C15H14N4O
129618-40-2
Human Immunodeficiency Virus Type 1 (HIV-1) Infection
Nevirapine is a first-generation non-nucleoside reverse transcriptase inhibitor (NNRTI) and was the first agent in its class to receive regulatory approval for the treatment of Human Immunodeficiency Virus Type 1 (HIV-1) infection.[1] It functions as a potent, allosteric inhibitor of the HIV-1 reverse transcriptase enzyme, a critical component of the viral replication cycle.[2] Nevirapine is indicated for use exclusively in combination with other antiretroviral (ARV) agents for the management of HIV-1 in adults and children and has also played a significant role in strategies for the prevention of mother-to-child transmission (PMTCT) of the virus.[2]
The clinical utility of Nevirapine is defined by a distinct and challenging safety profile. The drug carries a U.S. Food and Drug Administration (FDA) Boxed Warning for severe, life-threatening, and sometimes fatal hepatotoxicity and severe cutaneous adverse reactions (SCARs), including Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN).[5] The risk of hepatotoxicity is paradoxically elevated in treatment-naïve patients with more robust immune systems. Consequently, initiation of Nevirapine is not recommended in adult females with CD4+ T-cell counts greater than 250 cells/mm³ or in adult males with CD4+ T-cell counts greater than 400 cells/mm³, unless the potential benefit is determined to unequivocally outweigh the substantial risk.[2]
Despite its established efficacy in suppressing viral load and its favorable impact on lipid profiles compared to other agents in its class, Nevirapine's low genetic barrier to resistance and its significant potential for drug-drug interactions have further limited its clinical application. In contemporary HIV management, particularly in resource-rich settings, Nevirapine has been largely superseded by newer antiretroviral agents with superior safety and resistance profiles, such as integrase strand transfer inhibitors and second-generation NNRTIs. Its use is now primarily confined to specific PMTCT protocols and as a potential alternative agent in select, carefully monitored patient populations, often in resource-limited settings where cost and historical availability remain factors.[8]
Nevirapine is a small molecule drug identified by DrugBank Accession Number DB00238 and Chemical Abstracts Service (CAS) Number 129618-40-2.[2] It was originally developed and marketed by Boehringer Ingelheim under the brand names Viramune® for immediate-release (IR) formulations and Viramune XR® for the extended-release (XR) version.[2] While these brand name products have been discontinued, Nevirapine is widely available as a generic medication from various manufacturers, including Mylan (now Viatris).[12] Common abbreviations used in clinical and research settings include NVP and NEV.[15]
Systematic chemical nomenclature for the compound includes the formal name 11-cyclopropyl-5,11-dihydro-4-methyl-6H-dipyrido[3,2-b:2',3'-e]diazepin-6-one and the IUPAC name 2-cyclopropyl-7-methyl-2,4,9,15-tetrazatricyclo[9.4.0.03,8]pentadeca-1(11),3,5,7,12,14-hexaen-10-one.[3] Structurally, Nevirapine is classified as a member of the dipyridodiazepinone chemical class and also contains a cyclopropane moiety.[10] Its molecular formula is
C15H14N4O, and it is identified by standard chemical informatics strings such as InChI and SMILES for computational purposes.[2]
Nevirapine is a white to off-white or light yellow crystalline solid or powder.[3] It has a molecular weight of approximately 266.30 g/mol.[3]
A defining feature of Nevirapine is its solubility profile. It is highly lipophilic and poorly soluble in water at neutral pH, with reported solubility of approximately 0.1 g/L.[3] However, its solubility increases significantly in acidic conditions (pH < 3).[22] It demonstrates solubility in organic solvents such as dimethyl sulfoxide (DMSO).[15] This combination of high permeability and low aqueous solubility classifies Nevirapine as a Biopharmaceutics Classification System (BCS) Class 2 drug, a characteristic that influences its formulation design and absorption kinetics.[20]
The melting point of Nevirapine is consistently reported within the range of 244°C to 249°C.[19] The compound is noted to be heat sensitive and is stable for at least four years when stored appropriately at controlled room temperature (20°C to 25°C) or under refrigeration, depending on the supplier's recommendations.[15]
Nevirapine has been approved by the U.S. FDA in three primary oral formulations to accommodate different patient populations and dosing strategies:
The development of these formulations reflects a broader trend in antiretroviral therapy aimed at improving patient adherence by simplifying dosing regimens. The original IR formulation required twice-daily dosing after a lead-in period, which can be a barrier to long-term adherence.[24] The subsequent approval of the once-daily XR formulation in 2011 represented a significant step toward reducing pill burden and improving convenience, which are critical factors for achieving the near-perfect adherence required for durable virologic suppression in HIV management.[26]
The regulatory timeline for Nevirapine highlights its status as a foundational antiretroviral agent:
Table 1: Nevirapine - Key Identifiers and Physicochemical Properties
Property | Value | Source(s) |
---|---|---|
DrugBank ID | DB00238 | 2 |
Type | Small Molecule | 10 |
CAS Number | 129618-40-2 | 10 |
IUPAC Name | 2-cyclopropyl-7-methyl-2,4,9,15-tetrazatricyclo[9.4.0.03,8]pentadeca-1(11),3,5,7,12,14-hexaen-10-one | 10 |
Molecular Formula | C15H14N4O | 2 |
Molecular Weight | 266.30 g/mol | 3 |
Appearance | White to off-white/light yellow crystalline powder | 3 |
Melting Point | 244°C - 249°C | 19 |
Water Solubility | ~0.1 g/L (at neutral pH) | 19 |
pKa | 2.8 | 20 |
BCS Class | Class 2 (High Permeability, Low Solubility) | 20 |
Nevirapine is a potent and highly selective non-nucleoside reverse transcriptase inhibitor (NNRTI) specific to HIV-1.[17] Its mechanism of action is fundamentally different from that of nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs). While NRTIs act as competitive inhibitors by binding to the polymerase active site of the reverse transcriptase (RT) enzyme, Nevirapine binds to a distinct, non-substrate allosteric site.[2]
This binding site, often referred to as the "NNRTI pocket," is a hydrophobic pocket located within the p66 subdomain of the HIV-1 RT enzyme, approximately 10 Ångstroms from the catalytic active site.[2] The binding of Nevirapine to this pocket is non-competitive with respect to the natural substrates of the enzyme (nucleoside triphosphates) and the nucleic acid template.[3] This interaction induces a critical conformational change in the three-dimensional structure of the enzyme, disrupting the catalytic site and rendering it inactive.[3] By locking the enzyme in an inactive conformation, Nevirapine effectively blocks both RNA-dependent and DNA-dependent DNA polymerase activities, thereby halting the conversion of viral RNA into proviral DNA and inhibiting viral replication.[3] One study demonstrated that it specifically inhibits the initiation of the RNA plus-strand with a 50% inhibitory concentration (
IC50) of 0.45 μM.[15]
The inhibitory activity of Nevirapine is highly specific. It does not inhibit HIV-2 RT and has no effect on eukaryotic DNA polymerases, including human DNA polymerases α, β, γ, or δ.[3] This specificity accounts for its targeted antiviral effect and its lack of certain host-cell-related toxicities, such as myelosuppression, that can be associated with less specific inhibitors of DNA synthesis.
The clinical behavior of Nevirapine is governed by its distinct pharmacokinetic profile, most notably its capacity for autoinduction.
Absorption: Nevirapine is readily and rapidly absorbed following oral administration, with an excellent oral bioavailability exceeding 90% in both healthy volunteers and HIV-1-infected individuals.[3] The absolute bioavailability is reported as 93 ± 9% for the 50 mg tablet and 91 ± 8% for the oral solution, confirming that these formulations are bioequivalent and interchangeable at doses up to 200 mg.[3] Following a single 200 mg dose, peak plasma concentrations (
Cmax) of approximately 2 µg/mL are achieved in about 4 hours (Tmax).[3] The extent of absorption is not significantly affected by food, including high-fat meals, or by co-administration of antacids, which allows for flexible dosing with respect to meals.[3]
Distribution: As a highly lipophilic compound that is essentially non-ionized at physiological pH, Nevirapine distributes widely throughout the body.[3] This is reflected in its large apparent volume of distribution (
Vdss) of 1.21 ± 0.09 L/kg.[3] Plasma protein binding is moderate at approximately 60% across a clinically relevant concentration range of 1–10 µg/mL.[3] Importantly, Nevirapine readily crosses the placenta and is found in breast milk, properties that are foundational to its use in PMTCT but also require consideration during pregnancy and lactation.[3] The drug also achieves significant concentrations in the cerebrospinal fluid (CSF), reaching levels approximately 45% of those in plasma, which corresponds to its unbound fraction and suggests it can exert antiviral pressure within this key viral sanctuary site.[3]
Metabolism and Autoinduction: Nevirapine undergoes extensive biotransformation in the liver. It is metabolized via oxidative pathways mediated primarily by the cytochrome P450 isoenzymes CYP3A4 and, to a lesser extent, CYP2B6.[2] This process yields several hydroxylated metabolites, such as 4-carboxynevirapine and 12-hydroxynevirapine, which are subsequently inactivated through glucuronide conjugation before excretion.[3]
A critical pharmacokinetic characteristic of Nevirapine is its role as a potent inducer of its own metabolic enzymes. It induces the activity of both CYP3A4 and CYP2B6 by approximately 20–25%.[3] This process, known as autoinduction, leads to a progressive increase in its own apparent oral clearance by 1.5- to 2-fold over the first two to four weeks of continuous therapy.[3] This self-induced increase in metabolism is the primary reason for the mandatory 14-day dose-escalation (lead-in) period. Starting with a lower dose allows the induction process to stabilize, preventing initially high and potentially toxic drug concentrations while ensuring that therapeutic steady-state levels are achieved and maintained once the full maintenance dose is administered. This pharmacokinetic principle is directly linked to the clinical strategy for mitigating the risk of early adverse events, particularly rash.[7] Furthermore, this potent enzyme induction is the mechanistic basis for the majority of Nevirapine's clinically significant drug-drug interactions, as it can accelerate the clearance of many co-administered medications that are also CYP3A4/2B6 substrates.[6]
Excretion: The primary route of elimination for Nevirapine and its metabolites is renal. Following a radiolabeled dose, over 80% is recovered in the urine, almost exclusively in the form of glucuronidated metabolites.[3] Less than 3% of the total dose is excreted as unchanged parent drug in the urine, indicating that direct renal excretion of active Nevirapine is minimal.[3] Fecal excretion accounts for approximately 10% of the dose.[3]
The autoinduction process has a direct impact on the drug's half-life. After a single dose, the terminal half-life is approximately 45 hours. However, with continuous multiple dosing, as its clearance increases, the steady-state half-life shortens to 25–30 hours.[3]
Table 2: Pharmacokinetic Parameters of Nevirapine in Adults
Parameter | Value | Comment/Source(s) |
---|---|---|
Bioavailability | >90% | Excellent oral absorption 3 |
Cmax (single 200 mg dose) | ~2 µg/mL | Peak plasma concentration 3 |
Tmax | ~4 hours | Time to reach peak concentration 3 |
Volume of Distribution (Vdss) | 1.21 ± 0.09 L/kg | Indicates wide tissue distribution 3 |
Plasma Protein Binding | ~60% | Moderate binding 3 |
Primary Metabolism | Hepatic (CYP3A4, CYP2B6) | Extensive oxidative metabolism 3 |
Primary Elimination Route | Renal (as metabolites) | >80% of dose excreted in urine as glucuronidated metabolites 3 |
Half-life (single dose) | ~45 hours | Before autoinduction 3 |
Half-life (steady-state) | 25–30 hours | After autoinduction is complete 3 |
In vitro, Nevirapine demonstrates potent antiviral activity against a range of laboratory and clinical isolates of HIV-1, with 50% inhibitory concentration (IC50) values typically falling between 10 and 100 nM.[3] When used in cell culture, it exhibits additive to synergistic effects when combined with agents from other antiretroviral classes, including NRTIs (e.g., zidovudine, lamivudine) and protease inhibitors (PIs), providing the rationale for its use in combination therapy.[3]
A defining pharmacodynamic limitation of Nevirapine, and all first-generation NNRTIs, is its low genetic barrier to resistance. This means that as few as a single amino acid substitution in the reverse transcriptase gene can confer high-level resistance to the drug.[3] HIV-1 isolates with 100- to 250-fold reduced susceptibility emerge rapidly both in vitro and in vivo, particularly when the drug is used as monotherapy or in suboptimal regimens.[3] In early trials where Nevirapine was administered alone, resistance mutations were detectable within one to two weeks, and by eight weeks, 100% of patients harbored virus with high-level resistance.[3] This rapid selection pressure underscores the absolute contraindication of using Nevirapine as a single agent for HIV treatment.
The key resistance-associated mutations (RAMs) that emerge under Nevirapine pressure include K103N, V106A, Y181C, Y188C, and G190A.[3] Of these, the Y181C and K103N mutations are particularly significant. There is extensive cross-resistance among the first-generation NNRTIs; virus resistant to Nevirapine is almost always resistant to efavirenz and delavirdine as well.[3] Conversely, there is no cross-resistance with the NRTI or PI classes.[3]
The specific pattern of RAMs selected by Nevirapine has profound implications for long-term treatment sequencing. While both Nevirapine and efavirenz can select for the K103N mutation, Nevirapine failure is more frequently associated with the emergence of the Y181C mutation.[3] This distinction is critical because the Y181C mutation confers significant cross-resistance to the second-generation NNRTIs, etravirine and rilpivirine, effectively compromising their utility as future treatment options.[38] In contrast, the K103N mutation, which is the hallmark of efavirenz failure, does not confer resistance to etravirine or rilpivirine.[36] This differential impact on future drug options provides a strong strategic argument for preferring efavirenz over Nevirapine as a first-line NNRTI in settings where long-term treatment sequencing is a priority. This strategic consideration, alongside its more favorable safety profile, has contributed to the preferential recommendation of efavirenz in many global treatment guidelines.[9]
Nevirapine is indicated by regulatory agencies, including the U.S. FDA, for the treatment of HIV-1 infection in adults, adolescents, and children.[2] A cardinal rule of its use is that it must always be administered as a component of a combination antiretroviral therapy (ART) regimen, never as a single agent, to mitigate the rapid development of drug resistance.[18]
A second major application for Nevirapine is the prevention of mother-to-child transmission (PMTCT) of HIV-1. Due to its rapid absorption and ability to cross the placenta, it has been effectively used in regimens involving a single oral dose given to the mother at the onset of labor, followed by a single dose of oral suspension given to the infant within 48-72 hours of birth.[2] Numerous clinical trials have validated this approach and explored extended prophylaxis regimens for breastfed infants.[40]
The therapeutic use of Nevirapine is sharply curtailed by safety considerations, leading to specific, CD4+ count-based restrictions for treatment initiation in ART-naïve patients. Due to an unacceptably high risk of severe and potentially fatal hepatotoxicity, Nevirapine should not be initiated in:
In these patient populations, Nevirapine should only be considered if the potential benefit is judged to clearly outweigh the significant risk of liver injury.[7]
The clinical development of Nevirapine is marked by foundational trials establishing its efficacy, a landmark head-to-head comparison that defined its relative strengths and weaknesses, and subsequent analyses that clarified its long-term strategic implications.
The regulatory approval of Nevirapine was based primarily on the results of one principal clinical trial, BI 1090, along with smaller supportive studies.[34] The BI 1090 study was a large-scale, double-blind, placebo-controlled trial that enrolled 2,249 HIV-1-infected patients with advanced disease (median baseline CD4+ count of 96 cells/mm³) who were largely treatment-experienced.[3] The trial compared a regimen of Nevirapine plus lamivudine and background ART against a control arm of placebo plus lamivudine and background ART.
The primary outcome demonstrated a clear and significant virologic benefit for Nevirapine. At 48 weeks, 18.0% of patients in the Nevirapine group had achieved an undetectable viral load (HIV RNA <50 copies/mL), compared to just 1.6% in the placebo group.[3] The Nevirapine arm also showed a significantly greater increase in CD4+ cell counts over one year (a mean increase of 64 cells/mm³ vs. 22 cells/mm³ in the placebo group) and a reduction in the rate of progression to new AIDS-defining clinical events.[3] Importantly, a sub-analysis of treatment-naïve patients within the trial showed that Nevirapine was effective even in those with very high baseline viral loads, suggesting that baseline viremia was not a predictor of treatment failure, a finding that helped shape the understanding of NNRTI efficacy at the time.[2]
The 2NN study was a pivotal, large-scale, randomized trial that provided the first direct, head-to-head comparison of Nevirapine and efavirenz in ART-naïve patients.[47] The study had four arms, comparing Nevirapine dosed once-daily (QD), Nevirapine dosed twice-daily (BID), and efavirenz QD, all with a backbone of stavudine and lamivudine. A fourth arm testing a combination of Nevirapine and efavirenz was also included but was found to offer no additional benefit while increasing toxicity.[48]
At 48 weeks, the primary endpoint of treatment failure (a composite of virologic failure, disease progression, or death) showed no statistically significant difference between the efavirenz arm (37.8% failure rate) and the Nevirapine BID arm (43.7% failure rate).[48] The proportion of patients achieving a viral load below 50 copies/mL was also statistically similar across the single-NNRTI arms.[48]
However, the trial revealed important nuances. While not statistically significant in the overall population, a trend toward greater virologic potency favored efavirenz, particularly in the difficult-to-treat subgroup of patients with high baseline viral loads (>100,000 copies/mL). In this group, 61% of efavirenz-treated patients achieved virologic success, compared to 53.7% of those on Nevirapine BID.[49] Subsequent meta-analyses that included the 2NN data have concluded that efavirenz-based regimens are associated with a significantly lower risk of virologic failure compared to Nevirapine-based regimens.[9]
The 2NN study was instrumental in delineating the distinct safety and tolerability trade-offs between the two drugs. Nevirapine was associated with a significantly higher incidence of Grade 3/4 cutaneous rash and hepatotoxicity. Conversely, efavirenz was associated with a significantly higher incidence of central nervous system (CNS) side effects, such as dizziness and abnormal dreams.[49] Two deaths in the trial were attributed to Nevirapine toxicity (one from fulminant hepatitis and one from complications of SJS).[49] On the other hand, Nevirapine demonstrated a more favorable effect on lipid profiles, causing significantly greater increases in beneficial high-density lipoprotein (HDL) cholesterol compared to efavirenz, which was shown in a modeling analysis to translate to a lower long-term cardiovascular risk.[49]
Table 3: Comparative Efficacy and Safety Outcomes from the 2NN Study (Nevirapine vs. Efavirenz)
Outcome (at 48 weeks) | Nevirapine BID (N=387) | Efavirenz (N=400) | Comment/Source(s) |
---|---|---|---|
Treatment Failure (Primary Endpoint) | 43.7% | 37.8% | Difference not statistically significant 48 |
Virologic Success (<50 copies/mL) - Overall | 63.6% | 67.8% | Difference not statistically significant 49 |
Virologic Success (<50 copies/mL) - Baseline VL >100k | 53.7% | 61.3% | Trend favored efavirenz 49 |
Discontinuation due to Adverse Event | 21.2% | 15.5% | Higher discontinuation rate with Nevirapine 50 |
Grade 3/4 Rash | 3.1% | 1.8% | Higher risk with Nevirapine 49 |
Grade 3/4 Hepatotoxicity | 2.1% | 0.3% | Higher risk with Nevirapine 50 |
Grade 3/4 CNS Events | 3.6% | 5.5% | Higher risk with efavirenz 50 |
Change in HDL Cholesterol | Larger Increase | Smaller Increase | More favorable lipid profile with Nevirapine 50 |
There are no major clinical trials comparing Nevirapine head-to-head with second-generation NNRTIs like etravirine or rilpivirine for initial therapy, as these agents were developed for different clinical settings (etravirine for treatment-experienced patients, rilpivirine for select treatment-naïve patients). The most critical comparison, therefore, lies in the analysis of resistance patterns after first-line failure and the implications for sequencing therapy.
As discussed in Section 3.3, the choice of first-line NNRTI has a profound impact on the viability of second-generation NNRTIs in a subsequent regimen. Retrospective analyses of patient samples following virologic failure on a first-line NNRTI-based regimen have shown that:
This evidence demonstrates that initiating therapy with efavirenz preserves more future treatment options within the NNRTI class compared to initiating with Nevirapine. This strategic advantage is a key factor that has contributed to the decline of Nevirapine as a preferred first-line agent in global HIV treatment guidelines.
The clinical use of Nevirapine is dominated by its safety profile, which is characterized by the risk of severe, life-threatening adverse reactions. These risks are prominent enough to be highlighted in a Boxed Warning by the U.S. FDA and similar warnings by other global regulatory bodies.
Nevirapine is associated with a significant risk of severe, life-threatening, and in some cases, fatal hepatotoxicity.[5] This can manifest as fulminant hepatitis, cholestatic hepatitis, hepatic necrosis, or outright hepatic failure.[34]
Incidence, Timing, and Presentation: Symptomatic hepatic events occur in approximately 4% of patients receiving Nevirapine, compared to 1% in control groups.[45] The period of greatest risk is the first 18 weeks of therapy, with a critical window of highest incidence within the first 6 weeks.[6] The clinical presentation can be insidious, beginning with non-specific prodromal symptoms such as fatigue, malaise, anorexia, and nausea, before progressing to more definitive signs like jaundice and liver failure.[6] In many cases, the hepatotoxicity is a component of a broader hypersensitivity syndrome and is accompanied by rash, fever, and eosinophilia.[5]
Risk Factors: The risk of hepatotoxicity is not uniform across all patients. A unique and paradoxical feature of Nevirapine toxicity is its dependence on the patient's baseline immune status. The risk is significantly elevated in individuals with higher CD4+ T-cell counts—those who are immunologically healthier. This observation is likely linked to an immune-mediated mechanism, where a more robust immune system mounts a more vigorous and damaging inflammatory response to the drug or its metabolites. This hypersensitivity-like reaction explains why the drug is more dangerous in patients with stronger immune function.
Key identified risk factors include:
Monitoring and Management: Due to these risks, intensive monitoring is mandatory. Liver function tests (transaminases) should be performed at baseline and monitored frequently throughout the first 18 weeks of therapy. Transaminase levels must be checked immediately in any patient who develops a rash.[6] If a patient develops signs or symptoms of clinical hepatitis, or experiences significant transaminase elevations accompanied by systemic symptoms (e.g., rash, fever), Nevirapine must be
permanently discontinued and should never be restarted, even after recovery.[2]
Table 4: Risk Factors and Monitoring Schedule for Nevirapine-Induced Hepatotoxicity
Risk Category | Specific Risk Factor | Clinical Recommendation/Monitoring Action | Source(s) |
---|---|---|---|
Immune Status (Treatment-Naïve) | Adult Females with CD4+ > 250 cells/mm³ | Initiation not recommended unless benefit outweighs risk. | 7 |
Adult Males with CD4+ > 400 cells/mm³ | Initiation not recommended unless benefit outweighs risk. | 7 | |
Co-morbidities | Hepatitis B or C co-infection | Increased risk of symptomatic events; monitor closely. | 45 |
Monitoring Schedule | All Patients (Baseline) | Obtain baseline liver function tests (LFTs). | 51 |
All Patients (Weeks 1-18) | Intensive clinical and laboratory monitoring. | 6 | |
All Patients (Weeks 1-6) | Period of greatest risk; requires extra vigilance. | 6 | |
Symptom-Triggered Action | Any patient developing a rash | Check transaminase levels immediately. | 6 |
Event Management | Clinical hepatitis or severe LFT elevation with systemic symptoms | Permanently discontinue Nevirapine. DO NOT RESTART. | 6 |
Nevirapine is also associated with a Boxed Warning for severe, life-threatening skin reactions.[6] These include cases of Stevens-Johnson Syndrome (SJS), Toxic Epidermal Necrolysis (TEN), and Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS).[5]
The overall incidence of rash of any grade is high, reported in 15–21% of patients.[6] While most rashes are mild to moderate, severe or life-threatening (Grade 3/4) rashes occur in approximately 2–7% of patients.[6] The highest risk period for these reactions mirrors that of hepatotoxicity, occurring predominantly within the first 6 weeks of treatment.[4]
A key risk mitigation strategy is the mandatory 14-day lead-in dosing period (200 mg once daily). Clinical data have demonstrated that this dose-escalation strategy significantly reduces the overall frequency of rash and must be strictly adhered to.[7] If a rash develops during this lead-in period, the dose must not be escalated until the rash has resolved.[34]
Management of SCARs is unequivocal: any patient who develops a severe rash or a rash accompanied by constitutional symptoms (e.g., fever, blistering, oral lesions, conjunctivitis, muscle/joint aches, malaise) must permanently discontinue Nevirapine immediately and seek urgent medical evaluation.[2]
Contraindications: Nevirapine is absolutely contraindicated in the following patient populations:
Drug-Drug Interactions (DDI): Nevirapine’s DDI profile is extensive and clinically significant, driven by its dual role as both a substrate and a potent inducer of CYP3A4 and CYP2B6 enzymes.[2]
Concomitant use of St. John’s Wort, a potent CYP3A4 inducer, is not recommended as it can dramatically lower Nevirapine levels, leading to virologic failure and resistance.[6]
Table 5: Clinically Significant Drug-Drug Interactions with Nevirapine
Interacting Drug/Class | Effect on Nevirapine | Effect on Interacting Drug | Clinical Recommendation/Management | Source(s) |
---|---|---|---|---|
HIV Protease Inhibitors | ||||
Atazanavir/ritonavir | ↑ NVP levels | ↓↓↓ Atazanavir levels | Co-administration is not recommended. | 6 |
Lopinavir/ritonavir | No significant effect | ↓↓ Lopinavir levels | Increase Lopinavir/r dose. Refer to product labeling. | 6 |
Saquinavir, Indinavir | No significant effect | ↓ Saquinavir/Indinavir levels | Appropriate doses not established; avoid if possible. | 6 |
Other HIV Antivirals | ||||
Efavirenz | May be altered | May be altered | Co-administration not recommended (additive toxicity, no benefit). | 6 |
Antimicrobials | ||||
Rifampin | ↓↓↓ NVP levels | No significant effect | Do not co-administer. May cause NVP failure. | 6 |
Rifabutin | No significant effect | ↑ Rifabutin levels | Use with caution; monitor for rifabutin toxicity. | 6 |
Clarithromycin | No significant effect | ↓↓ Clarithromycin levels | May reduce efficacy; consider alternative like azithromycin. | 6 |
Ketoconazole, Itraconazole | ↑ NVP levels | ↓↓ Antifungal levels | Do not co-administer. May cause antifungal failure. | 6 |
Fluconazole | ↑ NVP levels | No significant effect | Use with caution; monitor for NVP toxicity. | 2 |
Other Agents | ||||
Methadone | No significant effect | ↓↓ Methadone levels | Monitor for opiate withdrawal; may require methadone dose increase. | 6 |
Hormonal Contraceptives | No significant effect | ↓ Estrogen/Progestin levels | Alternative or additional contraceptive methods should be used. | 6 |
Warfarin | No significant effect | May be ↑ or ↓ | Monitor anticoagulation levels closely. | 6 |
St. John's Wort | ↓↓↓ NVP levels | N/A | Do not co-administer. Risk of virologic failure. | 6 |
Correct dosing of Nevirapine is critical for both efficacy and safety, particularly adherence to the mandatory lead-in period.
Adult Dosing:
Pediatric Dosing:
Dosing in children is complex and is based on either body surface area (BSA) or body weight, with different guidelines provided by various regulatory and health organizations.24
Administration Instructions:
Table 6: Dosing Recommendations for Nevirapine in Adult and Pediatric Populations
Population / Condition | Dosing Phase | Recommended Dose | Key Instructions / Max Dose | Source(s) |
---|---|---|---|---|
Adults (IR) | Lead-in (14 days) | 200 mg once daily | Mandatory to reduce rash risk. | 24 |
Maintenance | 200 mg twice daily | Total daily dose: 400 mg. | 24 | |
Adults (XR) | Maintenance | 400 mg once daily | Must complete IR lead-in first. Swallow whole. | 24 |
Pediatrics (IR) | Lead-in (14 days) | 150 mg/m² once daily | For patients ≥15 days old. | 54 |
Maintenance | 150 mg/m² twice daily | Max total daily dose: 400 mg. | 54 | |
Pediatrics (XR) | Maintenance | BSA-based: 200, 300, or 400 mg once daily | For patients ≥6 years old after IR lead-in. | 24 |
Neonatal Prophylaxis | 3-Dose Regimen | Weight-based (e.g., 12 mg for >2 kg) | Doses at birth, 48h, and 96h post-48h dose. | 24 |
Hemodialysis | Post-dialysis | 200 mg IR supplemental dose | Administer after each dialysis session. | 7 |
Treatment Interruption >7 Days | Restarting Therapy | Restart full 14-day lead-in | 200 mg (adult) or 150 mg/m² (pediatric) once daily. | 34 |
Nevirapine presents a complex risk-benefit profile that has been refined over decades of clinical use and research. Its position in the antiretroviral armamentarium is a case study in how a drug's role evolves as its liabilities become better understood and safer alternatives emerge.
Benefits:
Risks:
The decision to use Nevirapine in the modern era of HIV therapy must be highly individualized and guided by a rigorous assessment of its risks and benefits for a specific patient.
Published at: July 26, 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.