Fact-checked by Grok 2 weeks ago

Nitisinone

Nitisinone is a synthetic reversible of the enzyme 4-hydroxyphenylpyruvate dioxygenase (HPPD), a key component in the catabolic pathway, and is primarily indicated for the treatment of hereditary type 1 (HT-1), a rare autosomal recessive disorder caused by deficiency of fumarylacetoacetate (FAH). By blocking HPPD, nitisinone prevents the accumulation of toxic metabolites such as succinylacetone, thereby reducing liver and kidney damage, halting disease progression, and averting acute hepatic failure and neurological crises associated with HT-1. It is administered orally as capsules (available in 2 mg, 5 mg, and 10 mg strengths) in combination with a strict dietary restriction of and to manage elevated levels and optimize therapeutic outcomes. The typical dosage starts at 1 mg/kg/day, divided into two doses, and may be adjusted up to 2 mg/kg/day based on monitoring of biochemical markers like succinylacetone levels. Originally developed in the by Zeneca Agrochemicals as a triketone (known as NTBC) for its broad-spectrum properties, nitisinone's potential as a therapeutic agent was discovered serendipitously through studies in animals that revealed its potent inhibition of HPPD, leading to elevated levels mimicking human metabolic disorders. This insight prompted its repurposing for HT-1, with initial clinical use reported in 1992 in Quebec, Canada, where it dramatically improved survival rates from less than 30% to over 90% when combined with diet. The U.S. (FDA) approved nitisinone (marketed as Orfadin) for HT-1 in 2002 under status, followed by approval in 2005, transforming the management of this life-threatening condition with prevalence around 1 in 100,000 births. In June 2025, the FDA approved nitisinone (marketed as Harliku tablets at 2 mg strength) as the first disease-modifying for adult patients with (AKU), another rare genetic disorder involving HPPD inhibition to reduce urinary (HGA) excretion by up to 95%, thereby alleviating symptoms like joint pain, , and cardiovascular complications. Off-label investigations continue for conditions like type 1B, where it may enhance pigmentation by elevating substrate for synthesis, though evidence remains preliminary. Treatment with nitisinone requires vigilant monitoring for adverse effects, including corneal and conjunctival abnormalities due to hypertyrosinemia, , , and potential risks of liver neoplasms or porphyric crises, underscoring its role as a targeted yet supportive in metabolic .

Medical uses

Hereditary tyrosinemia type 1

Hereditary tyrosinemia type 1 (HT-1) is an autosomal recessive disorder caused by biallelic pathogenic variants in the FAH gene, resulting in deficiency of the enzyme fumarylacetoacetate hydrolase (FAH). This deficiency impairs the final step in , leading to accumulation of toxic metabolites such as fumarylacetoacetate, succinylacetoacetate, and succinylacetone in the liver and kidneys. Succinylacetone inhibits key enzymes like porphobilinogen synthase, contributing to hepatic dysfunction, renal tubular damage, and neurological crises. Nitisinone serves as an adjunct in HT-1 by competitively inhibiting 4-hydroxyphenylpyruvate dioxygenase (HPPD), an upstream in the tyrosine catabolic pathway, thereby blocking the production of toxic metabolites like succinylacetone and preventing , renal dysfunction, and neurological crises. This inhibition elevates levels, necessitating strict dietary restriction of and to avoid complications such as corneal lesions. The recommended starting dosage of nitisinone in HT-1 is 0.5 mg/kg administered orally twice daily (total of 1 mg/kg/day), with titration up to 1 mg/kg twice daily (total of 2 mg/kg/day) based on biochemical response and clinical monitoring; adjustments are made for age and weight in infants, children, and adults. For patients aged 5 years and older with stable control, once-daily dosing may be used after confirming undetectable succinylacetone levels. Clinical outcomes with nitisinone have transformed HT-1 management, preventing , renal tubular dysfunction, and porphyria-like neurological crises while improving and liver function. Long-term survival rates have risen from less than 20% in untreated patients (with 90% mortality within 2 years) to near-normal levels (93-96% at 2-4 years) when initiated early, particularly in newborns. Pivotal trials demonstrate reduced need for , from 6.4% in historical cohorts to 3.4% in nitisinone-treated groups, with no transplants required among those starting treatment before 28 days of age. Therapy requires regular monitoring, including plasma tyrosine levels (target <500 μmol/L to minimize risks like corneal issues), succinylacetone levels (target undetectable in blood or urine), nitisinone concentrations (20-60 μmol/L trough), and liver function tests such as alpha-fetoprotein and coagulation parameters, typically every 1-3 months depending on age and stability.

Alkaptonuria

Alkaptonuria (AKU) is a rare autosomal recessive disorder caused by mutations in the HGD gene, leading to a deficiency in the enzyme homogentisate 1,2-dioxygenase (HGD), which is essential for breaking down homogentisic acid (HGA) in the tyrosine catabolic pathway. This deficiency results in the accumulation and excretion of HGA, which oxidizes to form ochronotic pigment that deposits in connective tissues, causing ochronosis—a bluish-black discoloration—and progressive arthropathy, particularly affecting the spine and large joints. The condition typically manifests in adulthood with chronic pain, stiffness, and degenerative joint disease, though urine darkening upon alkalization or exposure to air may appear in infancy. Nitisinone treats AKU by inhibiting 4-hydroxyphenylpyruvate dioxygenase (HPPD), an enzyme upstream of HGD in the tyrosine degradation pathway (as detailed in the pharmacodynamics section), thereby reducing HGA production and excretion by 95-99% while elevating plasma tyrosine levels. This substantial decrease in HGA slows the progression of ochronosis and arthropathy in adults, potentially mitigating tissue damage and improving quality of life. The recommended dosing for nitisinone in AKU is 2 mg orally once daily under the brand name Harliku, approved by the FDA on June 19, 2025, as the first specific therapy for the condition, with commercial launch in July 2025. Treatment should be initiated in adults with confirmed AKU diagnosis via genetic testing or elevated urinary HGA levels exceeding 4 mmol/day, and it is not recommended for pediatric patients due to insufficient data on long-term effects. Plasma tyrosine levels must be monitored regularly to avoid elevations above 500 μmol/L, which could lead to ocular complications (as discussed in serious adverse effects). Clinical evidence supporting nitisinone's efficacy in AKU comes from the SONIA trials conducted between 2013 and 2020, including (a dose-response study) and (a multicenter, randomized controlled trial), which demonstrated sustained HGA reduction of over 95% at doses as low as 2 mg daily without significant tyrosine-related complications when monitored appropriately. These trials, involving over 170 patients, also showed decreased ochronotic pigmentation in skin and cartilage, alongside slower arthropathy progression compared to untreated controls. The FDA approval relied on surrogate endpoints of HGA reduction from an earlier NIH-led trial, confirming safety and tolerability in adults. Key outcomes include the potential to delay joint replacements and reduce spinal involvement by interrupting HGA-mediated tissue damage, though long-term data on clinical benefits remain under evaluation in ongoing studies. Early adopters report improved pain scores and mobility after 1-3 years, but comprehensive assessments of disease modification are pending extended follow-up.

Pharmacology

Pharmacodynamics

Nitisinone, chemically known as 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione, is a β-triketone compound that structurally mimics the substrate of (HPPD, EC 1.13.11.27). It exerts its primary therapeutic effect through competitive inhibition of HPPD, the enzyme responsible for catalyzing the conversion of 4-hydroxyphenylpyruvate to homogentisate in the tyrosine catabolic pathway. This inhibition halts tyrosine degradation at an early step, preventing the formation of downstream toxic metabolites while leading to accumulation of upstream substrates, particularly tyrosine. The metabolic consequences of HPPD inhibition by nitisinone are pivotal to its efficacy in tyrosine pathway disorders. In hereditary tyrosinemia type 1 (HT-1), it blocks the production of succinylacetone, a potent inhibitor of fumarylacetoacetate hydrolase that causes hepatotoxicity and renal damage. In alkaptonuria (AKU), inhibition reduces the accumulation of homogentisic acid (HGA), mitigating ochronosis and arthropathy associated with HGA oxidation products. These effects do not completely shut down the pathway but sufficiently normalize metabolite levels when combined with dietary management. Enzyme kinetics studies demonstrate that nitisinone acts as a reversible, tight-binding competitive inhibitor of human , with an IC50 value of approximately 244 nM in functional assays. The inhibition is non-covalent, allowing dissociation of the enzyme-inhibitor complex, which supports its safety profile in chronic use. At therapeutic doses, nitisinone exhibits high selectivity for HPPD with minimal off-target effects on other dioxygenases involved in related metabolic pathways. Although originally developed as a herbicide targeting plant HPPD, it shows no significant herbicidal activity in humans due to species-specific enzyme differences and dosing regimens. Therapeutically, nitisinone's inhibition synergizes with a low-tyrosine diet to manage elevated tyrosine levels, preventing complications like corneal lesions while maintaining pathway flux for essential functions. This approach allows partial restoration of metabolic homeostasis without risking complete tyrosine catabolism blockade.

Pharmacokinetics

Nitisinone is rapidly absorbed after oral administration. Peak plasma concentrations (C<sub>max</sub>) are typically achieved 2-4 hours following a dose. The presence of slightly delays the time to peak concentration but does not substantially alter the overall extent of . The apparent is approximately 8.2 L in healthy adults. Nitisinone demonstrates high greater than 95% at 50 μM concentration and minimally penetrates the blood-brain barrier. The drug is distributed to various tissues. Metabolism of nitisinone occurs primarily in the liver through the enzyme , yielding inactive metabolites. No major active metabolites have been identified. Elimination of nitisinone is characterized by a of 54-82 hours in adults, which may be prolonged in children to up to 100 hours. Approximately 3% is excreted unchanged in the , with the majority eliminated via non-renal routes (primarily ) following minor . Steady-state concentrations are attained after 1-2 weeks of consistent dosing. In special populations, dose reductions of 50% are recommended for moderate to severe hepatic impairment to account for potential alterations in metabolism and clearance. No dose adjustment is required for mild renal impairment. Nitisinone is metabolized by , leading to notable drug interactions; strong inducers such as rifampin can reduce systemic exposure by about 40%, whereas inhibitors like may increase exposure by roughly 30%, necessitating potential dose modifications.

Adverse effects

Common adverse effects

The most common adverse effect of nitisinone is elevated plasma levels, which occur in more than 10% of patients with hereditary type 1 (HT-1) and up to 95% of patients with (AKU) in clinical trials when ary restrictions are not strictly followed. This elevation is nearly universal without a low- , as nitisinone inhibits the 4-hydroxyphenylpyruvate dioxygenase , blocking . Dietary compliance with restriction of and is essential to maintain levels below 500 μmol/L and mitigate associated risks. Hematologic effects, including (incidence 3% in HT-1 trials) and (incidence 3% in HT-1 trials), are also common and typically transient, resolving with continued therapy, dose adjustment, or . In AKU trials, occurred in 10% of patients. Regular of platelet and counts is recommended to manage these effects. Gastrointestinal effects such as , , , and are reported in patients on nitisinone, particularly with the oral suspension formulation due to its content, and are often transient and dose-related. These effects occur at rates below 1% in controlled HT-1 trials but may be more noticeable in clinical practice; and switching to capsules can alleviate them. , fever, and have been reported in post-marketing experience, which generally resolve with dose adjustment or supportive care. Management of these common effects emphasizes symptomatic relief, routine laboratory monitoring, and adherence to a to prevent tyrosine-related complications. Incidences are similar across HT-1 and AKU indications.

Serious adverse effects

Nitisinone treatment is associated with ocular toxicity primarily due to elevated plasma levels exceeding 500 μmol/L, manifesting as corneal dendritic keratopathy, , , corneal opacities, and eye pain; in clinical trials for , occurred in 15% of patients. These effects are generally reversible upon implementation of dietary restriction and use of topical lubricants. Hematologic adverse effects include severe (platelet count <50,000/μL) and anemia, reported in approximately 3% of patients, particularly during early treatment of hereditary tyrosinemia type 1 (HT-1); these are often reversible and linked to the underlying disease process. Complete blood count monitoring is recommended monthly initially, then periodically. Hepatic effects involve transient elevations in transaminases (ALT/AST >3x upper limit of normal) reported in <1% of cases, which rarely progress to ; nitisinone is contraindicated in patients with decompensated . should be monitored regularly alongside levels in HT-1 patients. Other rare serious adverse effects include porphyria-like symptoms and reactions such as (<1%). Regarding , there are limited data on nitisinone use in pregnant women to inform a drug-associated of adverse developmental outcomes. reproduction studies showed incomplete skeletal in mice and rabbits at exposures greater than the maximum recommended dose, and decreased pup survival in mice at exposures similar to human levels. The background of major birth defects and in the general U.S. population is 2-4% and 15-20%, respectively. Prevention strategies emphasize strict control of plasma tyrosine levels, targeting below 400-500 μmol/L through dietary phenylalanine and tyrosine restriction, alongside ophthalmologic slit-lamp examinations every 6-12 months or more frequently if symptoms arise; discontinuation is advised if irreversible damage occurs.

History and society

Development and discovery

Nitisinone, chemically known as 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione, was first synthesized in 1982 by David L. Lee and colleagues at Stauffer Chemical Company (a predecessor to ICI, now part of Syngenta) as part of a program to develop triketone herbicides targeting the enzyme 4-hydroxyphenylpyruvate dioxygenase (HPPD) in plants. The compound's design drew inspiration from the natural β-triketone leptospermone, a phytotoxin isolated from the Australian bottlebrush plant (Callistemon citrinus), which exhibits similar inhibitory activity against plant HPPD and causes bleaching symptoms in weeds. Initial testing revealed potent herbicidal efficacy through disruption of carotenoid biosynthesis, leading to chlorosis and necrosis in susceptible plants. Development as a was halted in the late due to its non-selective bleaching effects on crops, such as damage to , and concerning toxicity profiles observed in . In rats and dogs, chronic exposure induced elevated plasma levels () and reversible ocular lesions, including corneal opacities and , attributed to HPPD inhibition disrupting . These findings prompted Stauffer/ICI to prioritize structurally related analogs, like , for agricultural use while shelving nitisinone. The therapeutic potential of nitisinone emerged in the early 1990s through research on hereditary tyrosinemia type 1 (HT-1), a rare caused by deficiency of fumarylacetoacetate (FAH), leading to accumulation of toxic metabolites like succinylacetone. researchers, led by Lindstedt and Elisabeth Holme at the , identified that HPPD inhibition could block the upstream formation of these metabolites, effectively rescuing the phenotype in preclinical models. This insight, building on biochemical understanding of degradation, paved the way for nitisinone (also termed NTBC) as a for HT-1. Preclinical validation included studies in FAH-deficient mice, where HPPD inhibition prevented neonatal lethality and metabolite buildup, confirming the approach's viability. Compassionate use began in 1991 with the initiation of the NTBC Study at , administering nitisinone to five infants with acute HT-1; rapid normalization of succinylacetone and other metabolites was observed, averting . The NTBC study continued from 1991 to 1997 and enrolled 207 patients, demonstrating sustained biochemical control and improved survival without transplantation in most cases. In 1994, distribution rights were licensed to Swedish Orphan International AB (now , or Sobi), which focused initial European trials on neonatal and infant populations to establish dosing and safety. No significant advancements occurred until the , when investigations into nitisinone for revived interest in its broader metabolic applications.

Regulatory approvals and availability

Nitisinone received its initial regulatory approval from the () as Orfadin on February 21, 2005, for the treatment of hereditary type 1 (HT-1) in patients of , in combination with dietary restriction of and . In the United States, the () granted and approved Orfadin on January 18, 2002, for the same indication in adult and pediatric patients. These approvals established nitisinone as a standard therapy for HT-1, with indications covering all age groups following the FDA's 2002 decision. The scope of approvals expanded for (AKU). In September 2020, the EMA's Committee for Medicinal Products for Human Use issued a positive opinion for Orfadin in adult AKU patients to reduce urinary , leading to full authorization based on the SONIA-2 trial results demonstrating clinical benefits. On June 19, 2025, the FDA approved Harliku specifically for reducing urine in adult AKU patients, marking the first FDA-approved treatment for this rare condition and supported by evidence from the SONIA . Nitisinone is marketed under several brand names, including Orfadin by (capsules in 2 mg, 5 mg, 10 mg, and 20 mg strengths), Nityr by Cycle Pharmaceuticals (oral suspension ), and Harliku by Cycle Pharmaceuticals (2 mg tablets). Generic versions have been available since 2019, with FDA approvals for nitisinone capsules by manufacturers such as Novitium Pharma; additional generics followed, including a temperature-stable approved in 2022 by Dipharma and an acquisition of rights by Eton Pharmaceuticals in 2023 for further distribution. As an in both the US and EU, nitisinone benefits from regulatory incentives and is covered under reimbursement programs, though its high cost—approximately $300,000 per year—necessitates patient assistance initiatives from manufacturers to improve accessibility. It is approved in over 50 countries worldwide, including , , and several in the and , but availability remains limited in low-resource settings due to cold-chain storage requirements for suspension formulations.