalidomide profits. This pattern reflects broader resistance to retrospective , prioritizing scientific defensibility over victim redress until public and legal pressures mounted.

Broader Lessons on Innovation vs. Safety Trade-offs

The thalidomide tragedy, which resulted in an estimated 10,000–20,000 children born with severe and other malformations due to maternal ingestion during from 1957 to 1961, exposed critical vulnerabilities in pre-market drug testing, particularly the failure to conduct studies in animals. Prior to this, many jurisdictions, including and pre-1962 , approved drugs based primarily on in non-pregnant adults without mandating proof or comprehensive teratogenicity assessments, enabling rapid market entry but overlooking developmental risks. This causal oversight—stemming from assumptions that adult safety equated to fetal safety—demonstrated how driven by commercial pressures can bypass essential preclinical safeguards, leading to widespread harm when extrapolated to vulnerable populations like pregnant women. In direct response, the enacted the Kefauver-Harris Amendments on October 10, 1962, mandating that manufacturers prove both safety and efficacy through "adequate and well-controlled investigations," with FDA pre-approval required before marketing, alongside requirements for in trials and reporting of adverse events. These reforms, credited with preventing a U.S. thalidomide disaster—thanks to FDA reviewer Frances Kelsey's insistence on additional data—marked a toward rigorous standards, substantially reducing post-approval surprises like teratogenic effects in subsequent drugs. However, empirical analyses indicate trade-offs: average drug approval times lengthened from about 7 months pre-1962 to over 2 years by the 1970s, while compliance costs rose, correlating with a decline in new drug applications and molecular entities approved annually, from a peak of 40–50 in the early 1960s to under 20 by the mid-1970s. Quantitatively, econometric studies attribute a 30–60% drop in innovative output to these heightened barriers, as smaller firms and marginal therapies faced prohibitive R&D expenses without guaranteed returns, though proponents argue the safety gains—evidenced by fewer thalidomide-scale withdrawals—outweigh delays by averting catastrophes that could erode public trust and halt broader pharmaceutical progress. Thalidomide's own post-1990s repurposing for leprosy and multiple myeloma under stringent risk evaluation and mitigation strategies (REMS), which restricted distribution to prevent pregnancy exposure, illustrates a viable equilibrium: controlled access enables high-risk innovations to deliver net benefits, as seen in its FDA approval for erythema nodosum leprosum on July 16, 1998, after prospective registries confirmed efficacy with minimal fetal risks. This duality underscores that while expedited approvals foster rapid deployment of novel therapies, they necessitate parallel investments in targeted safety data—such as species-specific animal models for teratogens—to avoid causal blind spots, without succumbing to regulatory stasis that stifles causal discoveries in unmet needs.

References

  1. [1]
    Thalidomide Has 37-Year History - JAMA Network
    THALIDOMIDE, a glutamic acid derivative (α[N=Phthalimido] glutarimide), was first described in 1953 by the Swiss pharmaceutical firm Ciba.The company.
  2. [2]
    Thalidomide‐induced teratogenesis: History and mechanisms - NIH
    Jun 4, 2015 · Between 1957 and 1962, thalidomide caused severe birth defects in over 10,000 children. Almost any tissue/organ could be affected by thalidomide ...
  3. [3]
    Molecular Mechanisms of the Teratogenic Effects of Thalidomide
    When pregnant women took thalidomide between day 20 and day 36 after fertilization, multiple birth defects occurred [29]. A single tablet (50 mg) of thalidomide ...
  4. [4]
    Thalidomide - StatPearls - NCBI Bookshelf - NIH
    Jun 12, 2023 · Thalidomide is a medication used to manage and treat advanced leprosy and multiple myeloma, and various other solid and hematologic malignancies.
  5. [5]
    Thalidomide as a novel therapeutic agent: new uses for an old product
    This drug is approved in the USA for cutaneous manifestations of lepromatous leprosy and is in Phase III trials for multiple myeloma. Thalidomide and its ...Missing: repurposed | Show results with:repurposed
  6. [6]
    Antitumor Activity of Thalidomide in Refractory Multiple Myeloma
    Nov 18, 1999 · Thalidomide is active against advanced myeloma. It can induce marked and durable responses in some patients with multiple myeloma, including those who relapse ...
  7. [7]
    Thalidomide | C13H10N2O4 | CID 5426 - PubChem - NIH
    Thalidomide | C13H10N2O4 | CID 5426 - structure, chemical names, physical and chemical properties, classification, patents, literature, ...
  8. [8]
    (+/-)-Thalidomide 50-35-1 | Tokyo Chemical Industry (India) Pvt. Ltd.
    Properties (reference). Melting Point, 276 °C. Maximum Absorption Wavelength, 300 nm. Solubility in water, Insoluble. Degree of solubility in water, 60 mg/l 25 ...
  9. [9]
    Thalidomide: Uses, Interactions, Mechanism of Action - DrugBank
    IUPAC Name. 2-(2,6-dioxopiperidin-3-yl)-2,3-dihydro-1H-isoindole-1,3 ... chemical ADMET properties. (23092397). Spectra. Mass Spec (NIST): Not Available ...Identification · Categories · Chemical Identifiers · References
  10. [10]
    Molecular mechanisms of thalidomide and its derivatives - PMC - NIH
    When a ligand such as thalidomide binds to CRBN, it recognizes various 'neosubstrates' depending on the shape of the ligand. CRL4CRBN binds many neosubstrates ...
  11. [11]
    How Thalidomide Works Against Cancer - PMC - NIH
    The small-molecule drugs thalidomide, lenalidomide, and pomalidomide bind to the protein cereblon (CRBN), which activates the enzymatic activity of the CRBN E3 ...
  12. [12]
    Enantiomerization mechanism of thalidomide and the role of water ...
    Nov 5, 2012 · We have identified the two most plausible interconversion pathways for isolated thalidomide: 1) proton transfer from the chiral carbon center ...
  13. [13]
    Enantiomerization Mechanism of Thalidomide and the Role of Water ...
    Oct 12, 2012 · We have identified the two most plausible interconversion pathways for isolated thalidomide: 1) proton transfer from the chiral carbon center ...
  14. [14]
    Thalidomide induces limb defects by preventing angiogenic ...
    May 26, 2009 · Here we demonstrate that loss of immature blood vessels is the primary cause of thalidomide-induced teratogenesis and provide an explanation ...
  15. [15]
    Teratogenic effects of thalidomide: molecular mechanisms - PMC
    Our data suggest that thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting its ubiquitin ligase activity.
  16. [16]
    Free radical-mediated oxidative DNA damage in the mechanism of ...
    The teratogenicity of thalidomide may involve free radical-mediated oxidative damage to embryonic cellular macromolecules.
  17. [17]
    The Molecular Mechanisms of Thalidomide Teratogenicity ... - PubMed
    Thalidomide is a teratogen that affects many organs but primarily induces limb truncations like phocomelia. Rodents are thalidomide resistant.
  18. [18]
    Thalidomide in the treatment of erythema nodosum leprosum (ENL ...
    Oct 9, 2020 · The study enlightens the significant impact of thalidomide in controlling ENL in leprosy patients. It elicits a quick and effective response ...
  19. [19]
    Effective Treatment of Erythema Nodosum Leprosum with ...
    Abstract. The immunomodulatory drug thalidomide is the treatment of choice for erythema nodosum leprosum (ENL), an inflammatory cutaneous and systemic comp.Abstract · Patients, Materials, and Methods · Results
  20. [20]
    Double-blind trial of the efficacy of pentoxifylline vs thalidomide for ...
    As expected, overall, thalidomide proved to be more effective in the treatment of type II leprosy reaction. Nevertheless, continuous treatment with ...
  21. [21]
    Thalidomide in the treatment of leprosy - ScienceDirect.com
    All six studies reported that thalidomide therapy was effective in the amelioration of the signs and symptoms of ENL. Patients' clinical responses were dramatic ...
  22. [22]
    Thalidomide for Erythema Nodosum Leprosum and Other Applications
    Jan 16, 2012 · Thalidomide, an inhibitor of tumor necrosis factor a, is the best known agent for short-term treatment of ENL skin manifestations, as well as ...
  23. [23]
    Effects of Thalidomide on Intracellular Mycobacterium leprae in ...
    A distinct sequence of events explaining the mechanism of action for thalidomide's successful treatment of ENL has yet to be established. When administered ...
  24. [24]
    No Role for Thalidomide in Leprosy - PAHO/WHO
    Jan 14, 2000 · During the mid-1960s, the drug thalidomide was reintroduced as treatment for a complication of leprosy called Erythema nodosum leprosum (ENL).
  25. [25]
    The Rise, Fall and Subsequent Triumph of Thalidomide
    D'Amato and colleagues postulated that thalidomide-induced birth defects were due to inhibited blood vessel growth in the developing fetal limb bud and perhaps ...
  26. [26]
    Guideline for clinical use of thalidomide for management of ... - J-Stage
    Thalidomide was first approved for ENL by US FDA in 1998 (Table 1), where it may be prescribed only by licensed prescribers who are registered in the System ...<|separator|>
  27. [27]
    Thalidomide in the treatment of erythema nodosum leprosum (ENL ...
    Oct 9, 2020 · The study showed that thalidomide treatment for patients with ENL is possible in outpatientclinics. We also successfully prevented pregnancies to a larger ...
  28. [28]
    Clofazimine in Erythema Nodosum leprosum: where do we stand?
    Two trials showed significant long-term benefits with clofazimine treatment compared to thalidomide (fewer recurrences) or prednisolone (more treatment success) ...
  29. [29]
    Thalidomide and its analogues in the treatment of Multiple Myeloma
    Sep 11, 2012 · The VTD is often the induction regimen of choice outside of clinical trials, with overall response rates of 93%. Currently three-novel drug and ...
  30. [30]
    Thalidomide - NCI - National Cancer Institute
    Oct 5, 2006 · Thalidomide is approved to be used with dexamethasone to treat: Multiple myeloma in patients who have just been diagnosed with the disease.
  31. [31]
    [PDF] THALOMID® (thalidomide) capsules for oral use - accessdata.fda.gov
    THALOMID in combination with dexamethasone is indicated for the treatment of patients with newly diagnosed multiple myeloma (MM). 1.2 Erythema Nodosum Leprosum.
  32. [32]
    Maintenance therapy with thalidomide improves survival in patients ...
    Nov 15, 2006 · Our trial shows that thalidomide improves the quality of response after randomization. We have previously reported that the attainment of a very ...
  33. [33]
    The role of maintenance thalidomide therapy in multiple myeloma
    In this study, thalidomide maintenance therapy significantly improved PFS in newly diagnosed MM, and with the use of effective relapse treatment, this benefit ...
  34. [34]
    Comparative effectiveness and safety of thalidomide and ...
    Our results agree with the findings of recently published trials suggesting that thalidomide and lenalidomide are equivalent with respect to survival outcomes.
  35. [35]
    Analysis of the Efficacy of Thalidomide Plus Dexamethasone-Based ...
    Patients with disease that responded to the thalidomide-based regimen had better progression-free survival compared to those who did not (median, 8.8 vs. 2.5 ...
  36. [36]
    Thalidomide for treatment of graft-versus-host disease - PubMed - NIH
    Treatment of chronic GVHD in this model has shown thalidomide to be better tolerated and more successful than cyclosporine (CSA) or prednisone plus azathioprine ...
  37. [37]
    Thalidomide in the treatment of the mucocutaneous ... - PubMed
    Thalidomide is effective for treating the oral and genital ulcers and follicular lesions of the Behçet syndrome. A dosage of 100 mg/d is as effective as a ...
  38. [38]
    Thalidomide induces limb defects by preventing angiogenic ... - PNAS
    Thalidomide is a potent teratogen that induces a range of birth defects, most commonly of the developing limbs. The mechanisms underpinning the teratogenic ...
  39. [39]
    The Tragedy of Birth Defects and the Effective Treatment of Disease
    The sensitive period during pregnancy for thalidomide effects in humans is approximately days 20–34 after fertilization (Miller and Strömland, 1999; Miller et ...
  40. [40]
    Thalidomide Embryopathy: An Enigmatic Challenge - 2013
    Oct 31, 2013 · Some reports indicate thalidomide exposure in utero could result in a 50% risk of a birth defect [7, 14, 15]. Given the potency of the drug how ...<|separator|>
  41. [41]
    [PDF] Thalomid REMS: Pregnancy Exposure Registry Protocol - FDA
    THALOMID Pregnancy Prevention REMS Program ... Under this program, the System for Thalidomide Education and Prescribing Safety (THALOMID.
  42. [42]
    Thalidomide use in the US : experience with pregnancy testing in ...
    ... FDA for the treatment of erythema nodosum leprosum. To prevent fetal exposure to thalidomide, a restricted distribution risk management programme, the ...
  43. [43]
    [PDF] Prescriber Guide to THALOMID REMS Program
    A known teratogen, THALOMID® (thalidomide) is approved for marketing only under a restricted distribution program approved by the Food and. Drug Administration.
  44. [44]
    [PDF] Thalomid REMS: Patient Guide - FDA
    To avoid serious risks to unborn babies, THALOMID is only available under a restricted distribution program called the “THALOMID Risk Evaluation and Mitigation ...
  45. [45]
    Thalidomide-induced Neuropathy - PubMed
    Thalidomide induces a dose-dependent sensorimotor length-dependent axonal neuropathy; it should be judiciously used with close neurologic monitoring.Missing: side | Show results with:side
  46. [46]
    Incidence and Risk Factors for Thalidomide Neuropathy
    The incidence rate of thalidomide-induced neuropathy has been variously estimated, from less than 1% to more than 70%.
  47. [47]
    Peripheral neuropathy and new treatments for multiple myeloma
    Feb 1, 2010 · The neurological complications usually occur after prolonged exposure: 70% of the patients treated for 12 months will develop PN. The clinical ...Pathogenesis of peripheral... · Clinical aspects · Incidence, severity and risk...
  48. [48]
    Thalidomide-induced neuropathy | Neurology
    Objective: To describe the clinical, electrophysiologic, and pathologic features of thalidomide-induced peripheral neuropathy. Methods: Clinical and ...
  49. [49]
    Thalidomide-Induced Peropheral Neuropathy in Newly Diagnosed ...
    Nov 16, 2004 · Symptoms included paresthesias, tremor and dizziness; serial electromyographic studies revealed a symmetrical, mainly sensory peripheral ...
  50. [50]
    The adverse effects of thalidomide in relapsed and refractory ...
    Constipation (100% incidence) and sedation (87%) were the most common adverse effects. Neuropathy had low incidence and was late in onset (>12 months).
  51. [51]
    Thalidomide - LiverTox - NCBI Bookshelf - NIH
    Aug 30, 2022 · Rare but potentially severe adverse events include severe cutaneous reactions, seizures, tumor lysis syndrome and hypersensitivity reactions.Missing: excluding | Show results with:excluding<|control11|><|separator|>
  52. [52]
    Thalidomide Interactions Checker - Drugs.com
    There are 827 drugs known to interact with thalidomide, along with 5 disease interactions, and 1 alcohol/food interaction. Of the total drug interactions, 207 ...
  53. [53]
    Thalidomide: Side Effects, Uses, Dosage, Interactions, Warnings
    Sep 6, 2017 · What Other Drugs Interact with Thalidomide? · amobarbital · butabarbital · butalbital · chlorpromazine · cholera vaccine · dichlorphenamide · ethanol ...What Is · Side Effects · Drug Interactions
  54. [54]
    Drug Interactions of Thalidomide with Midazolam and Cyclosporine A
    The present results suggest that total midazolam metabolism or cyclosporine A clearance may be increased by thalidomide in a dose-dependent manner.
  55. [55]
    Thalidomide – Detailed History and Timeline - RMS Consultancy
    27th November, Chemie Grünenthal reluctantly withdrew Thalidomide from the West German market. 1961 2nd December, Distillers Company (Biochemicals) Ltd ...
  56. [56]
    The History of Thalidomide - PubMed
    The first paper describing the pharmacological actions of thalidomide was published in 1956. The drug, then designated as K17, was thought to have sedative ...Missing: pre- | Show results with:pre-
  57. [57]
    A History of Regulatory Animal Testing: What Can We Learn?
    Aug 19, 2022 · Even in the case of thalidomide, animal testing in 10 strains of rats, 11 breeds of rabbits, two breeds of dogs, three strains of hamsters, ...Missing: pre- 1950s<|separator|>
  58. [58]
    Thalidomide - Science Museum
    Dec 11, 2019 · It took five years for the connection between thalidomide taken by pregnant women and the impact on their children to be made.Introduction · Thalidomide in the marketplace · The thalidomide scandal
  59. [59]
    Sixty years on: the history of the thalidomide tragedy
    Dec 2, 2021 · After being on the market for five years as a treatment for morning sickness in pregnant women, it had finally been established that the ...
  60. [60]
    FBR - About Animal Research
    Feb 4, 2025 · The thalidomide tragedy could have been avoided if researchers had studied the drug's effects with pregnant animals before releasing it to the ...Missing: pre- | Show results with:pre-
  61. [61]
    File:Thalidomide synthesis 01.svg - Wikimedia Commons
    ... Synthesis of thalidomide - first Grünenthal version (GB 768 821 (Chemie Grünenthal; 20.2.1957). Date, 19 March 2013. Source, Own work. Author, Steffen ...Missing: initial | Show results with:initial
  62. [62]
    History | About us | Grünenthal - Grunenthal
    In 1957, the sleep and sedative drug Thalidomide (marketed as Contergan in Germany) is introduced to the market. After its teratogenic effects were discovered, ...Missing: synthesis date<|separator|>
  63. [63]
    The Tragic History of Thalidomide and Phocomelia - PBS
    Apr 5, 2016 · On Oct. 1, 1957, the company launched thalidomide and began aggressively advertising and marketing it as completely safe. Thalidomide was used ...
  64. [64]
    The Canadian Tragedy | Thalidomide
    It was then marketed in several other countries, including the United Kingdom (1958), Japan (1958) and Norway (1959). The drug would have entered the Canadian ...Missing: global timeline
  65. [65]
    The history of the Thalidomide tragedy
    However, Grünenthal itself did not promote taking the medication for morning sickness. In November 1961, it became known that the drug caused severe deformities ...
  66. [66]
    Thalidomide: the tragedy of birth defects and the effective treatment ...
    It became apparent in the 1960s that thalidomide treatment resulted in severe birth defects in thousands of children.
  67. [67]
    Thalidomide: history, withdrawal, renaissance, and safety concerns
    Thalidomide is used around the world today to effectively treat inflammatory and cancer conditions. Yet, 60 years ago, it was withdrawn from the market.Missing: investigational besides
  68. [68]
    Causes of birth defects: lessons from history - PubMed
    The teratogenic effects of thalidomide were first noted in 1961 by William McBride, a Sydney obstetrician, and independently confirmed by Widukind Lenz, a ...
  69. [69]
    William McBride: alerted the world to the dangers of thalidomide in ...
    Aug 6, 2018 · In May 1961, McBride delivered a baby with malformed arms and severe internal damage. The baby died shortly after birth. A few weeks later he ...
  70. [70]
    Thalidomide: an unanticipated adverse effect.
    On the 16 December 1961 The Lancet published a letter by an Australian obstetrician, William McBride (1961). He reported that one out of every five women ...
  71. [71]
    Learning about the Safety of Drugs — A Half-Century of Evolution
    Dec 8, 2011 · McBride, WG. Thalidomide and congenital abnormalities. Lancet 1961;2:1358-1358. Go to Citation. Crossref · Web of Science · Google Scholar. 4 ...
  72. [72]
    of foetal deformity (McBride 1961; Lenz 1962
    SOONafter the first reports that thalidomide might be the cause of foetal deformity (McBride 1961; Lenz 1962; Pfeiffer and. Kosenow 1962) Speirs (1962) ...<|separator|>
  73. [73]
    Thalidomide: a byword for tragedy - O&G Magazine
    In humans, the critical period of exposure to thalidomide is 34–50 days after the last menstrual period (LMP). In biological and basic scientific texts ...
  74. [74]
    Congenital Malformations | Thalidomide
    Recognition of birth defects caused by thalidomide · Absence of the auricles with deafness. · Defects of the muscles of the eye and of the face. · Absence or ...
  75. [75]
    Thalidomide: the untold American story in “Wonder Drug”
    Jul 17, 2023 · The U.S. never approved the drug, which killed perhaps 10,000 children in Europe and caused thousands more to be born with serious birth defects ...
  76. [76]
    Brazil's new generation of Thalidomide babies - BBC News
    Jul 24, 2013 · About 10,000 Thalidomide babies were born worldwide until the drug was withdrawn in the early 1960s. In most countries the Thalidomide children ...
  77. [77]
    Thalidomide - MotherToBaby | Fact Sheets - NCBI Bookshelf - NIH
    The birth defects usually seen with thalidomide exposure during pregnancy are very short or missing arms and legs, missing parts of the ears, and deafness.Missing: timeline | Show results with:timeline
  78. [78]
    Studies of Thalidomide's Effects on Rodent Embryos from 1962-2008
    Mar 7, 2014 · Thalidomide is a sedative drug introduced to European markets on 1 October 1957 after claims of extensive testing on rodent embryos to ensure ...
  79. [79]
    Myths and facts about animal testing
    When thalidomide was tested on pregnant animals, the same birth defects were caused in the animals' offspring as those seen in humans. The failure to test ...
  80. [80]
  81. [81]
    Why did thalidomide's makers ignore warnings about their drug?
    Dec 7, 2015 · In the 1950s, German company Chemie Grünenthal developed a “wonder drug” sleeping pill that it marketed around the world as safe for everyone, including ...
  82. [82]
    How the thalidomide scandal led to safer drugs - MedicalNewsToday
    Dec 15, 2020 · Between 1957 and 1962, more than 10,000 babies were born with physical abnormalities caused by the drug thalidomide.
  83. [83]
    Frances Oldham Kelsey: Medical reviewer famous for averting ... - FDA
    Feb 1, 2018 · Dr. Kelsey, despite constant pressure from the company, refused to approve the application because of its inadequate evidence. Frances Oldham ...
  84. [84]
    How a courageous physician-scientist saved the U.S. from a birth ...
    Mar 10, 2020 · As an FDA reviewer in 1961, Frances Kelsey, MD, PhD, resisted pressure to approve the drug thalidomide, later shown to cause severe birth ...<|separator|>
  85. [85]
    The Devil in the Details, Chapter One: The Doctor Who Said No to ...
    Sep 11, 2024 · Starting with her rejection of an FDA application for thalidomide in 1960, physician and pharmacist Frances Oldham Kelsey took a stand against the now infamous ...
  86. [86]
    Was Thalidomide Safe? Frances Oldham Kelsey Was Not Convinced
    Sep 19, 2024 · The clinical human studies were incomplete. They hadn't looked at long-term safety. There were inconsistencies. There were confusing parts.
  87. [87]
    The Story of Thalidomide in the U.S., Told Through Documents
    Mar 24, 2020 · A trove of archival records shows how in the early 1960s the Food and Drug Administration investigated the use of a drug that caused severe ...
  88. [88]
    Thalidomide scandal: How Australia's response has 'lagged behind'
    Apr 2, 2019 · Although Japan and Canada kept it on shelves longer than others, Australia made a comparatively weak and late effort to get it out of surgeries ...<|separator|>
  89. [89]
    U of T expert on why we need answers to the thalidomide tragedy to ...
    Oct 25, 2018 · Why did Canada wait three months longer than Germany and the U.K. to decide that thalidomide was too unsafe to be sold? Some may wonder why ...
  90. [90]
    What's happened to Thalidomide babies? - BBC News
    Nov 3, 2011 · The sedative Thalidomide was withdrawn after thousands of mothers gave birth to disabled babies. That ageing Thalidomide generation now faces rising care bills.Missing: warnings | Show results with:warnings<|control11|><|separator|>
  91. [91]
    US Regulatory Response to Thalidomide (1950-2000)
    Apr 1, 2014 · Prior to discovering thalidomide's teratogenic effects in the early 1960s, the US Food and Drug Administration (FDA) did not place regulations ...<|separator|>
  92. [92]
    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, ...
  93. [93]
    Milestones in US Food and Drug Law - FDA
    Jan 30, 2023 · Kefauver-Harris Drug Amendments passed to ensure drug efficacy and greater drug safety. For the first time, drug manufacturers are required ...
  94. [94]
    [PDF] A Historical Perspective on the Pace of Drug Approval, 1950s-2009.
    On the background of the thalidomide crisis in the early 1960s, however, the drug approval process was reframed. The 1962 Kefauver-Harris Amendments ensured a ...
  95. [95]
    Thalidomide in Canada | The Canadian Encyclopedia
    Feb 12, 2025 · The drug thalidomide was developed in Germany in the 1950s and was first available in Canada in 1959. It was originally prescribed as a ...<|separator|>
  96. [96]
    Reform, Regulation, and Pharmaceuticals — The Kefauver–Harris ...
    in the form of “adequate and well-controlled investigations”— before ...
  97. [97]
    Countering the Problem of Falsified and Substandard Drugs - NCBI
    In the 1940s and 1950s, before the thalidomide crisis of the early 1960s, federal drug regulators in the United States began to regulate efficacy (Carpenter, ...
  98. [98]
    A Brief History of the Center for Drug Evaluation and Research - FDA
    Jan 31, 2018 · On October 10 President Kennedy signed the Drug Amendments of 1962, also known as the Kefauver-Harris Amendments. These Amendments required ...
  99. [99]
    Two Thalidomide Disasters | Cato Institute
    One could make an argument that a federal government agency should check drugs for safety before marketing, realizing that some safety problems are rare and won ...Missing: deficiencies | Show results with:deficiencies
  100. [100]
    The 50th Anniversary of the Kefauver‐Harris Amendments: Efficacy ...
    Oct 3, 2012 · The practice of informed consent and the creation of institutional review boards, for example, are directly traceable to them, and ongoing ...
  101. [101]
    [PDF] Promoting Safe and Effective Drugs for 100 Years | FDA
    "Also critically, the 1962 amendments required that the FDA specifically approve the marketing application before the drug could be marketed, another major ...
  102. [102]
    Thalidomide Tragedy Prompts Passage of the Kefauver-Harris ...
    Grünenthal began offering thalidomide as an over-the-counter drug (but one that it encouraged physicians to recommend) under a variety of trade names, such as ...
  103. [103]
    An historical overview over Pharmacovigilance - PMC - NIH
    In Europe (1965), the disaster of thalidomide stimulated the development of a European legislation with the EC Directive 65/65 [15]. In 1966, a pilot study of ...
  104. [104]
    Thalidomide, Drug Safety Regulation, and the British ... - SpringerLink
    It is well known that, in the wake of the thalidomide tragedy, drug safety regulation became more stringent, and that this would have a profound effect on the ...
  105. [105]
    The penumbra of thalidomide, the litigation culture and the licensing ...
    Aug 19, 2012 · An unintended consequence of these changes was that drugs have become ruinously expensive. It can now take more than 10 years and cost more than ...<|separator|>
  106. [106]
    A 1962 law slowed drug development. Changing it could slash drug ...
    Oct 1, 2018 · Reversing the 1962 amendments could slash new drug costs virtually overnight, stimulate innovation, and add years to our lives. The amendments ...
  107. [107]
    An Evaluation of Consumer Protection Legislation: The 1962 Drug ...
    The compliance costs are shown to have engendered a marked reduction in drug innovation. Consumer surplus analysis is then adapted and supplemented with "expert ...
  108. [108]
    [PDF] the impact on Patients of Delayed Drug approvals - Manhattan Institute
    This paper reexamines the costs of the drug- development process in this light and illustrates empirically how incorporating what economists refer to as ...
  109. [109]
    Lost medicines: a longer view of the pharmaceutical industry with ...
    The Kefauver–Harris Amendment was a hindrance to the pharmaceutical industry because it decreased the sales of many existing medicines. At the same time, this ...<|separator|>
  110. [110]
    [PDF] REGULATION OF PHARMACEUTICAL INNOVATION
    The net effect of the 1962 amendments, then, may well be more risk of adverse drug reactions and less safety than if the law had never been changed.
  111. [111]
    Fifty years after thalidomide; what role for drug regulators? - PMC - NIH
    Fifty years after thalidomide, there is still an important role for drug regulators, but the time has come to bring patients fully into the decision process, ...
  112. [112]
    The revival of thalidomide: From tragedy to therapy
    Jun 5, 2011 · Despite its tragic history, thalidomide has found its place in the treatment of leprosy, HIV/AIDS, and cancer.Missing: repurposed | Show results with:repurposed
  113. [113]
    The drug of good and evil
    Nov 1, 2014 · In 1964, an Israeli doctor at the Hadassah University Hospital, Jacob Sheskin, prescribed thalidomide for a leprosy patient's insomnia.Troubled History · Discovering Other Uses · Finding More Puzzle Pieces
  114. [114]
    THALIDOMIDE IN THE TREATMENT OF LEPRA REACTIONS
    THALIDOMIDE IN THE TREATMENT OF LEPRA REACTIONS. ... Author. J SHESKIN. PMID: 14296027; DOI: 10.1002/cpt196563303. MeSH terms. Arsenicals*; Drug Therapy*; Humans ...Missing: 1964 | Show results with:1964
  115. [115]
    A review of thalidomide's history and current dermatological ...
    In 1998, thalidomide became FDA approved for the acute treatment and suppression of the cutaneous manifestations of erythema nodosum leprosum (ENL).
  116. [116]
    Thalomid/Thalidomide for Multiple Myeloma | The IMF
    Thalomid (Thalidomide) is less commonly prescribed for multiple myeloma, but may be a good alternative for patients with low blood cell counts.Missing: leprosy | Show results with:leprosy<|control11|><|separator|>
  117. [117]
    Thalidomide sNDA Granted Approval for Newly Diagnosed Multiple ...
    Jun 1, 2006 · The US Food and Drug Administration (FDA) has granted accelerated approval to its Supplemental New Drug Application (sNDA) for thalidomide (Thalomid) in ...
  118. [118]
    EU/3/01/067 - orphan designation for treatment of multiple myeloma
    Jul 29, 2008 · Update: Thalidomide (Thalidomide Celgene) has been authorised in the EU since 16 April 2008 for the treatment of myeloma.
  119. [119]
    Phase II trial of thalidomide in renal-cell carcinoma
    Thalidomide has been reported to yield anti-tumor activity in cancer. We performed a phase II trial of this drug in patients with metastatic renal cell ...
  120. [120]
    A systematic review of phase II trials of thalidomide/dexamethasone ...
    Thalidomide monotherapy in relapsed/refractory multiple myeloma (MM) has a response rate of 30%. The combination of thalidomide with dexamethasone (Thal/Dex) ...
  121. [121]
    Survival Outcomes of Patients Receiving Thalidomide ...
    Nov 16, 2006 · The overall response rate was 72%, including 12% with CR. Median overall survival (OS) of those who received autologous stem cell transplant ( ...
  122. [122]
    Superiority of thalidomide and dexamethasone over vincristine ...
    The aim of the present study was to compare thalidomide-dexamethasone (Thal-Dex) and vincristine-doxorubicin-dexamethasone (VAD) as primary therapy in prep.Missing: key | Show results with:key
  123. [123]
    S.T.E.P.S.: a comprehensive program for controlling and ... - PubMed
    program uses a three-pronged approach: (1) controlling access to the drug; (2) educating prescribers, pharmacists, and patients; and (3) monitoring compliance.Missing: details | Show results with:details
  124. [124]
    Evaluation of the Thalidomide Fetal Exposure Prevention Program ...
    Background. Thalidomide is a drug known to cause serious birth defects if taken during pregnancy. In 1998, FDA approved thalidomide under restricted ...
  125. [125]
    Pregnancy prevention programs for medications used in dermatology
    Apr 17, 2020 · The FDA has developed the REMS program for thalidomide, isotretinoin, and acitretin, all with a high risk of embryopathy [Table 1]. The goal of ...
  126. [126]
    [PDF] Thalomid (thalidomide) capsules - accessdata.fda.gov
    Mar 24, 2023 · RISK EVALUATION AND MITIGATION STRATEGY (REMS) REQUIREMENTS. The REMS for Thalomid (thalidomide) was originally approved on August 3, 2010, and.
  127. [127]
    About the THALOMID REMS program
    THALOMID (thalidomide) is only available under a restricted distribution program called “THALOMID Risk Evaluation and Mitigation Strategy (REMS).”
  128. [128]
    iPLEDGE REMS - Home
    The iPLEDGE REMS is a safety program to manage the risk of isotretinoin's teratogenicity and to minimize fetal exposure.
  129. [129]
    Thalidomide—Cost and Development: In Response
    Celgene has been marketing Thalomid using S.T.E.P.S. since 1998, and among the more than 100,000 Americans who have received the drug, no malformations have ...
  130. [130]
    Effectiveness of Risk Evaluation and Mitigation Strategies (REMS ...
    Jan 10, 2017 · The lenalidomide and thalidomide REMS programs enhance patient understanding of safe-use messages, resulting in high levels of compliance with the birth ...
  131. [131]
    Trends in Use and Evidence of Adherence to Risk Evaluation and ...
    May 16, 2024 · Thalidomide, lenalidomide, and pomalidomide have had safety programs intended to prevent embryo-fetal exposure since their approval.
  132. [132]
    The Return of Thalidomide | Drug Safety
    Nov 25, 2012 · This mandatory, but untested, programme promises to be effective at preventing fetal exposures to thalidomide, provided that patients, ...Missing: no | Show results with:no
  133. [133]
    TPM Regimen (Thalidomide, Prednisone and Methotrexate) in LGLL
    The investigators used TPM regimen (thalidomide + prednison + methotrexate ) to treat LGLL since 2013, and 18/20 patients (90%) obtained clinical response, ...
  134. [134]
    Thalidomide-based regimen shows promising efficacy in large ...
    Mar 12, 2025 · This multicenter study evaluated the efficacy and safety of a thalidomide, prednisone, and methotrexate (TPM) regimen in 52 symptomatic, methotrexate- and ...
  135. [135]
    NCT06146478 | Deciphering Effects of Thalidomide on Red Blood ...
    The goal of this Non-Randomized Clinical Trial is to determine the effects of thalidomide on red blood cells in transfusion dependent beta thalassemia patients.
  136. [136]
    Thalidomide Combined With R-CHOP in Newly Diagnosed ...
    If a study with a recruitment status of recruiting; not yet recruiting; or active, not recruiting has not been confirmed within the past 2 years, the ...
  137. [137]
    Thalidomide for Recurrent Bleeding Due to Small-Intestinal ...
    Nov 1, 2023 · In this placebo-controlled trial, treatment with thalidomide resulted in a reduction in bleeding in patients with recurrent bleeding due to SIA.Missing: emerging | Show results with:emerging
  138. [138]
  139. [139]
    Thalidomide in Treating Crohn's Disease | ClinicalTrials.gov
    Therefore the aim of this study is to investigate the efficacy on clinical and endoscopic response and the adverse effects of using low-dose thalidomide in ...
  140. [140]
    A review of thalidomide and digestive system related diseases
    May 28, 2025 · Thalidomide is currently used as an antineoplastic therapeutic agent in multiple myeloma (43–46), melanoma (47), glioblastoma multiforme (48), ...Missing: investigational besides
  141. [141]
    Thalidomide With or Without Probiotics in Radioactive Oral Mucositis
    An entry on ClinicalTrials.gov that contains a summary of a clinical study's protocol information, including the recruitment status; eligibility criteria; ...
  142. [142]
    NCT02201043 | Phase II Clinical Study of Thalidomide in the ...
    A multicenter, randomized, double-blind, placebo-controlled, Phase II clinical study of thalidomide in the treatment of ankylosing spondylitis.
  143. [143]
    Cereblon is a direct protein target for immunomodulatory and ...
    May 3, 2012 · Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide | Leukemia.
  144. [144]
    The novel mechanism of lenalidomide activity - PMC - NIH
    Lenalidomide acts by a novel drug mechanism—modulation of the substrate specificity of the CRL4CRBN E3 ubiquitin ligase. In multiple myeloma, lenalidomide ...
  145. [145]
    Cereblon expression is required for the antimyeloma activity of ...
    We investigated the role of cereblon (CRBN), a primary teratogenic target of thalidomide, in the antimyeloma activity of IMiDs.
  146. [146]
    The story of the development of generic lenalidomide
    Syntheis and clinical development thalidomide. Lenalidomide, or CC-5013, was first synthesized in 1999 as a structural analogue of thalidomide [1], [24], [25].
  147. [147]
    Development of Analogs of Thalidomide - Encyclopedia.pub
    Lenalidomide is the first analog of thalidomide which is marketed. ... In October 2001 lenalidomide was granted orphan status for the treatment of MM. In mid-2002 ...
  148. [148]
    The tale of lenalidomide clinical superiority over thalidomide and ...
    Lenalidomide (Revlimid®) was the first thalidomide-like drug obtaining a marketing authorization (Figure 1). It was developed by Celgene® Co and approved by the ...
  149. [149]
    Lenalidomide use in multiple myeloma (Review)
    Nov 28, 2023 · Lenalidomide was the first thalidomide analog to be marketed, and it was more potent than its parent drug despite only two differences at ...<|separator|>
  150. [150]
    From thalidomide to pomalyst - The D'Amato Lab
    Robert D'Amato, MD, PhD, developed Pomalyst®, recently approved to treat multiple myeloma and the most potent analog to date of thalidomide.
  151. [151]
    [PDF] 204026Orig1s000 - accessdata.fda.gov
    Feb 7, 2013 · Pomalidomide is an immune-modulator and analogue of thalidomide, developed to treat patients with relapsed/refractory multiple myeloma.
  152. [152]
    [PDF] 204026Orig1s000 - accessdata.fda.gov
    Dec 13, 2012 · Pomalidomide is an immune-modulator and analogue of thalidomide, developed to treat patients with relapsed/refractory multiple myeloma.
  153. [153]
    Novel immunomodulatory drugs and neo-substrates
    Jan 9, 2020 · Thalidomide, lenalidomide and pomalidomide are immunomodulatory drugs (IMiDs) effective in the treatment of multiple myeloma, myelodysplastic syndrome (MDS)
  154. [154]
    [PDF] 204026Orig1s000 - accessdata.fda.gov
    Feb 7, 2013 · Pomalidomide is a thalidomide analogue for the treatment of patients with multiple myeloma that has relapsed after prior therapy. It is the ...
  155. [155]
    Pomalidomide is nonteratogenic in chicken and zebrafish embryos ...
    We found that Pomalidomide, displays a high degree of cell specificity, and has no detectable teratogenic, antiangiogenic or neurotoxic effects at potent anti- ...<|separator|>
  156. [156]
    [PDF] POMALYST (pomalidomide) capsules - accessdata.fda.gov
    POMALYST, in combination with dexamethasone, is indicated for patients with multiple myeloma who have received at least two prior therapies including ...
  157. [157]
    After 60 years, scientists uncover how thalidomide produced birth ...
    Aug 1, 2018 · Building on years of previous research, the researchers found that thalidomide acts by promoting the degradation of an unexpectedly wide range ...Missing: pharmacology | Show results with:pharmacology
  158. [158]
    Structural studies reveal thalidomide's mechanism of action and ...
    Thalidomide initiates its teratogenic effects by binding to CRBN and inhibiting the associated ubiquitin ligase activity. Building off the research of Ito et al ...
  159. [159]
    Immunological Effects of Thalidomide and Its Chemical ... - PubMed
    Its effectiveness in the clinic has been ascribed to wide-ranging properties, including anti-TNF-alpha, T-cell costimulatory and antiangiogenic activity.
  160. [160]
    Recent advances in the molecular mechanism of thalidomide ...
    Thalidomide can inhibit basic fibroblast growth factor (bFGF)-mediated angiogenesis, and blood vessels are essential for embryonic development, leading to the ...Missing: pharmacology | Show results with:pharmacology
  161. [161]
    Thalidomide, the trailblazing molecular glue degrader
    Jun 13, 2022 · Thalidomide's action as a molecular glue brings together two proteins that would normally not interact, recruiting cereblon (CRBN), an E3 ligase, to bind to ...Missing: studies | Show results with:studies
  162. [162]
    Thalidomide in cancer medicine - Annals of Oncology
    Recent data suggest that this drug can block the activation of nuclear factor (NF)-kB through a mechanism involving the inhibition of IkB kinase activity [8].
  163. [163]
    About Thalidomide
    Thalidomide is a drug that was marketed as a sedative and treatment for morning sickness in pregnant women in the late 50s and early 60s.
  164. [164]
    Thalidomide survivors still struggle 60 years on – DW – 11/27/2021
    Nov 27, 2021 · Four to five out of every 10 children who were exposed to the drug died shortly after birth. "As harmless as a sugar cookie" is how thalidomide ...Missing: outcomes | Show results with:outcomes
  165. [165]
    Living and ageing with birth defects caused by the drug thalidomide
    Jul 30, 2013 · Given a life expectancy of 19 years, Mandy was one of more than 10,000 babies worldwide born with a disability caused by the drug thalidomide.Missing: outcomes | Show results with:outcomes
  166. [166]
    The health and quality of life of Thalidomide survivors as they age
    Jan 16, 2019 · A high proportion of Thalidomide survivors reported multiple health problems (see Fig 2). Just 3% had no health problem but almost half (46%) ...Missing: outcomes | Show results with:outcomes
  167. [167]
    The changing health of Thalidomide survivors as they age
    Thalidomide survivors were born with a range of impairments. Most commonly they have missing or short and/or deformed limbs (Phocomelia).
  168. [168]
    Health‐related quality of life after 50 years in individuals with ...
    Jun 4, 2022 · A longitudinal decreasing HRQOL in individuals with thalidomide embryopathy is known, as well as high prevalence of mental disorders and chronic pain syndromes.Missing: survival | Show results with:survival<|separator|>
  169. [169]
    Life situation of women impaired by Thalidomide embryopathy in ...
    Apr 3, 2019 · Both studies show that women impaired by Thalidomide embryopathy face a poorer health status than women their age in the general population.
  170. [170]
    The health and quality of life of Thalidomide survivors as they age
    Jan 16, 2019 · Having lived relatively independent lives, many Thalidomide survivors are now having to adjust to growing disability.Missing: studies | Show results with:studies
  171. [171]
    [PDF] The Health and Wellbeing of Thalidomide Survivors in Middle Age
    The findings from a UK study commissioned by the. Thalidomide Society suggested that the health of Thalidomide survivors was deteriorating more quickly than ...
  172. [172]
    End of criminal proceedings against Grünenthal: 50 years later
    50 years ago today, the Aachen Regional Court terminated the Thalidomide proceedings against nine Grünenthal senior employees.Missing: compensation | Show results with:compensation
  173. [173]
    Statement: Government money for thalidomide survivors
    The drug's UK manufacturer, Distillers Biochemicals, paid around £28 million compensation in the 1970s following a legal battle by the families of those ...
  174. [174]
    Thalidomide: Britons launch legal case - BBC News
    Jun 5, 2014 · The claimants are suing Grunenthal, which manufactured the drug, and Diageo Scotland which now owns Distillers. 'Held to account'. Fraser ...
  175. [175]
    Court formally approves $89m payout for Thalidomide victims
    Feb 6, 2014 · The Victorian Supreme Court has formally signed off on a $89 million class action settlement for Thalidomide victims in Australia and New Zealand.
  176. [176]
    Australia, NZ thalidomide victims win $81 mln settlement | Reuters
    More than 100 Australian and New Zealand victims of thalidomide have won a A$89 million ($81 million) settlement from the parent of the company that ...
  177. [177]
    Seeking justice for thalidomide survivors | About - Torys LLP
    Jul 4, 2025 · Since then, countless lawsuits have been filed in Canada, the United States and Australia against the manufacturers, administrators, and ...
  178. [178]
    Why thalidomide survivors have such a tough time getting ...
    Dec 8, 2015 · In 2014, a court ruled that Grunenthal should pay around 35 million euros to 22 Spaniards affected by thalidomide. Tap to unmute. Your ...
  179. [179]
    Australia thalidomide victims win $81m payout | News - Al Jazeera
    Dec 3, 2013 · Grunenthal, which apologised to thalidomide victims last year, has said it has paid about $680.77 million as compensation by 2010.Missing: scandal | Show results with:scandal<|separator|>
  180. [180]
    German thalidomide maker Gruenenthal issues apology - BBC News
    Sep 1, 2012 · The maker of thalidomide issues its first apology in 50 years to people born with disabilities as a result of their mothers taking the drug.
  181. [181]
    Thalidomide apology insulting, campaigners say - BBC News
    Sep 1, 2012 · German-based Gruenenthal has issued its first apology in 50 years, but said the drug's possible side-effects "could not be detected" before it ...
  182. [182]
    Thalidomide Maker Apologizes After More Than 50 Years - NPR
    Aug 31, 2012 · More than a half-century after a German drugmaker took thalidomide off the market because of birth defects, the company said it was sorry.
  183. [183]
    FDA Drug Review Reforms | Mercatus Center
    Jan 9, 2020 · The direct outcomes of the amendment were longer approval times and higher premarketing costs of drug development. The drug development process ...
  184. [184]
    Lost medicines: a longer view of the pharmaceutical industry with ...
    This project catalogues over-the-counter drugs and compounds lost following implementation of the 1962 Kefauver-Harris Amendment.