Small Molecule
C19H17ClN2O3
248281-84-7
Laquinimod is an investigational small molecule immunomodulator classified chemically as a quinoline-3-carboxamide derivative.[1] It has been assigned the DrugBank accession number DB06685 and the Chemical Abstracts Service (CAS) Registry Number 248281-84-7.[1] For regulatory and tracking purposes, it is also identified by the Unique Ingredient Identifier (UNII) 908SY76S4G.[3]
The compound's formal chemical name, according to the International Union of Pure and Applied Chemistry (IUPAC), is 5-Chloro-N-ethyl-4-hydroxy-1-methyl-2-oxo-N-phenyl-1,2-dihydroquinoline-3-carboxamide.[3] Throughout its development and in scientific literature, Laquinimod has been referred to by numerous synonyms and codes. These include the primary development code ABR-215062 (and its variants ABR 215062), the code TV-5600, and the proposed brand name Nerventra.[1]
The molecular structure of Laquinimod is defined by the chemical formula C19H17ClN2O3.[3] Its structure is captured by standard chemical notation systems, including the Simplified Molecular Input Line Entry System (SMILES) string
CCN(C1=CC=CC=C1)C(=O)C2=C(C3=C(C=CC=C3Cl)N(C2=O)C)O and the International Chemical Identifier (InChI) key GKWPCEFFIHSJOE-UHFFFAOYSA-N.[3] These identifiers provide an unambiguous representation of its atomic composition and connectivity.
Laquinimod is a synthetic organic compound that presents as a solid, off-white to beige powder under standard conditions.[6] Its molecular weight (molar mass) is consistently reported as approximately 356.8 g/mol.[1]
The compound's solubility profile is a critical determinant of its formulation and pharmacokinetic behavior. It is poorly soluble in water, with a reported solubility of less than 1 mg/mL, but demonstrates significantly higher solubility in organic solvents like dimethyl sulfoxide (DMSO), where concentrations of up to 61 mg/mL can be achieved.[6]
Computed properties provide further insight into its potential as an orally administered therapeutic. The partition coefficient (logP), a measure of lipophilicity, is estimated to be around 2.77, suggesting a favorable balance for crossing biological membranes.[1] Other computed descriptors include a topological polar surface area (TPSA) of 60.9 Ų, one hydrogen bond donor, and three hydrogen bond acceptors.[3] These characteristics are consistent with a molecule designed for oral bioavailability and central nervous system (CNS) penetration.
Table 1: Physicochemical and Structural Properties of Laquinimod
Property | Value | Source(s) |
---|---|---|
DrugBank ID | DB06685 | 1 |
CAS Number | 248281-84-7 | 1 |
IUPAC Name | 5-chloro-N-ethyl-4-hydroxy-1-methyl-2-oxo-N-phenylquinoline-3-carboxamide | 3 |
Molecular Formula | C19H17ClN2O3 | 3 |
Molar Mass | 356.8 g/mol | 3 |
Physical Form | Off-white solid powder | 6 |
Water Solubility | <1 mg/mL (Insoluble) | 6 |
logP | 2.77 | 1 |
InChIKey | GKWPCEFFIHSJOE-UHFFFAOYSA-N | 3 |
SMILES | CCN(C1=CC=CC=C1)C(=O)C2=C(C3=C(C=CC=C3Cl)N(C2=O)C)O | 3 |
The development of Laquinimod emerged from a targeted medicinal chemistry program aimed at improving upon a predecessor compound, roquinimex (also known as linomide).[5] Roquinimex, another quinoline-3-carboxamide derivative, showed initial promise in treating multiple sclerosis (MS) but was ultimately withdrawn from Phase III trials due to unacceptable cardiovascular toxicity, including cases of serositis and myocardial infarction.[15] Laquinimod was selected from a library of over 60 analogues based on its superior potency in animal models and a more favorable preclinical toxicological profile.[16] This lineage is fundamental to understanding Laquinimod's development, as the program was a deliberate attempt to retain the therapeutic mechanism of the quinoline-3-carboxamide class while engineering out the specific toxicities that led to the failure of roquinimex. This history established a high level of scrutiny on the cardiovascular safety of Laquinimod from the outset of its clinical evaluation.
The chemical synthesis of Laquinimod involves a multi-step process starting from 2-amino-6-chlorobenzoic acid.[6] A key step involves a cyclization reaction to form an isatoic anhydride intermediate. While early literature routes employed the highly toxic reagent phosgene for this transformation, a safer industrial-scale process was developed using solid phosgene (triphosgene).[6] Subsequent steps include N-methylation of the anhydride, followed by condensation with diethyl malonate to construct the core quinoline ring system.[6] The final step involves appending the N-ethyl-N-phenylamine moiety to the carboxylic acid at the 3-position of the quinoline ring, a reaction for which pyridine was identified as an effective acid-binding agent.[6]
While the precise molecular target of Laquinimod was not fully understood for a significant portion of its clinical development, subsequent research has definitively identified the aryl hydrocarbon receptor (AhR) as its primary pharmacological target.[18] The AhR is a ligand-activated transcription factor belonging to the Per-ARNT-Sim (PAS) family of proteins.[21] Historically studied for its role in mediating the toxic effects of environmental pollutants like dioxins, the AhR is now recognized as a critical regulator of cellular differentiation, metabolism, and, most importantly for Laquinimod, the immune response.[21]
The essential role of AhR in Laquinimod's mechanism of action was conclusively demonstrated in preclinical studies. In the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, the therapeutic effects of Laquinimod—including reduction in clinical score, CNS inflammation, and demyelination—were completely abolished in AhR knockout mice (AhR−/−).[18] This finding provided unequivocal evidence that AhR activation is necessary for the drug's efficacy. Genome-wide expression analyses in Laquinimod-treated mice further substantiated this, revealing consistent and significant up-regulation of prototypical AhR target genes, such as
Cyp1a1 and Ahrr, in the spleen, blood, and brain.[18]
Activation of the AhR by Laquinimod initiates a cascade of downstream events that collectively shift the immune system from a pro-inflammatory to a more tolerogenic or anti-inflammatory state. This dual action, affecting both the innate and adaptive immune systems, forms the basis of its immunomodulatory profile.
Laquinimod exerts its primary effects on the peripheral innate immune system, particularly on antigen-presenting cells (APCs) such as dendritic cells (DCs) and monocytes.[2] By targeting AhR expressed on these cells, Laquinimod effectively "re-programs" them to adopt a tolerogenic phenotype.[19] This reprogramming involves the modulation of key intracellular signaling pathways, including nuclear factor-kappa B (NF-κB), signal transducer and activator of transcription 1 (STAT1), and mitogen-activated protein kinase (MAPK) pathways.[11] The functional consequence of this is a reduced capacity of APCs to stimulate pro-inflammatory T-cell responses.
The alteration of APC function by Laquinimod has profound secondary effects on the adaptive immune system, specifically on T-cell differentiation. By promoting a tolerogenic APC phenotype, Laquinimod suppresses the development of pathogenic, pro-inflammatory T-cell subsets that are central to the pathology of MS, most notably T helper 1 (Th1) and T helper 17 (Th17) cells.[15] This is evidenced by a significant reduction in the production of the hallmark Th17 cytokine, interleukin-17 (IL-17), in preclinical models.[18]
Simultaneously, Laquinimod promotes an anti-inflammatory environment by augmenting the numbers and function of regulatory T-cells (Tregs), which are crucial for maintaining immune homeostasis and suppressing autoimmune responses.[11] This shift in the balance—away from pathogenic Th1/Th17 cells and toward protective Tregs—is a central tenet of its immunomodulatory mechanism.
A distinguishing feature of Laquinimod's pharmacological profile is its ability to cross the blood-brain barrier and exert direct effects within the CNS, independent of its peripheral immune activity.[18] This dual-action mechanism, combining peripheral immunomodulation with direct neuroprotection, provided the core scientific rationale for investigating Laquinimod not only in inflammatory conditions like relapsing-remitting MS (RRMS) but also in diseases with a more prominent neurodegenerative component, such as primary progressive MS (PPMS) and Huntington's disease.[18] This strategic expansion of the clinical program beyond pure autoimmunity was predicated on the strong preclinical evidence for these CNS-intrinsic effects.
Within the CNS, Laquinimod modulates the activity of resident glial cells, including astrocytes and microglia.[11] In the context of neurodegenerative diseases, these cells can become chronically activated, contributing to a persistent state of neuroinflammation, tissue damage, and neuronal loss. Laquinimod has been shown to reduce this reactive gliosis.[11] A key mechanism underlying this effect is the inhibition of astrocytic NF-κB activation, a critical signaling pathway that drives the production of pro-inflammatory mediators within the CNS.[11]
Perhaps the most significant neuroprotective action of Laquinimod is its demonstrated ability to up-regulate the production of Brain-Derived Neurotrophic Factor (BDNF).[2] BDNF is a critical protein that supports the survival, growth, and differentiation of neurons and is essential for neuroplasticity and repair. By increasing BDNF levels, Laquinimod may directly counteract the neurodegenerative processes that lead to irreversible disability in diseases like MS. This specific mechanism provides a compelling biological explanation for the consistent observations of reduced brain atrophy in clinical trials.
A later but crucial discovery significantly refined the understanding of Laquinimod's pharmacology, revealing that it functions as a prodrug.[34] The parent compound, Laquinimod, is metabolized in the liver via CYP-dependent N-de-alkylation to its major metabolite, N-desethyl-laquinimod, also known as DELAQ.[34]
Further investigation revealed that DELAQ, and not the parent Laquinimod molecule, is a very potent activator of the AhR.[34] Laquinimod itself is a relatively weak agonist. This finding explains the apparent paradox of why Laquinimod's efficacy in vivo is entirely dependent on a functional AhR, despite its own modest affinity for the receptor. The biotransformation to DELAQ is therefore the critical step for its therapeutic activity. This prodrug mechanism has important clinical implications. First, it introduces a potential source of inter-patient variability, as differences in CYP3A4 metabolic activity could lead to varying levels of the active DELAQ metabolite, potentially contributing to the inconsistent efficacy observed in clinical trials. Second, it complicates the safety assessment, as the toxicological profiles of both the parent drug and the active metabolite must be considered. The structural basis for DELAQ's higher potency is thought to be its ability to form an internal hydrogen bond, resulting in a more linear and lipophilic conformation that is better suited for binding to the AhR ligand-binding pocket.[34]
Laquinimod exhibits a pharmacokinetic profile suitable for once-daily oral administration, characterized by rapid absorption, extensive distribution into tissues including the CNS, and slow elimination.[14]
Absorption: Following oral administration, Laquinimod is rapidly absorbed, with peak plasma concentrations (Tmax) typically reached within 2 hours.[15] Studies have demonstrated high oral bioavailability, in the range of 80% to 90%, ensuring efficient systemic delivery.[14] Pharmacokinetic analyses have shown that its systemic exposure is proportional to the dose administered in the range of 0.9 mg to 2.7 mg, indicating linear pharmacokinetics.[36]
Distribution: Laquinimod is extensively bound to plasma proteins, with a binding fraction greater than 98%.[14] It has a small volume of distribution, and as a small, lipophilic molecule, it readily crosses the blood-brain barrier.[28] This ability to penetrate the CNS is a cornerstone of its proposed neuroprotective mechanism of action, allowing it to directly engage with glial cells and neurons.
Metabolism and Excretion: The drug is characterized by a low rate of total clearance and a long terminal elimination half-life of approximately 80 hours.[14] This long half-life supports a convenient once-daily dosing regimen and means that steady-state plasma concentrations are achieved within approximately 14 days of initiating treatment.[36] Metabolism is the primary route of elimination and occurs predominantly in the liver.[1] The resulting metabolites are then excreted, primarily via the urine. Only a small fraction of the parent drug, estimated at 5% to 10%, is excreted unchanged.[15]
The metabolism of Laquinimod is almost exclusively mediated by the hepatic cytochrome P450 enzyme system, with CYP3A4 identified as the major enzyme responsible for its biotransformation.[1] The primary metabolic pathway is N-de-ethylation, which converts Laquinimod into its pharmacologically active metabolite, DELAQ.[34] Additional metabolic pathways include hydroxylation at various positions on the quinoline ring structure.[1]
The heavy reliance of Laquinimod on a single, highly prevalent metabolic pathway—CYP3A4—creates a significant potential for clinically relevant drug-drug interactions (DDIs). This represents a notable clinical liability, particularly for patient populations with MS or Huntington's disease, who are often treated with multiple concomitant medications for symptom management. The complexity of managing these potential interactions would have posed a challenge to its clinical use had it been approved. Since the active moiety is the metabolite DELAQ, interactions that inhibit CYP3A4 could paradoxically decrease efficacy by reducing the formation of the active drug, adding a further layer of complexity.
CYP3A4 Inhibitors: Co-administration of Laquinimod with drugs that are strong or moderate inhibitors of CYP3A4 can decrease its metabolism. This leads to an increase in the serum concentration of the parent drug, which could elevate the risk of concentration-dependent adverse effects.[1] A wide range of commonly used medications fall into this category.
CYP3A4 Inducers: Conversely, co-administration with drugs that are strong inducers of CYP3A4 can accelerate the metabolism of Laquinimod. This leads to a decrease in its serum concentration, which could potentially compromise its therapeutic efficacy.[1]
Effects of Laquinimod on Other Drugs: Laquinimod may also act as an inhibitor or inducer of other metabolic pathways, affecting the concentrations of co-administered drugs. For instance, it has been shown to have the potential to increase the serum concentration of haloperidol.[1] A dedicated DDI study was also conducted to assess its effect on a commonly used oral contraceptive combination (ethinylestradiol and levonorgestrel), indicating regulatory concern over its interaction potential.[7]
Table 2: Selected Drug-Drug Interactions Affecting Laquinimod Exposure via CYP3A4
Interacting Drug/Drug Class | Effect on Laquinimod | Example Drugs | Source(s) |
---|---|---|---|
Strong CYP3A4 Inhibitors | Metabolism can be decreased; serum concentration can be increased. | Ritonavir, Ketoconazole, Clarithromycin, Amiodarone | 1 |
Moderate CYP3A4 Inhibitors | Metabolism can be decreased; serum concentration can be increased. | Cyclosporine, Diltiazem, Erythromycin, Fluconazole | 1 |
Strong CYP3A4 Inducers | Metabolism can be increased; serum concentration can be decreased. | Rifampin, Carbamazepine, Phenytoin, Phenobarbital | 1 |
Moderate CYP3A4 Inducers | Metabolism can be increased; serum concentration can be decreased. | Apalutamide, Efavirenz, Lumacaftor, Mitotane | 1 |
The clinical development of Laquinimod was extensive, encompassing three large-scale Phase III trials in relapsing-remitting multiple sclerosis (RRMS) and a Phase II trial in primary progressive multiple sclerosis (PPMS). The program was characterized by a recurring and ultimately pivotal pattern: modest and inconsistent effects on traditional inflammatory endpoints, contrasted with more robust and consistent effects on measures of neurodegeneration, such as brain atrophy.
Table 3: Summary of Major Phase II/III Clinical Trials of Laquinimod
Trial Name (Acronym) | Phase | Condition | N (approx.) | Arms | Primary Endpoint | Key Result Summary |
---|---|---|---|---|---|---|
ALLEGRO | III | RRMS | 1,106 | Laquinimod 0.6 mg vs. Placebo | Annualized Relapse Rate (ARR) | Met: 23% reduction in ARR; 36% reduction in disability progression 8 |
BRAVO | III | RRMS | 1,332 | Laquinimod 0.6 mg vs. Placebo vs. IFN-β-1a | Annualized Relapse Rate (ARR) | Not Met: No significant ARR reduction vs. placebo in primary analysis 8 |
CONCERTO | III | RRMS | 2,199 | Laquinimod 0.6 mg vs. Placebo | Time to 3-month Confirmed Disability Progression (CDP) | Not Met: No significant effect on disability progression vs. placebo 8 |
ARPEGGIO | II | PPMS | 374 | Laquinimod 0.6/1.5 mg vs. Placebo | Percent Brain Volume Change (PBVC) | Not Met: No significant effect on brain volume loss vs. placebo 8 |
LEGATO-HD | II | Huntington's Disease | 352 | Laquinimod 0.5/1.0 mg vs. Placebo | Change in UHDRS-Total Motor Score | Not Met: No significant effect on motor symptoms 40 |
The ALLEGRO study was a 24-month, randomized, double-blind, placebo-controlled trial that enrolled 1,106 patients with RRMS across 24 countries.[5] The trial successfully met its primary endpoint, demonstrating that a once-daily oral dose of 0.6 mg Laquinimod resulted in a statistically significant 23% reduction in the annualized relapse rate (ARR) compared to placebo (0.30 vs. 0.39,
p=0.002).[8] The study also showed significant benefits on key secondary endpoints. The risk of 3-month confirmed disability progression (CDP), as measured by the Expanded Disability Status Scale (EDSS), was reduced by 36% (hazard ratio 0.64,
p=0.01).[8] Furthermore, Laquinimod treatment was associated with a 33% reduction in the rate of brain volume loss compared to placebo (
p<0.001), providing the first major clinical evidence of its potential neuroprotective effect.[8]
The BRAVO study was a similarly designed 24-month trial that enrolled 1,332 RRMS patients. It had a more complex design, comparing 0.6 mg Laquinimod against both placebo and an active comparator, interferon beta-1a (Avonex), in a rater-blinded fashion.[8] The trial delivered a significant setback, as it failed to meet its primary endpoint in the primary analysis; the reduction in ARR for Laquinimod versus placebo was not statistically significant (18% reduction,
p=0.075).[8]
This result was complicated by the discovery of an imbalance in baseline MRI characteristics between the treatment groups. A pre-specified sensitivity analysis designed to correct for this imbalance did yield a statistically significant 21% reduction in ARR (p=0.026).[8] Despite the failure on the primary endpoint, BRAVO replicated the positive signals seen in ALLEGRO on neurodegenerative measures. It demonstrated a significant reduction in brain atrophy and a 33.5% reduction in the risk of 3-month CDP compared to placebo, although the latter did not reach statistical significance (
p=0.063).[8]
In an attempt to secure regulatory approval based on the strong signal on disability progression, a third Phase III trial, CONCERTO, was initiated. This large study enrolled approximately 2,200 patients and was specifically designed with time to 3-month CDP as its primary endpoint.[48] The trial evaluated two doses, 0.6 mg and 1.2 mg. However, during the study, the 1.2 mg arm was discontinued due to an imbalance of cardiovascular events, a major safety concern.[38]
Ultimately, the CONCERTO trial failed to meet its primary endpoint. The 0.6 mg dose of Laquinimod did not significantly delay the time to 3-month CDP compared to placebo (hazard ratio 0.94, p=0.706).[8] This failure was a decisive blow to the MS program. Consistent with the previous trials, however, CONCERTO did show nominally significant positive effects on several secondary and exploratory endpoints, including a 40% reduction in brain volume change versus placebo (
p<0.0001), a 28% reduction in the risk of first relapse, and a 25% reduction in ARR.[38]
The collective results of the RRMS Phase III program revealed a central paradox. The effect of Laquinimod on markers of active inflammation, such as relapses, was modest (21-23% reduction) and inconsistent across trials. In contrast, its effect on slowing brain atrophy, a marker of the underlying neurodegenerative process, was more robust and consistently demonstrated across all three studies. A post-hoc analysis highlighted this dissociation, showing that the observed 29% reduction in disability progression in pooled data was far greater than the 5% reduction that would be predicted based on its modest effect on relapses alone.[53] This suggested that Laquinimod's primary clinical benefit might stem from a direct neuroprotective mechanism rather than potent anti-inflammatory activity. However, this created a significant regulatory hurdle, as the failure to consistently meet traditional, relapse-based primary endpoints in RRMS trials made its clinical benefit appear uncertain to regulators.
Based on the strong signal of slowing brain atrophy, the development program was expanded to include PPMS, a form of MS characterized primarily by neurodegeneration with minimal overt inflammation.[31] The ARPEGGIO trial was a proof-of-concept study in 374 PPMS patients, designed to test the neuroprotection hypothesis directly by using percent brain volume change as the primary endpoint.[31]
The trial was another significant failure. Neither the 0.6 mg nor the 1.5 mg dose of Laquinimod demonstrated a significant reduction in the rate of brain atrophy compared to placebo.[8] This outcome severely undermined the core hypothesis that the drug's most potent effect—slowing neurodegeneration—would be most beneficial in a progressive disease subtype. This failure, coupled with the negative outcome of CONCERTO, effectively ended the viability of the Laquinimod program for any form of multiple sclerosis.
The LEGATO-HD trial represented the most significant exploration of Laquinimod outside of MS, testing its potential in Huntington's disease, a fatal, inherited neurodegenerative disorder.[55] The Phase II study enrolled 352 patients and evaluated two doses (0.5 mg and 1.0 mg) against placebo over 12 months.[40] The primary endpoint was clinical: the change from baseline in the Unified Huntington's Disease Rating Scale-Total Motor Score (UHDRS-TMS).[40]
The trial's results remarkably mirrored the paradoxical findings from the MS program. Laquinimod failed to meet its primary endpoint, showing no significant effect on the progression of motor symptoms compared to placebo (p=0.4853).[40] However, the study yielded a highly significant and compelling result on a key secondary endpoint. The 1.0 mg dose of Laquinimod significantly slowed the rate of caudate volume loss, a key imaging biomarker of HD progression, by a relative -1.76% compared to placebo (
p=0.0002).[40]
This cross-disease replication of the dissociation between clinical/functional outcomes and structural imaging biomarkers provided the strongest evidence that Laquinimod's primary biological activity is geared toward preserving brain structure. While this effect was insufficient to translate into a measurable clinical benefit over the one-year trial period, it was noted as the first clinical observation of a therapeutic intervention modulating this core pathological feature of HD, suggesting that neuroinflammation is a viable therapeutic target in the disease.[41] It also reinforced the conclusion that the clinical trial designs, with their focus on functional primary endpoints over relatively short durations, may have been mismatched to the drug's true, slower-acting, structure-modifying mechanism.
Reflecting the broad initial interest in its immunomodulatory properties, Laquinimod was also investigated in other autoimmune conditions. A Phase IIa study in patients with active Crohn's Disease (NCT00737932) was completed, as were early-stage explorations in Systemic Lupus Erythematosus (SLE).[20] However, as the focus of the development program narrowed to neurology and the challenges in that area mounted, further clinical development for these indications was postponed and ultimately not pursued.[60]
The safety profile of Laquinimod was extensively characterized through a clinical program that exposed thousands of patients to the drug. While the 0.6 mg dose was generally considered to have a manageable safety profile in clinical trials, significant concerns arose from preclinical toxicology studies and from cardiovascular events observed at higher clinical doses.
A pooled analysis of the two largest Phase III trials, ALLEGRO and BRAVO, provides a robust overview of the safety profile of the 0.6 mg dose. This dataset included 983 patients treated with Laquinimod and 1,005 patients on placebo, with a mean treatment duration of over 600 days.[61]
The overall rate of adverse events (AEs) was slightly higher in the Laquinimod group (81.8%) compared to the placebo group (76.2%).[63] The rate of serious adverse events (SAEs), however, was similar between the groups (approximately 9%), as were rates of malignancies and infections.[61] Early treatment discontinuations due to AEs were infrequent but slightly higher with Laquinimod (6.4%) than placebo (4.7%).[62] The most commonly reported AEs that occurred more frequently with Laquinimod were generally mild to moderate and included headache, back pain, neck pain, arthralgia, cough, and abdominal pain.[2]
Table 4: Frequency of Common Adverse Events (≥5% in Laquinimod Arm) from Pooled ALLEGRO and BRAVO Data
Adverse Event | Laquinimod 0.6 mg (n=983) % | Placebo (n=1005) % | Source |
---|---|---|---|
Headache | 18.2 | 15.1 | 63 |
Back pain | 13.6 | 8.2 | 63 |
Arthralgia | 7.2 | 6.0 | 63 |
Alanine aminotransferase increased | 5.9 | 2.7 | 63 |
Urinary tract infection | 5.7 | 4.2 | 63 |
Cough | 5.2 | 3.1 | 63 |
Abdominal pain | 5.0 | 2.6 | 63 |
Appendicitis: An unusual and unexpected safety signal that emerged from the pooled Phase III data was a higher incidence of appendicitis. Six cases were reported in the Laquinimod group compared to only one in the placebo group.[47] While no clear causative mechanism was identified, this imbalance led to appendicitis being classified as an important identified risk of the drug.[47]
Cardiovascular Events: The most critical clinical safety finding was the emergence of cardiovascular events at doses higher than 0.6 mg/day. During the CONCERTO and ARPEGGIO trials, a number of non-fatal cardiovascular events were observed in patients receiving the 1.2 mg and 1.5 mg doses.[39] This prompted the data monitoring committees to recommend the discontinuation of all higher-dose arms across the entire clinical program.[38] This finding is particularly significant given that Laquinimod's predecessor, roquinimex, was withdrawn due to severe cardiovascular toxicity.[15] This parallel suggests a potential class effect for quinoline-3-carboxamides, where the chemical modifications in Laquinimod may have raised the threshold for toxicity but did not eliminate it entirely. This safety signal effectively capped the maximum feasible dose at 0.6 mg, precluding any exploration of whether higher doses might have yielded greater efficacy.
Liver Enzymes: A consistent and well-characterized laboratory finding was the occurrence of asymptomatic, transient elevations in liver enzymes (alanine aminotransferase and aspartate aminotransferase).[2] These elevations were typically mild, non-progressive, occurred within the first 6 to 9 months of treatment, and often stabilized or reverted to baseline levels even with continued dosing.[62] In the pivotal trials, 4.7% of Laquinimod-treated subjects experienced ALT elevations greater than three times the upper limit of normal, with no associated increases in bilirubin or signs of clinical liver failure.[2]
Inflammatory Markers: Paradoxically for an anti-inflammatory drug, Laquinimod treatment was associated with mild, asymptomatic increases in systemic inflammatory markers, including fibrinogen and C-reactive protein (CRP).[47]
Despite a clinically manageable safety profile at the 0.6 mg dose, significant safety signals from preclinical animal studies cast a long shadow over the entire development program and were ultimately decisive in its regulatory failure.
Carcinogenicity: In standard 2-year carcinogenicity studies in rats, long-term exposure to Laquinimod was associated with an increased incidence of specific tumors, namely uterine adenocarcinomas and oral cavity tumors in female rats.[14] Although the developers argued that the proposed mechanisms were species-specific and not relevant to humans, this finding created significant uncertainty about the long-term cancer risk in humans, a major point of concern for the European Medicines Agency (EMA).[14]
Teratogenicity: Laquinimod demonstrated clear teratogenic effects in rats. At exposure levels similar to the intended clinical dose, it caused urogenital malformations, specifically hypospadias.[2] These findings led to the implementation of stringent contraception requirements for all women of childbearing potential in the clinical trials.[14] This risk was another key factor in the EMA's negative assessment, as the potential for harm to an unborn child could not be excluded.[67]
The failure of Laquinimod to gain regulatory approval serves as a powerful case study in risk-benefit assessment. The decision was not driven by overt toxicity observed in the human trials, but rather by the conclusion that the drug's modest and inconsistent clinical efficacy was insufficient to outweigh the serious, albeit theoretical, long-term risks of cancer and teratogenicity extrapolated from these animal toxicology studies.[67]
The development of Laquinimod was driven by a long-standing partnership. The compound was originally discovered and developed by the Swedish company Active Biotech.[1] In 2004, Active Biotech licensed the global rights for the development and commercialization of Laquinimod to Teva Pharmaceutical Industries, a major global pharmaceutical company with a strong franchise in multiple sclerosis.[51] Teva then led the extensive and costly Phase III clinical development program.
In 2014, following the completion of the ALLEGRO and BRAVO trials, Teva submitted a Marketing Authorisation Application to the European Medicines Agency (EMA) for Laquinimod, under the proposed brand name Nerventra, for the treatment of RRMS.[5]
In January 2014, the EMA's Committee for Medicinal Products for Human Use (CHMP) adopted a negative opinion, recommending refusal of the marketing authorization.[9] After the applicant requested a re-examination, the CHMP confirmed its negative opinion in May 2014, finalizing the refusal.[8]
The CHMP's detailed reasoning reveals a classic risk-benefit calculation. The committee acknowledged that Laquinimod had a positive effect, but deemed its impact on reducing relapses to be "modest".[60] While the effect on slowing disability progression was noted as "encouraging," it was not considered sufficiently confirmed to be decisive.[67] This modest clinical benefit was then weighed against the significant potential risks identified in preclinical animal studies: the long-term risk of cancer and the teratogenic risk to an unborn child.[2] The CHMP concluded that, at the dose studied, the benefits of Nerventra did not outweigh these potential risks, leading to the refusal of marketing authorisation.[67] This outcome underscores a critical regulatory principle: a drug with significant safety questions must demonstrate a substantial and unequivocal clinical benefit to achieve a positive risk-benefit assessment.
Following the EMA's definitive rejection and the subsequent failures of the CONCERTO trial to meet its primary disability endpoint and the ARPEGGIO trial in PPMS, Teva Pharmaceutical Industries made the strategic decision to discontinue all further development of Laquinimod for multiple sclerosis.[8]
In accordance with their licensing agreement, in September 2018, Teva returned the full global development and commercialization rights for Laquinimod to its originator, Active Biotech. This transfer included the complete and extensive data package generated over more than a decade of preclinical and clinical research.[20]
Faced with a failed late-stage asset, Active Biotech executed a clever strategic pivot designed to salvage value from the Laquinimod molecule. The company shifted its focus away from systemic administration for neurodegenerative diseases and initiated a new development program for a topical eye drop formulation of Laquinimod for the treatment of inflammatory eye diseases, with non-infectious uveitis (NIU) as the lead indication.[19]
This strategy is scientifically and commercially astute. By reformulating the drug for local, topical delivery to the eye, the goal is to achieve high, therapeutically effective concentrations at the site of inflammation while minimizing systemic exposure.[69] This approach directly addresses the primary reasons for the systemic program's failure: the preclinical toxicology concerns (carcinogenicity and teratogenicity) and the cardiovascular risks, all of which are driven by systemic drug levels.
Early validation for this new strategy has been positive. The Phase I LION study (NCT06161415), conducted in healthy volunteers and patients undergoing vitrectomy, demonstrated that the Laquinimod eye drop formulation was safe and well-tolerated.[19] Critically, the study also showed that the drug successfully penetrated the eye, achieving what were considered to be therapeutically relevant concentrations in both the anterior chamber and the vitreous humor.[19] This successful proof-of-concept has allowed Active Biotech to seek a new development partner to advance the ophthalmic program into Phase II efficacy studies.[72]
The failure of Laquinimod to achieve regulatory approval for multiple sclerosis was not the result of a single negative trial but rather a culmination of factors that, taken together, created an unfavorable risk-benefit profile. The key contributing elements were:
Despite the clinical failure of Laquinimod in neurology, the extensive research program provided significant validation for the aryl hydrocarbon receptor (AhR) as a legitimate and promising therapeutic target for neuroinflammatory and neurodegenerative diseases. The consistent biological signals observed across preclinical models and human clinical trials—including the modulation of CNS-resident glial cells, the suppression of pathogenic T-cell responses, and the robust effect on brain atrophy—strongly suggest that the AhR pathway is biologically active and relevant to the pathophysiology of these diseases. The Laquinimod program has thus paved the way for future research into second-generation AhR modulators that may have greater potency, a more favorable safety profile, or a better-optimized pharmacokinetic profile.
Active Biotech's strategic decision to repurpose Laquinimod as a topical ophthalmic agent represents a logical and promising path forward. The scientific rationale is sound: leveraging a known anti-inflammatory mechanism of action in a localized disease setting (non-infectious uveitis) while mitigating the risks associated with systemic exposure. The positive safety, tolerability, and ocular biodistribution data from the Phase I LION study provide crucial early validation for this approach, demonstrating that the drug can be delivered to the target tissues at relevant concentrations without systemic concerns.[69]
The future of the Laquinimod molecule now depends on the success of this new chapter. The primary challenges will be securing a strategic partner to fund the more expensive mid- and late-stage clinical development, and subsequently, demonstrating clear clinical efficacy in well-designed Phase II and III trials in patients with uveitis or other inflammatory eye diseases. While the ambitious program in neurology has concluded, the story of Laquinimod may not be over; it may yet find a valuable clinical niche through this strategic reformulation and change in therapeutic focus.
Published at: September 23, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.