Small Molecule
C42H50N8O6
1242087-93-9
[Ravidasvir is a potent, second-generation, pangenotypic direct-acting antiviral (DAA) developed for the treatment of chronic hepatitis C virus (HCV) infection. Classified as a non-structural protein 5A (NS5A) inhibitor, it targets a key viral protein essential for both RNA replication and virion assembly. In combination with the NS5B polymerase inhibitor sofosbuvir, ravidasvir has demonstrated exceptional clinical efficacy, achieving sustained virological response rates of 97% in large, diverse, multinational clinical trials. Its therapeutic profile is particularly notable for its high cure rates in historically difficult-to-treat patient populations, including those with HCV genotype 3, compensated cirrhosis, and HIV co-infection. The drug exhibits a favorable pharmacokinetic profile, characterized by once-daily oral administration, negligible metabolism, and a benign safety profile with a low incidence of serious adverse events. A key feature of its clinical pharmacology is a wide therapeutic index, which allows it to maintain efficacy even in the presence of drug-drug interactions that moderately increase its clearance.]
[The development of ravidasvir represents a paradigm shift in pharmaceutical innovation. Forged through a unique South-South collaboration led by the non-profit Drugs for Neglected Diseases initiative (DNDi), its research and development were guided by public health needs rather than market imperatives. This access-driven model, which established an affordable price target from its inception, has resulted in regulatory approvals in Malaysia, Egypt, and Russia, and inclusion on the World Health Organization's Model List of Essential Medicines. Ravidasvir stands as a critical tool in the global effort to eliminate HCV, offering a highly effective, safe, and affordable treatment option designed to facilitate large-scale public health implementation, particularly in low- and middle-income countries.]
Ravidasvir is a small molecule drug belonging to the class of direct-acting antivirals.[1] During its development, it was also identified by the codes PPI-668 and ASC16.[1]
The formal International Union of Pure and Applied Chemistry (IUPAC) name for ravidasvir is methyl N-pyrrolidin-2-yl]-3H-benzimidazol-5-yl]naphthalen-2-yl]-1H-imidazol-2-yl]pyrrolidin-1-yl]-3-methyl-1-oxobutan-2-yl]carbamate.[1] This complex nomenclature accurately describes the drug's symmetrical molecular architecture. The structure is built around a rigid, planar central core composed of linked benzimidazole, naphthalene, and imidazole heterocyclic rings. This core is flanked on both sides by identical chiral side chains, each consisting of an L-valine amino acid linked to an L-proline amino acid, which are in turn capped by methyl carbamate functional groups. The molecule possesses four defined stereocenters, all with an (S) configuration, which is critical for its specific binding to the viral target protein.[3]
[Key chemical structure identifiers are essential for its unambiguous identification in scientific literature and databases:]
The molecular formula for the ravidasvir free base is C₄₂H₅₀N₈O₆, corresponding to a molecular weight of 762.91 g/mol.[2] Elemental analysis of the compound is C, 66.12%; H, 6.61%; N, 14.69%; and O, 12.58%.[2] In its solid state, ravidasvir is described as a stable compound, suitable for shipping under ambient temperatures for several weeks. For long-term storage, conditions of -20°C in a dry, dark environment are recommended to ensure stability for months to years, with a reported stability of at least four years under such conditions.[2] Solubility data for the hydrochloride salt form indicate it is soluble in organic solvents such as dimethylformamide (DMF) and dimethyl sulfoxide (DMSO) at concentrations of 5 mg/mL.[6]
Table 1: Chemical Identifiers and Properties of Ravidasvir
Property/Identifier | Value (Free Base) | Value (Dihydrochloride Salt) | Source Database/Reference |
---|---|---|---|
DrugBank ID | DB15652 | - | DrugBank 1 |
CAS Number | 1242087-93-9 | 1303533-81-4 | ChemIDplus, DrugBank 1 |
Molecular Formula | C42H50N8O6 | C42H50N8O6⋅2HCl | PubChem, Cayman Chem 1 |
Molecular Weight | 762.91 g/mol | 835.82 g/mol | MedKoo, Axio-research 2 |
InChIKey | LCHMHYPWGWYXEL-ZYADHFCISA-N | JYLMWUZJMRNMDA-SPRBZRACSA-N | PubChem, Cayman Chem 1 |
UNII | AL3G001BI8 | HWN7376681 | FDA GSRS 1 |
ChEMBL ID | CHEMBL3121849 | - | ChEMBL 1 |
NCI Thesaurus Code | C152158 | - | NCI Thesaurus 1 |
KEGG ID | D12744 | - | KEGG 1 |
Ravidasvir is utilized in two primary chemical forms: a free base, identified by CAS number 1242087-93-9, and a dihydrochloride salt, identified by CAS number 1303533-81-4.[2] The dihydrochloride salt, with the molecular formula C₄₂H₅₀N₈O₆·2HCl and a molecular weight of 835.82 g/mol, is the form typically used for formulation and laboratory studies due to its enhanced physicochemical properties.[3]
The development of a salt form is a standard and critical step in pharmaceutical chemistry, particularly for large, complex molecules like ravidasvir that are likely to exhibit poor aqueous solubility in their free base state. By forming a dihydrochloride salt, the molecule's polarity is increased, which typically improves its solubility in water and its dissolution rate in the gastrointestinal tract. These are crucial factors for ensuring adequate and consistent absorption after oral administration, thereby enhancing the drug's bioavailability. The clinical formulation of ravidasvir, as listed by the World Health Organization (WHO), is an oral solid tablet containing 200 mg of the active substance.[1][ It is standard practice for the labeled dose to refer to the mass of the active moiety (the free base), while the actual ingredient in the tablet is the salt form, with the mass adjusted to provide the equivalent dose of the free base. This formulation strategy is a direct application of chemical principles to overcome a common drug development challenge, ensuring the potent molecule can be effectively delivered to its site of action in the body.]
Ravidasvir is classified as a direct-acting antiviral (DAA), a class of drugs that revolutionized the treatment of chronic HCV infection by directly targeting viral proteins essential for replication.[9] Specifically, ravidasvir is a highly potent, second-generation inhibitor of the HCV non-structural protein 5A (NS5A).[2] The NS5A protein is a multifunctional zinc-binding phosphoprotein that, despite lacking any known enzymatic activity, plays a central and indispensable role in the HCV life cycle.[12]
The molecular target of ravidasvir has been identified as Domain I of the NS5A protein.[12] By binding to this specific region, ravidasvir induces significant conformational changes in the protein's structure. This structural disruption impairs NS5A's ability to coordinate its multiple functions, leading to a profound antiviral effect that interrupts the viral life cycle at two distinct and critical stages [10][:]
In clinical practice, ravidasvir is used as part of a combination regimen with sofosbuvir, an inhibitor of the HCV NS5B RNA-dependent RNA polymerase.[14][ This dual-mechanism approach provides a powerful, synergistic antiviral effect by targeting two separate, essential viral proteins. This strategy not only maximizes the rate and likelihood of viral clearance but also presents a high barrier to the development of drug resistance, as the virus would need to acquire mutations in two different genes simultaneously to escape the effects of both drugs.]
The designation of ravidasvir as a "second-generation" NS5A inhibitor is a functional distinction that signifies a crucial improvement over earlier agents in its class, such as daclatasvir.[2] The primary challenge with first-generation DAAs was the selection of pre-existing or emergent viral variants with resistance-associated substitutions (RASs) that reduced drug susceptibility. A key feature of a second-generation inhibitor is its enhanced ability to overcome this challenge. Evidence confirms that ravidasvir possesses this characteristic; it effectively inhibits the replication of HCV variants that harbor RASs conferring resistance to other DAA classes.[15] This high barrier to resistance was substantiated in a clinical trial in China, where 100% of patients (76 out of 76) who had baseline NS5A RASs still achieved a cure (SVR12) when treated with a ravidasvir-containing regimen.[17][ This attribute is not merely an incremental improvement but a significant clinical advantage, making the regimen more robust and reliable, particularly in public health settings where pre-treatment resistance testing is often unavailable or impractical.]
In vitro studies using cell-based HCV replicon systems have confirmed ravidasvir's potent antiviral activity. The drug effectively inhibits HCV replication at concentrations in the picomolar to low nanomolar range, underscoring its high intrinsic potency.[6]
A defining feature of ravidasvir is its pangenotypic activity, meaning it is effective against a broad range of HCV genotypes. This is a critical attribute for a globally relevant HCV therapy, as it simplifies treatment by potentially eliminating the need for costly and complex pre-treatment genotype testing. Its in vitro potency against several key genotypes has been quantified [6][:]
[These data demonstrate exceptional potency against genotypes 1a and 1b, which are common worldwide, and potent activity against genotype 3a, which is known to be more challenging to treat with some DAA regimens. This broad spectrum of potent in vitro activity provides the fundamental pharmacological basis for the successful clinical outcomes observed across diverse patient populations.]
Ravidasvir is formulated for oral administration. Following ingestion, it is absorbed from the gastrointestinal tract, with population pharmacokinetic (PK) modeling suggesting that a model with three fixed transit absorption compartments best describes its absorption process.[9] In healthy volunteers receiving daily oral doses, plasma concentrations of ravidasvir reach steady state by the seventh day of administration.[5]
The presence of food has a modest effect on its absorption kinetics. When administered with food, the peak plasma concentration (Cmax) is reduced compared to fasting conditions. However, the total drug exposure over a 24-hour period, as measured by the area under the concentration-time curve (AUC), remains unchanged. This indicates that food slows the rate of absorption but not the overall extent, allowing the drug to be taken with or without food, which simplifies dosing instructions for patients.[5]
Pharmacokinetic studies in both healthy volunteers and HCV-infected patients have defined its key absorption and distribution parameters. In healthy subjects receiving the clinical dose of 200 mg, the median time to reach peak plasma concentration (Tmax) is 2.0 hours. The peak concentration (Cmax) averages 2,540 ng/mL, and the total 24-hour exposure (AUC0−24) is approximately 19,920 h●ng/mL.[5]
Once absorbed into the systemic circulation, ravidasvir is extensively distributed throughout the body, with an apparent volume of distribution (Vd) of approximately 100 L.[5] It is highly bound to plasma proteins, with an estimated unbound (pharmacologically active) fraction of only 1.9%.[5][ Population PK analysis has also revealed that higher plasma albumin levels are associated with a reduced central volume of distribution (]
Vc/F), a finding consistent with the drug's high affinity for this protein.[9]
Table 2: Summary of Ravidasvir Pharmacokinetic Parameters
Parameter | Value | Population | Dose | Source |
---|---|---|---|---|
Tmax (Time to Cmax) | 2.0 hours (median) | HCV Patients | 200 mg | 20 |
Cmax (Peak Concentration) | 2,540 ng/mL | Healthy Volunteers | 200 mg | 5 |
Ctrough (Trough Concentration) | 190 ng/mL | Healthy Volunteers | 200 mg | 5 |
AUC0−24 (Total Exposure) | 19,920 h●ng/mL | Healthy Volunteers | 200 mg | 5 |
17.3 µg●hr/mL (median) | HCV Patients | 200 mg | 20 | |
T1/2 (Plasma Half-life) | 7.37 hours | Healthy Volunteers | 200 mg | 5 |
6.6 hours (median) | HCV Patients | 200 mg | 20 | |
Vd (Volume of Distribution) | ~100 L | Not Specified | Not Specified | 5 |
CL/F (Apparent Oral Clearance) | 11.6 L/hr (median) | HCV Patients | 200 mg | 20 |
Protein Binding | >98% (1.9% unbound) | Not Specified | Not Specified | 5 |
The metabolic profile of ravidasvir is characterized by its simplicity and stability. The drug undergoes negligible metabolism in the body, with only minor mono-oxidation products observed.[5][ This lack of significant metabolic transformation means that the parent drug is the primary active entity.]
Consistent with its minimal metabolism, ravidasvir is primarily eliminated from the body unchanged. The main route of excretion is via biliary secretion into the feces.[9] Renal clearance is negligible, meaning very little of the drug is excreted through the kidneys into the urine.[5] This elimination pathway has important clinical implications, as it suggests that the drug's pharmacokinetics are unlikely to be affected by renal impairment. Consequently, dose adjustments are not expected to be necessary for patients with kidney disease, which simplifies treatment for a significant patient subpopulation.[5]
The plasma half-life (T1/2) of ravidasvir is approximately 7.4 hours in healthy volunteers and has a median of 6.6 hours in HCV-infected patients, supporting a once-daily dosing regimen.[5] The median apparent oral clearance (CL/F) in patients is 11.6 L/hr.[20]
A comprehensive population pharmacokinetic analysis, utilizing data from the large and diverse cohort of the STORM-C-1 trial, provided deeper understanding into the factors influencing ravidasvir's disposition in patients. The analysis confirmed that a two-compartment model with first-order elimination best described the drug's concentration-time data.[9][ The study identified several patient characteristics, or covariates, that significantly influenced its pharmacokinetic parameters:]
The most profound finding from this analysis is the relationship between the observed pharmacokinetic interactions and clinical outcomes. While the increased clearance of ravidasvir due to P-gp inducers like efavirenz is a statistically significant and pharmacokinetically observable drug-drug interaction, it did not lead to a reduction in clinical efficacy. The cure rate in the STORM-C-1 trial remained exceptionally high (97%) across all subgroups, including the 30% of patients with HIV co-infection who were on various ART regimens.[22] The study investigators concluded that the effect on ravidasvir exposure was "not clinically relevant".[9][ This apparent disconnect between a PK interaction and its clinical consequence points to a crucial property of ravidasvir: a wide therapeutic index. Its intrinsic potency is so high that even when plasma concentrations are moderately reduced by increased clearance, they remain well above the threshold required for complete viral suppression. This pharmacokinetic robustness is a major clinical and public health advantage. It greatly simplifies the management of HIV/HCV co-infected patients—a large and important population—by eliminating the need for clinicians to make complex and potentially disruptive changes to a patient's established and effective ART regimen, a common barrier to care in resource-limited settings.]
The pivotal evidence for ravidasvir's clinical efficacy comes from the STORM-C-1 trial (NCT02961426), a large, two-stage, open-label, single-arm, phase 2/3 study conducted across multiple centers in Malaysia and Thailand.[22] The trial was designed to evaluate the combination regimen of ravidasvir (200 mg once daily) plus sofosbuvir (400 mg once daily) in a real-world-like patient population. The treatment duration was stratified by disease severity: patients without cirrhosis received a 12-week course, while those with compensated cirrhosis (Child-Pugh Class A) were treated for 24 weeks.[22]
The patient cohort enrolled in the first stage (n=301) was intentionally diverse to reflect the complex challenges of HCV treatment globally. It included patients with a wide range of HCV genotypes, predominantly genotype 3 (52%) and genotype 1a (33%), with smaller numbers of genotypes 1b (9%), 6 (5%), and 2 (1%).[22] Critically, the study included a high proportion of difficult-to-treat patients: 27% had compensated cirrhosis, 30% had HIV co-infection, and 33% had previously failed interferon-based therapy.[22]
The primary endpoint of the trial was sustained virological response 12 weeks after the completion of treatment (SVR12), which is considered a definitive cure for HCV. The results from the first stage were exceptional: in the full analysis set, an SVR12 was achieved in 97% (291 out of 300) of patients (95% Confidence Interval [CI] 94–99).[22] Final results from the complete trial, encompassing 603 patients, confirmed this high level of efficacy, with an overall SVR12 rate of 96.8%.[26]
[The robustness of the ravidasvir plus sofosbuvir regimen was further demonstrated through detailed subgroup analyses, which showed consistently high efficacy across all key patient populations, particularly those known to be challenging to treat.]
The exceptional performance of the ravidasvir/sofosbuvir combination, especially in patients with genotype 3 and cirrhosis, is a significant clinical finding. When compared to historical data from pivotal trials of other leading DAA regimens, ravidasvir appears to offer a distinct advantage in this specific population. For example, studies of sofosbuvir plus daclatasvir (ALLY study) and sofosbuvir plus velpatasvir (ASTRAL-3 study) reported SVR12 rates of 63% and 91%, respectively, in the same GT3 cirrhotic subgroup.[25][ This suggests that the ravidasvir-based regimen is not merely an alternative but may be a superior option for this critical, hard-to-treat patient group. This clinical superiority provides a compelling rationale for its adoption in national treatment guidelines, particularly in regions where genotype 3 is prevalent, strengthening the case for its use beyond purely economic considerations.]
Table 3: Summary of SVR12 Rates from the STORM-C-1 Trial (Stage 1, n=300) by Subgroup
Subgroup | Number of Patients (n) | Number Cured (SVR12) | SVR12 Rate (%) | 95% Confidence Interval | Source |
---|---|---|---|---|---|
Overall | 300 | 291 | 97.0 | 94–99 | 22 |
No Cirrhosis | 219 | 213 | 97.3 | 94–99 | 22 |
Compensated Cirrhosis | 81 | 78 | 96.3 | 90–99 | 22 |
Genotype 1a | 98 | 95 | 96.9 | 91–99 | 22 |
Genotype 1b | 27 | 27 | 100.0 | 87–100 | 22 |
Genotype 3 | 158 | 153 | 96.8 | 93–99 | 22 |
Genotype 3 with Cirrhosis | 53 | 51 | 96.2 | 87–100 | 22 |
Genotype 6 | 16 | 15 | 93.8 | 70–100 | 22 |
HIV Co-infected | 90 | 87 | 96.7 | 91–99 | 22 |
Interferon-Experienced | 99 | 95 | 96.0 | 90–99 | 22 |
To establish its efficacy in HCV genotype 4, which is highly prevalent in the Middle East and North Africa, the PYRAMID 1 trial was conducted. This large, multicenter, phase 3 registrational study enrolled 300 patients in Egypt.[28] The trial employed a complex design, stratifying patients by cirrhosis status and prior treatment experience. The core regimen was ravidasvir plus sofosbuvir for 12 weeks, with or without the addition of ribavirin. For the most difficult-to-treat group—treatment-experienced patients with cirrhosis—the duration of the triple therapy was extended to 16 weeks in one arm.[28]
The results confirmed high efficacy, with SVR12 rates ranging from 86% to 100% across the various subgroups. Notably, the cure rate was 100% in treatment-naïve patients without cirrhosis treated with the two-drug regimen, and also 100% in treatment-experienced patients with cirrhosis who received the 16-week regimen with ribavirin. These findings solidified ravidasvir's position as a highly effective component of therapy for genotype 4 infection.[28]
In the ongoing effort to simplify HCV therapy and reduce costs, the EASE trial (NCT04885855) was initiated in Malaysia. This randomized study is designed to directly compare the efficacy of an 8-week course of sofosbuvir plus ravidasvir against the standard 12-week course in non-cirrhotic patients.[19]
Preliminary results from this trial have been reported and are highly promising. The 8-week regimen achieved an SVR12 rate of 93.42%, which was statistically non-inferior to the 93.46% SVR12 rate observed in the 12-week arm.[30][ These findings suggest that for the majority of HCV patients (those without cirrhosis), the treatment duration can be safely shortened to 8 weeks without compromising the high cure rate. This has significant implications for reducing the overall cost and pill burden of therapy, further enhancing the regimen's suitability as a public health tool.]
Across its extensive clinical development program, the combination of ravidasvir and sofosbuvir has consistently demonstrated a favorable safety and tolerability profile.[22][ The adverse events reported are typically mild to moderate in severity and are consistent with those seen with other DAA regimens.]
In the large, diverse STORM-C-1 trial (n=301), the most frequently reported treatment-emergent adverse events (TEAEs) were generally non-specific and self-limiting [22][:]
Similarly, in the PYRAMID 1 trial, which focused on patients with genotype 4, the most common adverse events were also mild and included headache (13%), abdominal discomfort (6%), fatigue (5%), and itching (4%).[28]
The benign nature of this safety profile is a cornerstone of the regimen's suitability for widespread public health implementation. The goal of the DNDi-led development program was to create a "public health tool" that could be deployed at scale, often in resource-limited settings.[22][ Such a tool requires a safety profile that minimizes the need for intensive monitoring or specialist intervention. The clinical trial data for ravidasvir aligns perfectly with this requirement. The absence of any significant safety signals related to major organ toxicity (e.g., hepatic, renal, cardiac) or hematological disturbances means the regimen can be prescribed and managed safely in decentralized, primary care settings. This direct link between the observed clinical trial safety data and the strategic feasibility of its intended use is critical; the safety profile is a key]
enabler[ of the public health approach, making it as important as the drug's efficacy and cost.]
Table 4: Profile of Treatment-Emergent Adverse Events (>5% Incidence) in Key Trials
Adverse Event | STORM-C-1 (n=301) Frequency (%) 22 | PYRAMID 1 (n=300) Frequency (%) 28 |
---|---|---|
Pyrexia (Fever) | 12 | <5 |
Headache | 7 | 13 |
Cough | 9 | <5 |
Upper Respiratory Tract Infection | 8 | <5 |
Abdominal Discomfort | <5 | 6 |
Fatigue | <5 | 5 |
Dizziness | 5 | Not Reported |
The incidence of serious adverse events and treatment discontinuations due to adverse events has been exceptionally low in clinical trials of ravidasvir. In the first stage of the STORM-C-1 trial, SAEs were reported in 6% of patients (19 out of 301). However, only a single event—a case of transient acute renal failure in an HIV/HCV co-infected patient—was considered possibly related to the study drugs.[26]
Crucially, there were no deaths attributed to the study medication, and no patients discontinued treatment due to drug-related SAEs in this 301-patient cohort.[22] Analysis of the full 603-patient trial population reinforced this excellent safety record, with treatment-related TEAEs leading to permanent discontinuation occurring in less than 1% of patients (2 individuals).[26][ This remarkably low rate of severe toxicity and high level of tolerability underscores the regimen's safety and suitability for broad application, even in patients with comorbidities.]
Systematic monitoring of laboratory parameters in clinical trials has not revealed any consistent or clinically meaningful patterns of abnormalities associated with ravidasvir treatment. In a study combining ravidasvir with danoprevir/ritonavir and ribavirin, observed laboratory abnormalities were mild to moderate (Grade 1-2) and not clinically significant.[17] In the PYRAMID 1 trial, a few patients who received ribavirin as part of their regimen developed anemia.[28][ This is a well-documented and expected side effect of ribavirin itself and is not attributed to ravidasvir. The ability to achieve high cure rates with a ribavirin-free regimen in most patient populations is a significant advantage of the ravidasvir plus sofosbuvir combination, avoiding the hematological toxicity and teratogenicity associated with ribavirin.]
The recommended clinical dose of ravidasvir is 200 mg administered orally once daily.[1][ It is intended for use as a component of a combination antiviral regimen and should not be used as monotherapy. The primary combination studied and recommended is with]
sofosbuvir 400 mg once daily.[9]
The tablets can be taken with or without food, as food does not significantly impact the overall drug exposure.[5][ The recommended duration of therapy is determined by the patient's cirrhosis status:]
The drug-drug interaction profile of ravidasvir is primarily dictated by its transport via the P-glycoprotein (P-gp) efflux pump; it is a substrate of P-gp.[9] Its disposition is therefore susceptible to alteration by potent inducers or inhibitors of this transporter. Importantly, ravidasvir does not appear to be a significant substrate, inhibitor, or inducer of the major cytochrome P450 (CYP450) metabolic enzymes, which simplifies its co-administration with many other drugs.[9]
[This interaction profile, characterized by a specific and predictable vulnerability to P-gp modulation but a lack of broad metabolic interactions, represents a clinically advantageous "sweet spot." It avoids the complex and numerous interactions associated with other DAA classes, such as the NS3/4A protease inhibitors, which are heavily metabolized by CYP3A4. The profile is manageable for non-specialists, who can ensure patient safety by avoiding a short, well-defined list of contraindicated medications. This feature enhances its suitability for widespread deployment in primary care and public health settings.]
Ravidasvir is positioned as a potent, pangenotypic NS5A inhibitor, a classification supported by both in vitro data and extensive clinical trial results showing high efficacy against HCV genotypes 1, 2, 3, 4, and 6.[9][ Its performance can be contextualized by comparing it to other prominent NS5A inhibitors.]
Table 5: Comparative Profile of Pangenotypic NS5A Inhibitors
Feature | Ravidasvir | Daclatasvir | Ledipasvir | Velpatasvir | Pibrentasvir |
---|---|---|---|---|---|
Pangenotypic Activity | Yes | Yes | No (GT 1, 4, 5, 6) | Yes | Yes |
Efficacy in GT3 w/ Cirrhosis (SVR12) | 96% (STORM-C-1) 22 | ~63% (ALLY) 25 | Not indicated | 91% (ASTRAL-3) 25 | >95% (SURVEYOR-II) |
Barrier to Resistance | High (2nd Gen) | Low (1st Gen) | Medium | High | Very High |
Key Clinical Advantages | High efficacy in GT3/cirrhosis; affordability; simple use with ART | First-in-class | First single-tablet regimen | First pangenotypic single-tablet regimen | 8-week duration for most; effective post-DAA failure |
A key differentiating feature among NS5A inhibitors is their resilience to resistance-associated substitutions (RASs). First-generation inhibitors like daclatasvir are susceptible to common RASs at amino acid positions 28, 30, 31, and 93, which can confer high-level resistance and cross-resistance within the class, complicating retreatment after failure.[40]
Ravidasvir, as a second-generation inhibitor, was specifically designed to have a higher barrier to resistance. It maintains activity against HCV variants that harbor RASs known to cause resistance to other DAAs.[15] This was clinically validated in a study where 100% of patients with baseline NS5A RASs were cured with a ravidasvir-containing regimen.[17] Furthermore, in vitro studies suggest that viral variants with reduced susceptibility to ravidasvir remain fully susceptible to other DAA classes (e.g., protease inhibitors), indicating that it would not compromise future retreatment options.[15] Later-generation inhibitors like velpatasvir and especially pibrentasvir also have improved resistance profiles, with pibrentasvir showing particularly robust activity against a broad range of RASs.[39]
To date, no direct, head-to-head randomized controlled trials have been published comparing the ravidasvir/sofosbuvir regimen against other WHO-recommended pangenotypic regimens like sofosbuvir/velpatasvir or glecaprevir/pibrentasvir.[33][ The absence of such trials is a direct consequence of ravidasvir's unique development model, which prioritized rapid, cost-effective development for LMICs over the expensive and lengthy process of competing in the saturated markets of the United States and Europe.]
[Despite this, the cumulative evidence from its targeted clinical trials allows for a clear positioning of ravidasvir within the therapeutic landscape. It is not merely a low-cost generic equivalent but a clinically distinct and valuable agent. Its value proposition is multifaceted: it competes on its dramatically lower price point, but also on its superior clinical performance in the difficult-to-treat GT3 cirrhotic population and its simplified use in polymedicated HIV-co-infected patients. This profile challenges the notion that affordable medicines developed outside the traditional pharmaceutical industry framework must be therapeutically inferior.]
In 2023, this clinical value was formally recognized by the WHO, which added ravidasvir to its Model List of Essential Medicines. It is included as a therapeutic alternative under the "square box" listing for pangenotypic DAAs, for use in combination with sofosbuvir, solidifying its role as a key option for national HCV treatment programs globally.[33]
The regulatory pathway for ravidasvir has been unconventional, deliberately bypassing the major regulatory agencies in high-income countries to focus on regions with the greatest public health need and where its development partners are located. There is no indication that ravidasvir has been submitted for approval to the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA).[44]
[Instead, its regulatory journey has been a targeted, country-by-country process:]
The story of ravidasvir is inseparable from the work of the Drugs for Neglected Diseases initiative (DNDi), a patient-needs-driven, non-profit Research and Development (R&D) organization. Ravidasvir's development is a landmark achievement for DNDi and a prime example of an alternative model of pharmaceutical innovation.[51] The project was conceived as a unique South-South collaboration, bringing together DNDi with public and private partners from the Global South, including Pharco Pharmaceuticals (Egypt), the Ministries of Health of Malaysia and Thailand, and Pharmaniaga (Malaysia).[27]
This partnership operated on the principle of "delinkage," which aims to separate the costs of R&D from the final price of the medicine.[52] From the project's inception, an affordable price target was a primary objective, not an afterthought. DNDi's role was comprehensive: it licensed the drug candidate from Presidio Pharmaceuticals, designed, managed, and co-funded the pivotal STORM-C-1 clinical trial, coordinated the complex multinational regulatory submissions, and established partnerships for local manufacturing and supply to ensure the final product would be accessible.[27]
[Ravidasvir's journey serves as a powerful proof-of-concept. It demonstrates that a collaborative, needs-driven innovation model can successfully bring a novel chemical entity from clinical development to regulatory approval and patient access, entirely outside the framework of major pharmaceutical companies and Western regulatory bodies. It challenges the prevailing argument that the high prices of new medicines are an unavoidable consequence of high R&D costs. The ravidasvir project shows that when R&D is structured differently—collaboratively, with a mix of public and non-profit funding, and with access as the primary goal—it is possible to produce cutting-edge medical innovation that is immediately and sustainably affordable. This has profound policy implications for future approaches to drug pricing, patent reform, and the funding of R&D for other global health crises.]
The cornerstone of the ravidasvir access strategy is its affordability. The DNDi-led partnership established an ambitious target price of US300toUS500 for a complete 12-week treatment course.[35][ At the time this target was set, it represented a dramatic reduction—in some cases by nearly 100%—from the prevailing prices of other DAA regimens in many middle-income countries.]
While formal, peer-reviewed cost-effectiveness studies comparing ravidasvir to other DAAs are not detailed in the available literature, its value proposition is clear. By combining clinical efficacy that is comparable or, in some key subgroups, potentially superior to other pangenotypic regimens with a radically lower price point, the ravidasvir/sofosbuvir combination is an inherently cost-effective intervention for health systems in LMICs.[35] It is important to note that the pricing landscape for DAAs is dynamic, and the cost of other generic regimens like sofosbuvir/velpatasvir has also decreased over time. A WHO review noted that in some MICs, the cost of ravidasvir/sofosbuvir may be similar to or even higher than sofosbuvir/velpatasvir, highlighting the need for continued price negotiations and market competition.[36]
Ultimately, the availability of an affordable, highly effective, safe, and simple-to-administer pangenotypic regimen like ravidasvir plus sofosbuvir is a critical enabler of the public health approach to HCV. It facilitates the large-scale "test-and-treat" programs necessary to find and cure the millions of people living with HCV and is a vital tool in the global effort to achieve the WHO's goal of eliminating viral hepatitis as a public health threat by 2030.[22]
[Ravidasvir has emerged as a formidable agent in the global fight against chronic hepatitis C. This comprehensive analysis confirms its identity as a potent, safe, and well-tolerated second-generation pangenotypic NS5A inhibitor. Its clinical profile is defined by high SVR12 rates, exceeding 96%, across a broad and diverse spectrum of HCV patients. Its performance is particularly noteworthy in difficult-to-treat populations, including those with genotype 3 infection and compensated cirrhosis, where cross-trial comparisons suggest it may offer advantages over other established direct-acting antiviral regimens.]
[The drug's pharmacokinetic properties are well-suited for clinical practice, allowing for once-daily oral administration without regard to food. Its negligible renal excretion and manageable drug-drug interaction profile, which is largely confined to P-gp modulation and lacks significant CYP450 involvement, simplify its use in patients with comorbidities or those on concomitant medications, especially antiretroviral therapy for HIV. The benign safety profile, characterized by mild, transient adverse events and a very low rate of serious complications, further solidifies its suitability for large-scale use.]
[Beyond its pharmacological and clinical attributes, the development of ravidasvir represents a landmark achievement in access-oriented pharmaceutical innovation. The DNDi-led South-South collaboration that brought ravidasvir to fruition serves as a powerful proof-of-concept for a public health-driven R&D model, demonstrating that novel, effective medicines can be developed affordably.]
[The journey of ravidasvir is ongoing, with several key future directions. The final, published results from the EASE trial, which is comparing 8-week versus 12-week treatment durations in non-cirrhotic patients, will be critical for further optimizing therapy to reduce costs and improve patient convenience. The successful preliminary findings of non-inferiority for the 8-week course are highly encouraging.]
[The most significant challenge and opportunity lie in the global rollout and scale-up of ravidasvir-based therapy. Leveraging its recent inclusion on the WHO Model List of Essential Medicines will be crucial for encouraging its adoption into national treatment guidelines and facilitating procurement by governments and global health agencies. Continued efforts to secure regulatory approval in more high-burden countries, particularly in Latin America and Southeast Asia, are essential to broaden its reach.]
[However, the availability of an affordable drug, while necessary, is not sufficient to achieve HCV elimination. The ravidasvir story highlights the remaining, formidable barriers. There is a profound need for sustained political will and increased domestic and international funding to support comprehensive HCV programs. This includes scaling up affordable and accessible diagnostics, such as rapid tests, to identify the millions of individuals who remain unaware of their infection. Furthermore, care delivery models must continue to be simplified and decentralized, integrating HCV testing and treatment into primary healthcare, harm reduction services, and HIV programs to reach the most vulnerable and marginalized populations.]
[In conclusion, ravidasvir is more than just another effective drug; it is a vital tool and a symbol of a more equitable approach to global health. Its success provides a blueprint for future access-oriented R&D, but its ultimate impact will depend on the collective commitment of the global community to build the health systems and financial frameworks necessary to deliver its curative potential to all who need it.]
Published at: August 20, 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.