Skip to main content
Elsevier Sponsored Documents logoLink to Elsevier Sponsored Documents
. 2025 May;6(5):None. doi: 10.1016/j.lanmic.2024.101002

Therapeutics for Nipah virus disease: a systematic review to support prioritisation of drug candidates for clinical trials

Xin Hui S Chan a,c,f,, Ilsa L Haeusler a, Bennett J K Choy a, Md Zakiul Hassan a,d,g, Junko Takata f, Tara P Hurst a, Luke M Jones a,b, Shanghavie Loganathan h, Elinor Harriss e, Jake Dunning a,d,i, Joel Tarning j,c, Miles W Carroll a,b, Peter W Horby a,d, Piero L Olliaro a,d
PMCID: PMC12062192  PMID: 39549708

Summary

Nipah virus disease is a bat-borne zoonosis with person-to-person transmission, a case-fatality rate of 38–75%, and well recognised potential to cause a pandemic. The first reported outbreak of Nipah virus disease occurred in Malaysia and Singapore in 1998, which has since been followed by multiple outbreaks in Bangladesh and India. To date, no therapeutics or vaccines have been approved to treat Nipah virus disease, and only few such candidates are in development. In this Review, we aim to assess the safety and efficacy of the therapeutic options (monoclonal antibodies and small molecules) for Nipah virus disease and other henipaviral diseases to support prioritisation of drug candidates for further evaluation in clinical trials. At present, sufficient evidence exists to suggest trialling 1F5, m102.4, and remdesivir (alone or in combination) for prophylaxis and early treatment of Nipah virus disease. In addition to well designed clinical efficacy trials, in-vivo pharmacokinetic–pharmacodynamic studies are needed to optimise the selection and dosing of therapeutic candidates in animal challenge and natural human infection.

Introduction

Nipah virus disease is a zoonotic infection acquired through contact with or ingestion of contaminated body fluids of infected mammals.1, 2, 3 Pteropid fruit bats (flying foxes) are the primary reservoir of Nipah virus. Secondary hosts include domestic animals4,5 (such as pigs, horses, and cows) and humans. Nipah virus also spreads through person-to-person transmission. Nipah virus disease occurs in all age groups.1,6 The clinical presentation of Nipah virus infection ranges from being asymptomatic7 to an acute respiratory syndrome and fatal encephalitis.1,6 After an incubation period of 4–14 days, fever, headache, and myalgia can occur, followed by shortness of breath and cough or confusion and seizures, which can rapidly progress to coma within 24–48 h.1,6 The case-fatality rate is estimated to be between 38% and 75%, and debilitating long-term neurological complications, such as paralysis, are common in individuals who have had Nipah virus infection.1,8

Nipah virus is part of the genus Henipavirus, along with Hendra virus, which also causes fatal encephalitis and respiratory diseases in horses and humans. Both Nipah and Hendra viruses are biosafety level 4 (BSL-4) pathogens that require the highest level of laboratory containment precautions. The other bat-borne members of the genus (Cedar and Kumasi viruses) are not known to cause human diseases.9

First identified in 1998, following an outbreak among pig farmers and abattoir workers in Malaysia10 and Singapore,11 Nipah virus is named after the Malaysian village from which the virus was first isolated. 283 cases of encephalitis and 109 deaths were recorded in that outbreak, with a case-fatality rate of 38·3%.12 The outbreak was contained with mass culling of over 1 million pigs and comprehensive modernisation of pig farming practice, including spatial separation of fruit tree plantations from pig farms.13

No further Nipah cases have been reported in Malaysia and Singapore since, and only one further outbreak of the Nipah virus Malaysia (NiV-M) strain has been reported in the Philippines in 2014,14 which was related to horse slaughter and consumption.

However, outbreaks of the Nipah virus Bangladesh (NiV-B) strain have been reported in Bangladesh3 and India (in the states of West Bengal15 and Kerala16) since 2001, with health-care workers15 and family members17 caring for infected patients emerging as another important risk group. Only NiV-B has known person-to-person transmission.

The highest mortality rates have been recorded in Bangladesh, where outbreaks occur almost every year in the winter following harvesting and consumption of contaminated raw date palm sap,2 a local delicacy. Since 2001, there have been 335 cases with 237 deaths in Bangladesh, with a case-fatality ratio of 70·7%.3 The 2023 outbreak in Bangladesh was the largest since 2015, with 14 cases and ten deaths. A second outbreak occurred 6 months later in Kerala, India, with six cases and two fatalities.18 Patient outcomes have not improved in the 25 years since the first reported outbreaks of the infection, reflecting the market failure typical of countermeasure development for a high-consequence pathogen.19

No approved vaccines or therapeutics are available for Nipah virus infection, and only a few candidates are in the development stage.20 In recognition of the urgent need for vaccines and therapeutics, Nipah virus infection has been a priority disease in the WHO R&D Blueprint since 2018.21 Clinical evaluation of vaccines and therapeutics for Nipah virus disease is restricted by the infeasibility of a controlled human infection model and small number of patients in sporadic outbreaks. Assessment of the effectiveness of therapeutics and vaccines is therefore reliant on animal challenge studies conducted in BSL-4 laboratory facilities.

This Review assessed the safety and efficacy of therapeutic options (monoclonal antibodies and small molecules) for Nipah virus and other Henipaviridae that cause human disease to support drug candidate prioritisation for evaluation in clinical trials.

Methods

This systematic review was registered prospectively on the PROSPERO database (CRD42022346563) and adheres to the PRISMA 2020 reporting guidelines (appendix pp 28–30).

Search strategy and selection criteria

An electronic literature search of the following bibliographic databases was conducted for journal articles, conference abstracts, and patents: PubMed, Ovid Embase, Ovid CAB Abstracts, Ovid Global Health, Scopus, Web of Science (all databases), and the WHO Global Index Medicus. “Henipavirus” or “Nipah” or “Hendra” along with “therapeutics” or “monoclonal” were the search terms for titles, abstracts, and subject headings, with synonyms and variant spellings serving as additional search terms.

The following trial registries were searched for clinical trials of henipavirus, Nipah virus, and Hendra virus at all stages of recruitment: Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, and the WHO International Clinical Trials Registry Platform. The Trip database and WHO website were searched for guidelines and reports. The full search strategies are detailed in the appendix (pp 3–4).

All searches were updated on Sept 5, 2024, and had no language or publication date criteria. The references identified were imported into EndNote, deduplicated, and then screened against eligibility criteria. Reference lists of the eligible records were checked for additional relevant studies.

Eligibility criteria

Studies containing primary data on the safety or efficacy, or both, of monoclonal antibodies (in vivo) or small molecules (in vivo or in vitro) for the treatment or prophylaxis, or both, of Nipah or Hendra infections, or both, were included. Studies on candidates without therapeutic applications (eg, monoclonal antibodies for diagnostics) or with only in-silico data were excluded.

Data extraction

We extracted data on the viruses studied, study characteristics (funder, year, location, and design), intervention characteristics (drug, dose, route, and administration timepoints), efficacy outcomes (all measures and all timepoints), and safety outcomes (all measures and all timepoints). Study investigators and experts were contacted for further information, as needed.

Data analysis

The review pilot identified substantial heterogeneity in study designs, outcome measures, and reporting. Quantitative data synthesis was not deemed possible. All available data were, therefore, prespecified to be summarised in a tabular format by individual therapeutic candidates as a narrative synthesis prioritising clinical and animal studies.

Quality assessment

Risk of Bias assessment was undertaken for the study designs using standardised tools: Risk of Bias 2 (RoB 222) for randomised clinical trials (RCTs); Risk of Bias in non-randomised studies of interventions (ROBINS-I23) for non-randomised clinical studies; and systematic review centre for laboratory animal experimentation (SYRCLE24) for animal studies.

Review team and tools

At least two independent reviewers (XHSC, ILH, BJKC, MZH, JTak, TPH, LMJ, and SL) performed screening (titles and abstracts, followed by full texts), discussed study eligibility, extracted data, and undertook Risk of Bias assessment using Covidence.

Results

Studies included

We identified 59 eligible studies (figure): 14 on monoclonal antibodies with clinical or animal data, or both (table 1 and appendix pp 7–11); 26 on small molecules with clinical or animal data, or both (table 2 and appendix pp 12–20); and 19 on small molecules with in-vitro data only (appendix pp 21–23).

Figure.

Figure

PRISMA flow chart

Table 1.

Therapeutic monoclonal antibodies against NiV and HeV by drug name (and mechanism)

Developer Reference Study design Funder
m102.4 (anti-HeV-G) Uniformed Services University, USA Sahay et al (2020)25 Clinical: compassionate use in postexposure prophylaxis during NiV outbreak in Kerala, India (n=1) US National Institutes of Health
Playford et al (2020)26 Clinical: healthy adult volunteers (18–50 years) phase 1 dose-escalation randomised controlled trial for safety, tolerability, and pharmacokinetics in Brisbane, Australia (n=40)
Mire et al (2016)27 Animal: African green monkey challenge with NiV-B for efficacy and safety (n=11)
 2.5 × 105 PFU intratracheal plus 2·5 × 105 PFU intranasal
Geisbert et al (2014)28 Animal: African green monkey challenge with NiV-M for efficacy and safety (n=16)
 5 × 105 PFU intratracheal
Bossart et al (2011)29 Animal: African green monkey challenge with HeV for efficacy and safety (n=14)
4 × 105 TCID50 intratracheal
Animal: African green monkey pharmacokinetics (n=4)
Bossart et al (2009)30 Animal: ferret challenge with NiV-M for efficacy (n=8)
 5 × 103 TCID50 oronasal
Zhu et al (2008)31 Animal: ferret pharmacokinetics (n=4)
1F5 and 12B2 (anti-NiV-F) Zeitlin et al (2024)32 Animal: African green monkey challenge with NiV-B for efficacy (n=13)
 4 × 104 PFU intranasal
Animal: hamster challenge with NiV-B for efficacy (n=16)
 5 × 106 PFU intranasal
h5B3.1 (anti-NiV-F) Mire et al (2020)33 Animal: ferret challenge with NiV-M or HeV for efficacy (n=11)
 5 × 103 PFU intranasal
NiV41 and NiV41-6 (anti-NiV-RBP) Wuhan Institute of Virology, China Chen et al (2024)34 Animal: hamster challenge with NiV-B for efficacy (n=12)
 105 TCID50 intraperitoneal
Chinese Academy of Sciences
Animal: hamster challenge with NiV-M for efficacy (n=48)
 1000 LD50 intraperitoneal
HENV-103, HENV-117, HENV-58, HENV-98, and HENV-100 (anti-HeV-RBP) Vanderbilt University, USA Doyle et al (2021)35 Animal: hamster challenge with NiV-B for efficacy (n=46)
 5 × 106 PFU intranasal
US National Institutes of Health
HENV-26 and HENV-32 (anti-HeV-RBP) Dong et al (2020)36 Animal: ferret challenge with NiV-B for efficacy (n=13)
 5 × 103 PFU intranasal
NipGIP1.7 and Nip3B10 (anti-NiV-G)
NipGIP35 and NipGIP3 (anti-NiV-F)
INSERM, France Guillaume et al (2006)37 Animal: hamster challenge with NiV-M for efficacy, dose titration, and therapeutic time window (n=124)
 7·5 × 102 PFU (100 LD50) intraperitoneal
Aventis Pharma, Bayer Pharma, INSERM, and Institut Pasteur
NipGIP35, NipGIP3, NipGIP21, and NipGIP7 (anti-NiV-F) Guillaume et al (2009)38 Animal: hamster challenge with HeV for efficacy and dose titration (n=54)
 103 PFU (100 LD50) intraperitoneal

HeV=Hendra virus. INSERM=Institut National de la Santé et de la Recherche Médicale. LD50=median lethal dose. NiV-B=Nipah virus Bangladesh. NiV-M=Nipah virus Malaysia. PFU=plaque-forming units. RBP=receptor binding protein. TCID50=median tissue culture infectious dose.

Please see appendix pp 7–11 for the full table.

Table 2.

Therapeutic small molecules against NiV and HeV by drug name (and mechanism)

Reference Study design Funder
Ribavirin (nucleoside analogue prodrug) Warrier et al (2020)39 Clinical: compassionate use for treatment in NiV outbreak in Kochi, India, 2019 (n=1) NA
Chandni et al (2020)40 Clinical: compassionate use for treatment in NiV outbreak in Kerala, India, 2018 (n=12: 6 treated, 6 untreated)
Banerjee et al (2019)41 Clinical: compassionate use for postexposure prophylaxis of health-care workers during NiV outbreak in Kerala, India, 2018 (n=8)
Kumar et al (2019)42 Clinical: compassionate use for treatment in NiV outbreak in Kerala, India, 2018 (n=5)
Playford et al (2010)43 Clinical: compassionate use during HeV outbreak in Australia, 2008, for treatment (n=2) and postexposure prophylaxis (n=1)
Chong et al (2001)44 Clinical: compassionate use for treatment in NiV outbreak in Malaysia, 1998–99
(n=194: 140 treated, 54 untreated)
Rockx et al (2010)45 Animal: African green monkey challenge with HeV (n=12)
 4 × 105 TCID50 intratracheal for efficacy
Ribavirin (nucleoside analogue prodrug), 6-azauridine (orotidine monophosphate decarboxylase inhibitor), and rintatolimod (toll-like receptor 3 agonist interferon inducer) Georges-Courbot et al (2006)46 Animal: hamster challenge with NiV-M for efficacy (n=18)
350 × LD50 intraperitoneal
Animal: hamster challenge with NiV-M for efficacy (n=18)
 35 × LD50 intraperitoneal
NA
Ribavirin (nucleoside analogue prodrug) and chloroquine (lysosome alkalinisation) Freiberg et al (2010)47 Animal: hamster challenge with NiV-M (n=41) and HeV (n=20) for efficacy (n=85)
 104 TCID50 intraperitoneal
US NIH
Chloroquine (lysosome alkalinisation) Pallister et al (2009)48 Animal: ferret challenge with NiV-M for efficacy and pharmacokinetics (n=8)
 5 × 103 TCID50 oronasal
US NIH
Remdesivir (nucleoside analogue) de Wit et al (2023)49 Animal: African green monkey challenge with NiV-B for efficacy (n=18)
 105 TCID50 intranasal plus 105 TCID50 intratracheal
US NIH
Lo et al (2019)50 Animal: African green monkey challenge with NiV-B for efficacy (n=8)
 105 TCID50 intranasal plus 105 TCID50 intratracheal
Jordan et al (2017)51 Animal: African green monkey challenge with NiV-B for efficacy
 Lethal dose (unspecified)
Favipiravir (nucleoside analogue prodrug) Dawes et al (2018)52 Animal: hamster challenge with NiV-M for efficacy (n=18)
 104 PFU intraperitoneal
US NIH
Griffithsin (fusion and cell entry inhibitor) Lo et al (2020)53 Animals: hamster challenge with NiV-B for efficacy (n=65)
 107 TCID50 intranasal
US NIH and US CDC
Periodate heparin (competitive inhibitor of transinfection) Mathieu et al (2015)54 Animal: hamster challenge with NiV-M for efficacy (n=15)
 500 × LD50 intraperitoneal
INSERM
Fusion inhibitory lipopeptides (fusion and cell entry inhibitors): VIKI-dPEG4-Chol, VIKI-dPEG4-Toco, VG-PEG24-chol, and VIKI-PEG4-chol Mathieu et al (2018)55 Animal: hamster challenge with NiV-M for efficacy (n=38)
 106 PFU (100 × LD50) intranasal
Animal: African green monkey challenge with NiV-M for efficacy (n=10)
2 × 107 PFU intratracheal
Animal: African green monkey biodistribution (n=4)
US NIH and INSERM
Mathieu et al (2017)56 Animal: hamster challenge with NiV-M for efficacy (n=13)
100 × LD50 intraperitoneal
Animal: Hamster biodistribution (n=6)
Porotto et al (2010)57 Animal: hamster challenge with NiV (strain unspecified) for efficacy (n=35)
100 × LD50 intraperitoneal
Defective interfering particles (virus-like particles containing defective interfering genomes that inhibit replication): DI-07, DI-10, DI-14, and DI-35 Welch et al (2022)58 Animal: hamster challenge with NiV-M for efficacy (n=153)
 Experiment 1: 104 TCID50 intraperitoneal
Experiment 2: 106 TCID50 intranasal
US CDC
Ceftriaxone (bacterial cell wall synthesis inhibitor), clarithromycin (bacterial protein synthesis inhibitor), and aciclovir (nucleoside analogue) Paton et al (1999)11 Clinical: empirical syndromic treatment during outbreak in Singapore, 1999 (n=11) NA

CDC=Centers for Disease Control and Prevention. dPEG=discrete polyethylene glycol. HeV=Hendra virus. INSERM=Institut National de la Santé et de la Recherche Médicale. LD50=median lethal dose. NA=not available. NIH=National Institutes of Health. NiV-B=Nipah virus Bangladesh. NiV-M=Nipah virus Malaysia. PFU=plaque-forming units. TCID50=median tissue culture infectious dose.

Please see appendix pp 12–16 for the full table.

Monoclonal antibodies

The 14 articles on monoclonal antibodies reported data on eight sets of antibodies from four research groups. Seven of the articles were on m102.4, an anti-Hendra virus (HeV)-G glycoprotein antibody (table 1 and appendix pp 7–11). m102.4 was the only Nipah drug candidate with clinical data from an RCT26 (phase 1) and in-vivo data from more than one animal species (African green monkeys27, 28, 29 and ferrets30,31) challenged with different henipaviruses (NiV-B,27 NiV-M,28,30 and HeV29; table 1).

The available data in humans support the safety of m102.4. A first-in-human dose-escalation randomised placebo-controlled trial26 of intravenous m102.4 (single doses of 1−20 mg/kg along with two doses of 20 mg/kg 72 h apart) in 40 healthy adult volunteers followed up for approximately 4 months did not report any serious adverse events. The frequency of adverse events, of which headache was the most common, was similar between the treatment and placebo groups. No anti-m102.4 antibodies were detected.

Before the trial, 14 individuals aged 8–59 years received m102.4 as postexposure prophylaxis on compassionate grounds for Hendra virus infection (n=13) in Australia and for Nipah virus infection (n=1) in the USA.26 Among the individuals, two had infusion-related febrile reactions that were attributed to an early production process of the antibody.26 In addition, one outbreak report described a single patient receiving m102.4 as postexposure prophylaxis in Kerala, India, in 2018.25 The patient was reported to have recovered completely, but no further details were provided.

Four animal challenge studies showed the efficacy of m102.4 in preventing death and severe disease in all treated animals when a single dose of m102.4 was administered to ferrets 10 h after oronasal NiV-M inoculation30 (n=3) or in a two-dose regimen administered 48 h apart in monkeys, starting within 5 days after intratracheal NiV-M28 (n=12) or HeV29 (n=12) challenge. In comparison, all control animals died within 8–10 days in the three studies. However, the treatment time window for the two-dose regimen after NiV-B challenge in monkeys27 was shorter than that after NiV-M challenge and HeV challenge, with only the animals treated within 3 days (n=6) from inoculation surviving to the end of the study and those treated on days 5 and 7 (n=2) showing similar outcomes as the controls (n=2).

The developers of m102.4 also investigated anti-NiV-F glycoprotein monoclonal antibodies: h5B.333 and 1F5.32 When administered intraperitoneally to ferrets in a regimen of two 20 mg/kg doses given 48 h apart starting within 3 days of intranasal challenge with NiV-M (n=6) or HeV (n=3), h5B.3 protected all treated ferrets from severe disease.33

1F5 was selected in favour of 12B2 (another anti-NiV-F monoclonal antibody) for its superior protection in NiV-B-infected hamsters.32 When administered intraperitoneally 24 h after intranasal inoculation, 1F5 prevented death in all five hamsters compared with the partial protection conferred after 12B2 treatment.32 1F5 also provided complete protection to NiV-B-inoculated monkeys at 5 days after challenge when administered intravenously at doses of 25 mg/kg (n=6) and 10 mg/kg (n=3), whereas intravenous m102.4 at a dose of 25 mg/kg resulted in the survival of only one of the six monkeys till the study endpoint.32 The data indicated that 1F5 has a longer treatment time window (5 days) than m102.4 (3 days) for NiV-B in monkeys.32

Anti-NiV receptor binding protein antibody NiV41 and its mature form NiV41–6 have been studied in NiV-challenged hamsters.34 NiV41–6 provided complete protection to hamsters against death at a dose of 10 mg/kg (n=6) and only partial protection at 3 mg/kg (n=6) when administered intraperitoneally 24 h before intraperitoneal NiV-M inoculation. When NiV41–6 was administered 3 h, 3 h and 3 days, and 1 day and 3 days after challenge (n=6 each group), protection was partial and declined the later the monoclonal antibody was initiated after challenge.34

Two studies35,36 described two sets of anti-HeV receptor binding protein antibodies. HENV-26 (n=5) and HENV-32 (n=5) administered intraperitoneally as two doses of 15 mg/kg on days 3 and 5 after intranasal NiV-B challenge each protected ferrets from death and severe disease compared with that observed in the controls (n=3).36 HENV-103 and HENV-117 protected all hamsters from intranasal NiV-B challenge in combination (n=5), but not individually (n=5 each), nor as two bispecific antibodies of different designs (n=5 each).35

Two other studies37,38 described two groups of anti-NiV-F and anti-NiV-G protein antibodies and provided details on protection, dose titration, and therapeutic time window; one study involved 124 hamsters37 and the other involved 54 hamsters.38 However, no further studies on these specific anti-NiV-F and anti-NiV-G protein antibodies have been conducted in the subsequent two decades.

Small molecules

In this Review, 26 articles on small molecules with in-vivo (table 2 and appendix pp 12–16) and in-vitro (appendix pp 17–20) data were identified. The articles described ten potential therapeutics and one group of syndrome-directed broad-spectrum empirical antimicrobials.

Ten of the 26 studies were on ribavirin, a repurposed nucleoside analogue prodrug, and included six clinical case series for treatment39,40,42, 43, 44 and postexposure prophylaxis41,43 for Nipah39, 40, 41, 42,44 and Hendra43 virus outbreaks (table 2); three animal challenge studies of African green monkeys (HeV45 only) and hamsters (HeV47 and NiV-M46,47; table 2); and three sets of in-vitro experiments with HeV47,59 and NiV-M46,47 (appendix pp 17–20).

The small number of patients administered ribavirin (n<10 in all, except the NiV-M outbreak in Malaysia44) and the pragmatic observational designs of the case series precluded definitive statements about clinical efficacy. Dose regimens were different among the four publications40,41,43,44 reporting on clinical efficacy and were not reported in the remaining two publications.39,42 All eight health-care workers in the only postexposure prophylaxis case series41 of ribavirin did not complete the prescribed course due to adverse effects: six of the eight had symptoms (such as fatigue or headache) or transient laboratory abnormalities (increased bilirubin or decreased haemoglobin concentrations, or both).

In the three animal studies, when administered intraperitoneally46,47 or subcutaneously45,46 at 24 h before challenge or within 12 h after challenge, ribavirin at 50–150 mg/kg per day delayed but did not prevent death or signs after NiV-M inoculation in hamsters46,47 (n=17) or HeV inoculation in African green monkeys45 (n=6), compared with that observed in the untreated controls. Ribavirin at 60 mg/kg per day neither delayed nor prevented death in HeV-challenged hamsters (n=5).47 Systemic toxicity owing to high-dose 200 mg/kg per day of intraperitoneal ribavirin was observed in infected and uninfected (control) hamsters, thus necessitating euthanasia.47

In-vitro experiments of ribavirin assessed viral replication through virus yield reduction,59 cytopathic effect,46 and dose–response47 assays in NiV-M-infected and HeV-infected Vero46,59 and HeLa47 cells. The ribavirin doses used to achieve 58-fold59 or 100% reductions46,47 in viral yield were high (50–409 μM) compared with the half-maximal inhibitory concentrations47 for NiV-M (4·18 μM) and HeV (4·96 μM).

Three studies assessed the widely used 4-aminoquinoline antimalarial chloroquine:47,48,60 two animal challenge studies47,48 (table 2) and two sets of in-vitro experiments47,60 (appendix pp 17–20). Ferrets administered 25 mg/kg per day chloroquine intravenously 24 h before (n=3) and 10 h after (n=3) NiV-M challenge presented disease courses identical to those observed in the controls.48 Hamsters inoculated with NiV-M and HeV and then treated intraperitoneally with 50 mg/kg chloroquine 6 h after challenge on alternate days as monotherapy (n=5 per virus) died earlier than the untreated controls, and hamsters treated in combination with ribavirin 30 mg/kg intraperitoneally twice a day (n=5 per virus) died at the same time as untreated controls.47 Chloroquine at 50 mg/kg per day intraperitoneally was also ineffective.47 Higher doses of 100 and 150 mg/kg per day of intraperitoneal chloroquine were consistently lethal by day 2 in both infected and uninfected hamsters.47

Four articles and one abstract on remdesivir, a nucleoside analogue, reported three NiV-B challenge studies in African green monkeys assessing intravenous administration49, 50, 51 (table 2) and in-vitro data from multiple assays on both the intravenous61 and oral62 formulations (appendix pp 17–20). Remdesivir 10 mg/kg per day administered from day 1 after challenge protected all four African green monkeys from death.50 Controls were all euthanised for disease severity after respiratory signs.50 The protective effect of remdesivir was time and dose dependent, with only partial protection when the treatment was started 3 days after challenge and declined further with dose reduction to 3 mg/kg.49 Reporter virus, cytopathic effect, and virus yield reduction assays for intravenous remdesivir (GS5734)61 and oral remdesivir (GS441524)62 and viral antigen reduction and minigenome assays for GS573461 were performed in multiple cell types, including HeLa and human small airway epithelial cells. The mean half-maximal effective concentration (EC50) values were submicromolar for both and an order of magnitude lower for GS5734 (0·029–0·066 μM)61 than those for GS441524 (0·19–0·95 μM).62

The single study on favipiravir,52 a nucleoside analogue prodrug, contained data from an NiV-M hamster challenge study (table 2) and in-vitro assays (appendix pp 17–20). Hamsters administered 600 mg/kg subcutaneous favipiravir immediately after challenge, followed by a maintenance dose of 300 mg/kg favipiravirorally twice a day (n=5) or subcutaneously daily (n=5) for 13 days, survived without clinical signs or detectable pathology or viral antigen on necropsy, whereas all the controls died or were euthanised because of disease severity by day 5–6. The doses used in the virus yield (100 μM) assay to attain 100% viral reductions and in the delayed treatment (250 μM) assay to attain ten-fold (at 1 h after infection) viral reductions were high. The EC50 values were 11·7 μM for HeV, 14·8 μM for NiV-B, and 44·2 μM for NiV-M.52

Six other groups of small molecules were studied, none of which provided complete protection from death at the doses used in the animal challenge models. 6-Azauridine, the nucleoside analogue metabolite of the previously licensed azaribine, delayed the mean time to death by approximately 1 day but did not prevent death when administered immediately before a full-dose NiV-M challenge (350 × median lethal dose [LD50]) as a 175 mg/kg per day continuous subcutaneous infusion for 14 days in hamsters.46 Rintatolimod, a toll-like receptor 3 agonist, provided partial protection at 3 mg/kg per day intraperitoneal administration for 10 days at 2 h after a low-dose (35 × LD50) NiV-M inoculation46 (table 2 and appendix pp 12–16).

Periodate heparin, an experimental glycosaminoglycan competitive inhibitor of transinfection, protected one of the five hamsters challenged with NiV-M subcutaneously at a dose of 10 mg/kg per day for 12 days from the day of infection.54 Despite promising in-vitro results, experimental cell entry inhibitors such as the lectin griffithsin (oxidation-resistant and trimeric monomer)53 and fusion inhibitory lipopeptides (cholesterol-based and tocopherol-based)55,57 administered at a dose of 10 mg/kg per day intranasally (hamsters55, 56, 57) or intratracheally (African green monkeys55) prevented death in up to half of each group of the NiV-B-challenged53 or NiV-M-challenged55,56 animals (table 2). Defective interfering virus particles administered intraperitoneally or intranasally also showed partial efficacy in NiV-M-challenged hamsters,58 whereas the virus yield reduction assays presented an order of magnitude greater reduction in NiV-M-infected Vero cells than in NiV-B-infected ones63 (appendix pp 17–20).

Risk of Bias

Most in-vivo studies had critical (six of eight case series) or high (18 of 26 animal studies) Risks of Bias. For animal studies, the high Risk of Bias was primarily because information related to most Risk of Bias domains had not been reported in study publications. Only three studies were assessed to have a low Risk of Bias: one RCT (of healthy volunteers),26 an outbreak report of a single case,39 and an NiV-B challenge study in African green monkeys.50 The remaining eight25,30,32, 33, 34,49,54,64 studies had an unclear Risk of Bias (appendix pp 26–27).

Discussion

To the best of our knowledge, this Review is the most detailed study of the therapeutics landscape for Nipah and Hendra virus diseases with the specific aim of supporting drug candidate prioritisation for clinical trials. We did not identify any ongoing or completed therapeutic efficacy RCTs for Nipah or Hendra virus infection.

The pipeline of therapeutics that can be deployed rapidly at the outset of a henipavirus outbreak is restricted to a few monoclonal antibodies and repurposed small molecules with efficacy data from animal challenge models (table 3). The comparative advantages of monoclonal antibodies and small molecules are summarised in table 4.

Table 3.

Clinical prioritisation of therapeutic candidates against NiV and HeV diseases

Efficacy Safety Feasibility Clinical prioritisation and proposed further evaluation
Monoclonal antibodies (see also table 1)
m102.4 (anti-HeV-G) Protected monkeys from death and pathology when administered as two doses 48 h apart, starting on day 1 or 3 after NiV-B27 (n=6) challenge or on day 1, 3, or 5 after NiV-M28 (n=12) and HeV29 (n=12) challenge.
Also protected ferrets from death when administered 10 h after NiV-M30 (n=3) challenge
No serious adverse events, similar rate of mild treatment emergent adverse events between treatment and placebo groups, and no anti-m102.4 antibodies in phase 1 RCT in healthy adults (n=30 treated)26 High cost of goods,20 low drug supply, parenteral route only High priority.
Phase 2a postexposure prophylaxis or early treatment RCT, or both, during NiV or HeV disease outbreak.
Shorter treatment window for NiV-B than for NiV-M.
Dose optimisation for cost recommended.
1F5 (anti-NiV-F) Protected monkeys from death and alleviated symptoms and viraemia when administered at doses of 25 mg/kg (n=6) and 10 mg/kg (n=3) on day 5 after NiV-B challenge, as compared with m102.4, which provided only partial protection at a dose of 25 mg/kg (1 of 6)32 No human studies to date.
No safety data reported in animal studies
High cost of goods,20 low drug supply, parenteral route only High priority.
Phase 1 first-in-human RCT for safety and pharmacokinetics.
Longer treatment window than that for m102.4 in animals with NiV-B.
Dose optimisation for cost recommended.
h5B.3 (anti-NiV-F) Protected ferrets from death but not minor clinical signs when administered at a dose of 20 mg/kg in two doses 48 h apart, starting on day 1 or 3 after NiV-M (n=6) or HeV (n=3) challenge33 No human studies to date. No safety data reported in animal studies. High cost of goods,20 low drug supply, parenteral route only Intermediate priority. Monkey studies with NiV-B challenge. Dose optimisation for cost necessary
NiV41-6 (anti-NiV-RBP) Protected hamsters from death when administered at a dose of 10 mg/kg (n=6) 24 h before NiV-M challenge34
HENV-26 (anti-HeV RBP) Protected ferrets (n=5) from death, symptoms, and viraemia when administered at a dose of 15 mg/kg on days 3 and 5 after NiV-B challenge36
HENV-103 plus HENV-117 (anti-HeV RBP) HENV-103 plus HENV-117 cocktail (5 mg/kg each) protected hamsters (n=5) from death when administered a day after NiV-B challenge35
Small molecules (see also table 2)
Remdesivir (nucleoside analogue) Protected monkeys (n=4) from death at a dose of 10 mg/kg when administered from day 1 after NiV-B challenge for 12 days,50 but provided only partial protection when administered from day 3 after challenge, with fewer monkeys protected at a dose of 3 mg/kg (2 of 6) than that of 10 mg/kg (4 of 6);49 in-vitro EC50 values=0·029–0·066 μM61 Sinus bradycardia and hepatotoxicity observed in humans65 Approved globally for COVID-19
Less affordable,20 parenteral route with oral formulation in development62
High priority.
Phase 2a postexposure prophylaxis or early treatment RCT, or both, during NiV or HeV outbreak.
Short treatment window for NiV-B.
Dose optimisation for efficacy recommended.
Favipiravir (nucleoside analogue) Fully protected hamsters (n=10) from death and alleviated symptoms and pathology when administered daily from time of NiV-M challenge; in-vitro EC50 values=11–44 μM52 Lethal toxicity in dogs and monkeys (>1 g/kg),66 teratogenicity across four animal species,66 transient hyperuricaemia in humans66,67 Approved in Japan for novel influenza.66
Affordable,20 oral route, but non-linear pharmacokinetics complicates dosing.68
Intermediate priority.
Monkey study with NiV-B challenge.
Dose optimisation for efficacy necessary.
Ribavirin (nucleoside analogue) Delayed but did not prevent death when administered before or within 12 h to monkeys (n=6) after HeV45 challenge and to hamsters (n=17) after NiV-M46,47 challenge; in-vitro IC50 values=4·2–5·0 μM47 Dose-dependent toxicity in hamsters (>100 mg/kg)47 and humans41 restricts safety and tolerability Approved globally for chronic hepatitis C.
Affordable20 but equivocal risk-to-benefit ratio.
Intermediate priority.
Monkey study with NiV-B challenge.
Dose optimisation for safety and efficacy crucial.
Time-to-event outcome measure when in phase 2a RCT.
Chloroquine (4-amino-quinoline) Did not protect ferrets48 (n=6) and hamsters47 (n=19) from death when administered as monotherapy before or within 12 h after NiV-M or HeV challenge Dose-dependent lethal toxicity in hamsters (>100 mg/kg)47 and humans (>3 μM plasma)69 Approved globally for malaria.
Affordable20 but unfavourable risk-to-benefit ratio.
Low priority.
Should not be used for the prophylaxis or treatment of NiV or HeV infection.

EC50=50% maximal effective concentration. HeV=Hendra virus. IC50=50% maximal inhibitory concentration. NiV-B=Nipah virus Bangladesh. NiV-M=Nipah virus Malaysia. RBP=receptor binding protein. RCT=randomised controlled trial.

High priority indicates that ample evidence exists for efficacy and safety for phase 2a trials. Intermediate priority indicates that further evidence is required for efficacy or potential major limitations in safety or feasibility, or both. Low priority indicates that no evidence exists for efficacy and major limitations in safety or feasibility, or both.

Table 4.

Comparison of the advantages of monoclonal antibodies and small molecules

Monoclonal antibodies Small molecules
Efficacy Highly specific Less specific
Safety, including in pregnancy or lactation and other immunosuppression Specificity confers low potential for off-target adverse effects
Typically safe in pregnancy or lactation and immunosuppression
Higher potential for off-target adverse effects
Often associated with hepatotoxicity, cardiotoxicity, or teratogenicity
Duration of effect Half-life-extending mutations can prolong protection from 1 month to 3–6 months Elimination half-lives of leading nucleoside analogue candidates between 1 and 5 h
Route of administration Parenteral Parenteral or oral, or both
Pharmacokinetic profile Typically linear Can be linear or non-linear
Cost Higher (US$1000–2000) Lower (US$10–100)
Development timeline Shorter Longer

Of the monoclonal antibodies, only m102.4 has been studied in humans, with safety and pharmacokinetic data from a phase 1 RCT in healthy adults.26 1F5 is also progressing to phase 1 safety testing. The longer therapeutic time window within which complete protection is seen with 1F5 (5 days) in NiV-B-infected monkeys than that with m102.4 (3 days) would be of substantial clinical interest if validated in human trials in India and Bangladesh. Further pharmacokinetic studies of monoclonal antibody candidates to identify minimal doses for efficacy could help to make scale-up more cost-effective.

Of the small molecules, animal efficacy data were supportive for remdesivir and favipiravir, equivocal for ribavirin, and not supportive for chloroquine. Remdesivir was the only small molecule with in-vivo data from challenge with NiV-B,49,50 a strain closely related to those causing the Nipah outbreaks in Bangladesh and India.70 In addition, remdesivir has accumulated acceptable safety data from its widespread intravenous use against COVID-19.71 The therapeutic time window of remdesivir seen in NiV-B-infected monkeys is short,49 with protection declining from 3 days after inoculation;49 moreover, the therapeutic time window of remdesivir requires confirmation in humans.

Although favipiravir prevented death in NiV-M-challenged hamsters after a subcutaneous loading dose followed by subcutaneous or oral maintenance doses52 and could be an attractive choice for prophylaxis after exposure with a licensed oral formulation,66 the non-linear clinical pharmacokinetics of favipiravir seen in Ebola virus disease,72 influenza,73 and COVID-1968 necessitate further dose optimisation before inclusion in trials on Nipah virus disease. The non-linearity is thought to be explained by the concentration-dependent aldehyde oxidase inhibition reproducible in non-human primates,74 and any additional infection-specific contribution remains unclear as of now. Pharmacokinetic studies of parenteral (including intravenous75) administration in NiV-B-inoculated non-human primates would be the next key step in favipiravir evaluation. Notably, favipiravir is associated with teratogenicity in four animal species,66 and further data on the safety of favipiravir in humans are needed.67,76

Ribavirin prolongs survival but does not prevent death in monkeys and hamsters challenged with HeV45 and NiV-M;46,47 in fact, ribavirin is toxic to hamsters at high doses.47 Clinical tolerability issues (fatigue, anaemia, and hyperbilirubinaemia)41 are further likely to reduce adherence to a postexposure prophylaxis regimen containing ribavirin. Clinical reports of ribavirin in henipavirus outbreaks have all been observational, with dosing based on that used for Lassa fever.44 Clinical and pharmacokinetic meta-analyses of studies on ribavirin treatment for Lassa fever highlight the absence of robust data for its effectiveness,77 and conventional dosing regimens are unlikely to reliably achieve the serum concentrations required to inhibit Lassa virus replication.78

Pharmacokinetic modelling for the existing ribavirin dosing regimens for Nipah and Hendra virus diseases is ongoing. Ribavirin remains part of the Nipah treatment guidelines in India79,80 but not in Bangladesh.81 Consultation with Nipah stakeholders would help to establish whether the unclear effectiveness of ribavirin justifies its further use as a drug to treat Nipah or Hendra virus disease, especially with the availability of more promising alternatives.

Chloroquine did not protect ferrets48 or hamsters47 from NiV-M and was lethal at high doses in hamsters.47 The narrow therapeutic dose window of chloroquine is well established in clinical practice. Despite being considered a safe and effective antimalarial drug, chloroquine has been used for rapid self-poisoning in deliberate overdoses.69,82 Chloroquine should not be used to treat or prevent Nipah or Hendra virus disease.

The promising in-vitro efficacy of the experimental small molecules listed in table 2 is yet to be translated into convincing in-vivo protection. The parent drug of 6-azauridine (azaribine) has been withdrawn from the market due to safety concerns of thrombosis,83 and the parent drug of ALS-8112 (lumicitabine) has been withdrawn from development due to safety concerns of paediatric neutropenia.84 Whether periodate heparin, fusion lipopeptides, and defective viral particles can be manufactured at scale or are stable for stockpiling remains to be resolved.

Antivirals appear to have a narrow temporal window within which they are most likely to have clinically relevant effectiveness, thereby restricting their use to prophylaxis (pre-exposure and postexposure) and possibly, early treatment.19 Antivirals could also play a key role in providing bridging protection before vaccine response or availability of vaccine. The time window for protection after challenge49 in monkeys is shorter with NiV-B than with NiV-M,27 although this finding is yet to be validated in humans.

Immunomodulators could be used in combination with pathogen-directed antivirals85 in the later phases of infection when immunopathology is thought to dominate,19 although no data on such combinations are available yet. Rintatolimod is the only host-directed agent with in-vivo efficacy data specifically for henipavirus infection that has been identified in this Review, and rintatolimod provides only partial protection after low-dose NiV-M challenge in hamsters.46

Overall, the paucity of drug candidates and clinical evidence underscores the challenges of clinical development of therapeutics for rare but high-threat infections with the potential to cause a pandemic.

Despite further Nipah outbreaks, cases under the current epidemiological situation are insufficient to obtain phase 3 RCT efficacy data necessary for approval86 or to attract substantial commercial investment. Alternative approaches similar to the regionally driven end-to-end West African Lassa fever Consortium87 framework are needed to bridge the gap.19

The requirement for BSL-4 precautions for preclinical studies of Nipah or Hendra virus also restricts the studies to a small number of specialist facilities. Establishing BSL-4 facilities in a larger number of locations, especially in henipavirus-endemic areas, could increase global capacity and allow endemic countries greater freedom to prioritise studies according to local needs. However, the high operational costs, small pool of specialist staff, and shortage of non-human primates following COVID-19 remain challenges for BSL-4 studies.

In the absence of RCTs in outbreak settings, evaluation of the efficacy of therapeutics for Nipah and Hendra virus diseases is mostly reliant on controlled animal challenge studies. The variable agreement between the results for in-vitro and in-vivo efficacy for most of the small molecules identified in this Review emphasises the importance of animal efficacy data for clinical prioritisation.

The US Food and Drug Administration allows for approval of drugs for conditions that threaten global human health security under the Animal Rule,88 when field trials are not possible, provided the following four criteria are met: sufficient understanding of the pathophysiology of the condition and the mechanism of its reduction by the product; efficacy shown in at least two animal species or one species that is a well characterised model for predicting the product’s response in humans; an animal study endpoint that is clearly related to the desired outcome in humans, typically reduction in mortality or major morbidity; and pharmacokinetic and pharmacodynamic data from animals and humans supporting selection of an effective dose in humans. The anti-infective agents approved under the Animal Rule include raxibacumab and obitoxaximab for anthrax; antibiotics such as ciprofloxacin for plague; and tecovirimat and brincidofovir for smallpox.

The European Medicines Agency has a similar Exceptional Circumstances89 mechanism for granting marketing authorisation to drugs whenever collection of data on their comprehensive efficacy and safety under typical conditions of use is not possible.

The clinical and pathological features and the strengths and limitations of the major animal models of Nipah virus disease (African green monkeys,90 ferrets,91 and Syrian golden hamsters92) have been reviewed93 by the Coalition for Epidemic Preparedness Innovations (CEPI). African green monkeys are closest in physiology to humans but have less consistent neurological signs compared with hamsters and ferrets.93 CEPI is also improving these models, particularly by standardising the virus challenge stock.93 Supporting access to NiV-B strains and standardisation of the dose and route of the challenge for each model could aid comparability across studies. For challenge studies involving therapeutics, having uninfected controls to assess drug toxicity thresholds (for candidates with a narrow or unknown therapeutic dose window) and pharmacokinetic–pharmacodynamic sampling to identify in-vivo EC50 values and concentration–efficacy relationships would be crucial for dose optimisation to de-risk human trials. Reporting guidelines can also play an important role in supporting greater transparency and consistency in publications as well as scientific reproducibility.

Human data remain essential for the evaluation of drug safety.88 Before deployment in outbreak settings, phase 1 first-in-human safety data need to be collected for new therapeutic agents, and the existing experience from repurposed agents has to be critically appraised for any potential exacerbation of adverse effects by the pathophysiology of Nipah or Hendra virus infection.

In an outbreak, new therapeutic agents should be evaluated in well designed phase 2 clinical trials integrated into and sustained in health systems94 using preapproved standardised protocols that maximise statistical and operational efficiency in the assessment of internationally agreed-upon core outcome measures.95 Wherever possible, drug concentrations should be measured at the same timepoints as efficacy and safety outcomes to characterise and quantify pharmacokinetic–pharmacodynamic relationships, including those at different stages of disease.

When RCTs are not possible, observational studies using enhanced clinical characterisation protocols96 that incorporate the same outcome measures could provide observational data of higher quality.97 The long-term neurological sequelae of Nipah encephalitis8,98,99 also merit more systematic characterisation and potential inclusion as outcomes.

Outbreaks of high-threat infections invoke the ethical duty100,101 to conduct inclusive research with speed and rigour. Community and stakeholder engagement,101 including that for design and interventions in trials, are key to support genuine informed consent and to maintain trust in the scientific process.102

Selection, optimisation, and stockpiling of potential therapeutics and their appropriate dosing regimens on the basis of all available clinical and preclinical evidence in advance of any outbreak are crucial. The continuous iterative process should be guided by disease-specific and, wherever appropriate, product-specific target product profiles103 (comprising indication, safety, efficacy, route, stability, and affordability characteristics) developed through consensus among all relevant stakeholders, including regulators, end-users, and communities. Systems pharmacology and statistical, mathematical, and economic modelling are powerful tools to support decision making as they provide a formal framework for integration of typically sparse data from multiple study types, species, and diseases and also inform the design efficiency of phase 1 and phase 2 RCTs.

Conclusions

At present, sufficient evidence is available to trial the monoclonal antibodies 1F5 and m102.4 along with the small molecule remdesivir (alone or in combination) for prophylaxis and early treatment of Nipah virus disease. In addition to well designed RCTs, in-vivo pharmacokinetic–pharmacodynamic studies are needed to support drug selection and dose optimisation for all high-threat infections.

Declaration of interests

We declare no competing interests.

Acknowledgments

XHSC is a UK National Institute of Health and Care Research (NIHR) Academic Clinical Lecturer in Infectious Diseases at the University of Oxford. MZH is a Moh Family Foundation Fellow at the Pandemic Sciences Institute, University of Oxford. JD is supported by the Moh Family Foundation, the NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, and the UK Public Health Rapid Support Team Research Programme (grant number IS-RRT-1015-001). JT is supported by the Wellcome Trust (grant number 220211). PWH is the Moh Family Foundation Professor of Emerging Infections and Global Health at the Pandemic Sciences Institute, University of Oxford. PWH and PLO are supported by the UK Foreign and Commonwealth, and Development Office, the Wellcome Trust (grant number 215091/Z/18/Z), and the Bill & Melinda Gates Foundation (grant number OPP1209135). The authors thank all members of the Pandemic Sciences Institute Henipavirus Programme and delegates of the International Pandemic Sciences Conference 2023 and the Nipah Virus Twenty-fifth Anniversary Symposium, both in Oxford, for the illuminating discussions. The funders of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report.

Contributors

XHSC, JD, PWH, and PLO conceptualised this Review. XHSC and EH wrote the study protocol and designed the search strategy. EH conducted the literature search. XHSC, ILH, and SL piloted this Review and screened studies. XHSC, ILH, BJKC, MZH, JTak, LMJ, and TPH extracted and tabulated the data. MZH and BJKC conducted the Risk of Bias assessment. MZH and XHSC generated the figures. XHSC, ILH, and BJKC wrote the initial draft. All authors critically reviewed, edited, and approved the final version of the manuscript.

Supplementary Material

Supplementary appendix
mmc1.pdf (1.9MB, pdf)

References

  • 1.Goh KJ, Tan CT, Chew NK, et al. Clinical features of Nipah virus encephalitis among pig farmers in Malaysia. N Engl J Med. 2000;342:1229–1235. doi: 10.1056/NEJM200004273421701. [DOI] [PubMed] [Google Scholar]
  • 2.Luby SP, Rahman M, Hossain MJ, et al. Foodborne transmission of Nipah virus, Bangladesh. Emerg Infect Dis. 2006;12:1888–1894. doi: 10.3201/eid1212.060732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nikolay B, Salje H, Hossain MJ, et al. Transmission of Nipah virus—14 years of investigations in Bangladesh. N Engl J Med. 2019;380:1804–1814. doi: 10.1056/NEJMoa1805376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chowdhury S, Khan SU, Crameri G, et al. Serological evidence of henipavirus exposure in cattle, goats and pigs in Bangladesh. PLoS Negl Trop Dis. 2014;8 doi: 10.1371/journal.pntd.0003302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Islam A, Cannon DL, Rahman MZ, et al. Nipah virus exposure in domestic and peridomestic animals living in human outbreak sites, Bangladesh, 2013–2015. Emerg Infect Dis. 2023;29:393–396. doi: 10.3201/eid2902.221379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hossain MJ, Gurley ES, Montgomery JM, et al. Clinical presentation of Nipah virus infection in Bangladesh. Clin Infect Dis. 2008;46:977–984. doi: 10.1086/529147. [DOI] [PubMed] [Google Scholar]
  • 7.Shete AM, Radhakrishnan C, Pardeshi PG, et al. Antibody response in symptomatic & asymptomatic Nipah virus cases from Kerala, India. Indian J Med Res. 2021;154:533–535. doi: 10.4103/ijmr.IJMR_4388_20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sejvar JJ, Hossain J, Saha SK, et al. Long-term neurological and functional outcome in Nipah virus infection. Ann Neurol. 2007;62:235–242. doi: 10.1002/ana.21178. [DOI] [PubMed] [Google Scholar]
  • 9.Broder CC, Wong KT. In: Neurotropic viral infections: volume 1: neurotropic RNA viruses. Reiss CS, editor. Springer; 2016. Henipaviruses; pp. 45–83. [Google Scholar]
  • 10.Chua KB, Goh KJ, Wong KT, et al. Fatal encephalitis due to Nipah virus among pig-farmers in Malaysia. Lancet. 1999;354:1257–1259. doi: 10.1016/S0140-6736(99)04299-3. [DOI] [PubMed] [Google Scholar]
  • 11.Paton NI, Leo YS, Zaki SR, et al. Outbreak of Nipah-virus infection among abattoir workers in Singapore. Lancet. 1999;354:1253–1256. doi: 10.1016/S0140-6736(99)04379-2. [DOI] [PubMed] [Google Scholar]
  • 12.Chua KB. Epidemiology, surveillance and control of Nipah virus infections in Malaysia. Malays J Pathol. 2010;32:69–73. [PubMed] [Google Scholar]
  • 13.Suit-B Y, Hassan L, Krauss SE, et al. Mental model of Malaysian pig farmers in implementing disease prevention and control practices. Front Vet Sci. 2021;8 doi: 10.3389/fvets.2021.695702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ching PK, de los Reyes VC, Sucaldito MN, et al. Outbreak of henipavirus infection, Philippines, 2014. Emerg Infect Dis. 2015;21:328–331. doi: 10.3201/eid2102.141433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chadha MS, Comer JA, Lowe L, et al. Nipah virus-associated encephalitis outbreak, Siliguri, India. Emerg Infect Dis. 2006;12:235–240. doi: 10.3201/eid1202.051247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Arunkumar G, Chandni R, Mourya DT, et al. Outbreak investigation of Nipah virus disease in Kerala, India, 2018. J Infect Dis. 2019;219:1867–1878. doi: 10.1093/infdis/jiy612. [DOI] [PubMed] [Google Scholar]
  • 17.Hassan MZ, Sazzad HMS, Luby SP, et al. Nipah virus contamination of hospital surfaces during outbreaks, Bangladesh, 2013–2014. Emerg Infect Dis. 2018;24:15–21. doi: 10.3201/eid2401.161758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.WHO Disease outbreak news: Nipah virus infection – India. Oct 3, 2023. https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON490
  • 19.Hassan MZ, Shirin T, Satter SM, et al. Nipah virus disease: what can we do to improve patient care? Lancet Infect Dis. 2024;24:e463–e471. doi: 10.1016/S1473-3099(23)00707-7. [DOI] [PubMed] [Google Scholar]
  • 20.Gómez Román R, Tornieporth N, Cherian NG, et al. Medical countermeasures against henipaviruses: a review and public health perspective. Lancet Infect Dis. 2022;22:e13–e27. doi: 10.1016/S1473-3099(21)00400-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mehand MS, Al-Shorbaji F, Millett P, Murgue B. The WHO R&D Blueprint: 2018 review of emerging infectious diseases requiring urgent research and development efforts. Antiviral Res. 2018;159:63–67. doi: 10.1016/j.antiviral.2018.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sterne JAC, Savović J, Page MJ, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366 doi: 10.1136/bmj.l4898. [DOI] [PubMed] [Google Scholar]
  • 23.Sterne JA, Hernán MA, Reeves BC, et al. Robins-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355 doi: 10.1136/bmj.i4919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hooijmans CR, Rovers MM, de Vries RBM, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE's risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14:43. doi: 10.1186/1471-2288-14-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sahay RR, Yadav PD, Gupta N, et al. Experiential learnings from the Nipah virus outbreaks in Kerala towards containment of infectious public health emergencies in India. Epidemiol Infect. 2020;148:e90. doi: 10.1017/S0950268820000825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Playford EG, Munro T, Mahler SM, et al. Safety, tolerability, pharmacokinetics, and immunogenicity of a human monoclonal antibody targeting the g glycoprotein of henipaviruses in healthy adults: a first-in-human, randomised, controlled, phase 1 study. Lancet Infect Dis. 2020;20:445–454. doi: 10.1016/S1473-3099(19)30634-6. [DOI] [PubMed] [Google Scholar]
  • 27.Mire CE, Satterfield BA, Geisbert JB, et al. Pathogenic differences between Nipah virus Bangladesh and Malaysia strains in primates: implications for antibody therapy. Sci Rep. 2016;6 doi: 10.1038/srep30916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Geisbert TW, Mire CE, Geisbert JB, et al. Therapeutic treatment of Nipah virus infection in nonhuman primates with a neutralizing human monoclonal antibody. Sci Transl Med. 2014;6 doi: 10.1126/scitranslmed.3008929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bossart KN, Geisbert TW, Feldmann H, et al. A neutralizing human monoclonal antibody protects African green monkeys from Hendra virus challenge. Sci Transl Med. 2011;3 doi: 10.1126/scitranslmed.3002901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bossart KN, Zhu Z, Middleton D, et al. A neutralizing human monoclonal antibody protects against lethal disease in a new ferret model of acute Nipah virus infection. PLoS Pathog. 2009;5 doi: 10.1371/journal.ppat.1000642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhu Z, Bossart KN, Bishop KA, et al. Exceptionally potent cross-reactive neutralization of Nipah and Hendra viruses by a human monoclonal antibody. J Infect Dis. 2008;197:846–853. doi: 10.1086/528801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zeitlin L, Cross RW, Woolsey C, et al. Therapeutic administration of a cross-reactive mAb targeting the fusion glycoprotein of Nipah virus protects nonhuman primates. Sci Transl Med. 2024;16 doi: 10.1126/scitranslmed.adl2055. [DOI] [PubMed] [Google Scholar]
  • 33.Mire CE, Chan Y-P, Borisevich V, et al. A cross-reactive humanized monoclonal antibody targeting fusion glycoprotein function protects ferrets against lethal Nipah virus and Hendra virus infection. J Infect Dis. 2020;221(suppl 4):S471–S479. doi: 10.1093/infdis/jiz515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen L, Sun M, Zhang H, et al. Potent human neutralizing antibodies against Nipah virus derived from two ancestral antibody heavy chains. Nat Commun. 2024;15:2987. doi: 10.1038/s41467-024-47213-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Doyle MP, Kose N, Borisevich V, et al. Cooperativity mediated by rationally selected combinations of human monoclonal antibodies targeting the henipavirus receptor binding protein. Cell Rep. 2021;36 doi: 10.1016/j.celrep.2021.109628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Dong J, Cross RW, Doyle MP, et al. Potent henipavirus neutralization by antibodies recognizing diverse sites on Hendra and Nipah virus receptor binding. Cell. 2020;183 doi: 10.1016/j.cell.2020.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Guillaume V, Contamin H, Loth P, et al. Antibody prophylaxis and therapy against Nipah virus infection in hamsters. J Virol. 2006;80:1972–1978. doi: 10.1128/JVI.80.4.1972-1978.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Guillaume V, Wong KT, Looi RY, et al. Acute Hendra virus infection: analysis of the pathogenesis and passive antibody protection in the hamster model. Virology. 2009;387:459–465. doi: 10.1016/j.virol.2009.03.001. [DOI] [PubMed] [Google Scholar]
  • 39.Warrier A. A single case outbreak of Nipah Encephalitis from India in May–June 2019. Int J Infect Dis. 2020;101(suppl 1):247. [Google Scholar]
  • 40.Chandni R, Renjith TP, Fazal A, et al. Clinical manifestations of Nipah virus-infected patients who presented to the emergency department during an outbreak in Kerala state in India, May 2018. Clin Infect Dis. 2020;71:152–157. doi: 10.1093/cid/ciz789. [DOI] [PubMed] [Google Scholar]
  • 41.Banerjee S, Niyas VKM, Soneja M, et al. First experience of ribavirin postexposure prophylaxis for Nipah virus, tried during the 2018 outbreak in Kerala, India. J Infect. 2019;78:491–503. doi: 10.1016/j.jinf.2019.03.005. [DOI] [PubMed] [Google Scholar]
  • 42.Kumar ASA, Sohanlal T, Prasad G, Gupta M, Gopal A. Single-center experience and lessons learnt from management of Nipah virus outbreak in India. Open Forum Infect Dis. 2019;6(suppl 2):S659–S660. [Google Scholar]
  • 43.Playford EG, McCall B, Smith G, et al. Human Hendra virus encephalitis associated with equine outbreak, Australia, 2008. Emerg Infect Dis. 2010;16:219–223. doi: 10.3201/eid1602.090552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chong HT, Kamarulzaman A, Tan CT, et al. Treatment of acute Nipah encephalitis with ribavirin. Ann Neurol. 2001;49:810–813. doi: 10.1002/ana.1062. [DOI] [PubMed] [Google Scholar]
  • 45.Rockx B, Bossart KN, Feldmann F, et al. A novel model of lethal Hendra virus infection in African green monkeys and the effectiveness of ribavirin treatment. J Virol. 2010;84:9831–9839. doi: 10.1128/JVI.01163-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Georges-Courbot MC, Contamin H, Faure C, et al. Poly(I)-poly(C12U) but not ribavirin prevents death in a hamster model of Nipah virus infection. Antimicrob Agents Chemother. 2006;50:1768–1772. doi: 10.1128/AAC.50.5.1768-1772.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Freiberg AN, Worthy MN, Lee B, Holbrook MR. Combined chloroquine and ribavirin treatment does not prevent death in a hamster model of Nipah and Hendra virus infection. J Gen Virol. 2010;91:765–772. doi: 10.1099/vir.0.017269-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Pallister J, Middleton D, Crameri G, et al. Chloroquine administration does not prevent Nipah virus infection and disease in ferrets. J Virol. 2009;83:11979–11982. doi: 10.1128/JVI.01847-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.de Wit E, Williamson BN, Feldmann F, et al. Late remdesivir treatment initiation partially protects African green monkeys from lethal Nipah virus infection. Antiviral Res. 2023;216 doi: 10.1016/j.antiviral.2023.105658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lo MK, Feldmann F, Gary JM, et al. Remdesivir (GS-5734) protects African green monkeys from Nipah virus challenge. Sci Transl Med. 2019;11 doi: 10.1126/scitranslmed.aau9242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jordan R, Hogg A, Warren T, et al. Broad-spectrum investigational agent GS-5734 for the treatment of Ebola, MERS coronavirus and other pathogenic viral infections with high outbreak potential. Open Forum Infect Dis. 2017;4(suppl 1):S737. [Google Scholar]
  • 52.Dawes BE, Kalveram B, Ikegami T, et al. Favipiravir (T-705) protects against Nipah virus infection in the hamster model. Sci Rep. 2018;8:7604. doi: 10.1038/s41598-018-25780-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lo MK, Spengler JR, Krumpe LRH, et al. Griffithsin inhibits Nipah virus entry and fusion and can protect Syrian golden hamsters from lethal Nipah virus challenge. J Infect Dis. 2020;221(suppl 4):S480–S492. doi: 10.1093/infdis/jiz630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mathieu C, Dhondt KP, Châlons M, et al. Heparan sulfate-dependent enhancement of henipavirus infection. mBio. 2015;6 doi: 10.1128/mBio.02427-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mathieu C, Porotto M, Figueira TN, Horvat B, Moscona A. Fusion inhibitory lipopeptides engineered for prophylaxis of Nipah virus in primates. J Infect Dis. 2018;218:218–227. doi: 10.1093/infdis/jiy152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mathieu C, Augusto MT, Niewiesk S, et al. Broad spectrum antiviral activity for paramyxoviruses is modulated by biophysical properties of fusion inhibitory peptides. Sci Rep. 2017;7 doi: 10.1038/srep43610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Porotto M, Rockx B, Yokoyama CC, et al. Inhibition of Nipah virus Infection in vivo: targeting an early stage of paramyxovirus fusion activation during viral entry. PLoS Pathog. 2010;6 doi: 10.1371/journal.ppat.1001168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Welch SR, Spengler JR, Harmon JR, et al. Defective interfering viral particle treatment reduces clinical signs and protects hamsters from lethal Nipah virus disease. mBio. 2022;13 doi: 10.1128/mbio.03294-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wright PJ, Crameri G, Eaton BT. RNA synthesis during infection by Hendra virus: an examination by quantitative real-time PCR of RNA accumulation, the effect of ribavirin and the attenuation of transcription. Arch Virol. 2005;150:521–532. doi: 10.1007/s00705-004-0417-5. [DOI] [PubMed] [Google Scholar]
  • 60.Porotto M, Orefice G, Yokoyama CC, et al. Simulating henipavirus multicycle replication in a screening assay leads to identification of a promising candidate for therapy. J Virol. 2009;83:5148–5155. doi: 10.1128/JVI.00164-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lo MK, Jordan R, Arvey A, et al. GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and Paramyxoviruses. Sci Rep. 2017;7 doi: 10.1038/srep43395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lo MK, Shrivastava-Ranjan P, Chatterjee P, et al. Broad-spectrum in vitro antiviral activity of ODBG-P-RVn: an orally-available, lipid-modified monophosphate prodrug of remdesivir parent nucleoside (GS-441524) Microbiol Spectr. 2021;9 doi: 10.1128/Spectrum.01537-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Welch SR, Tilston NL, Lo MK, et al. Inhibition of Nipah virus by defective interfering particles. J Infect Dis. 2020;221(suppl 4):S460–S470. doi: 10.1093/infdis/jiz564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wolf MC, Freiberg AN, Zhang T, et al. A broad-spectrum antiviral targeting entry of enveloped viruses. Proc Natl Acad Sci U S A. 2010;107:3157–3162. doi: 10.1073/pnas.0909587107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.British National Formulary Remdesivir. 2024. https://bnf.nice.org.uk/drugs/remdesivir/
  • 66.Pharmaceuticals and Medical Devices Agency Avigan (favipiravir) review report. March 4, 2014. https://www.pmda.go.jp/files/000210319.pdf Tokyo, Japan.
  • 67.Pilkington V, Pepperrell T, Hill A. A review of the safety of favipiravir - a potential treatment in the COVID-19 pandemic? J Virus Erad. 2020;6:45–51. doi: 10.1016/S2055-6640(20)30016-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Irie K, Nakagawa A, Fujita H, et al. Population pharmacokinetics of favipiravir in patients with COVID-19. CPT Pharmacometrics Syst Pharmacol. 2021;10:1161–1170. doi: 10.1002/psp4.12685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.White NJ, Watson JA, Hoglund RM, Chan XHS, Cheah PY, Tarning J. COVID-19 prevention and treatment: a critical analysis of chloroquine and hydroxychloroquine clinical pharmacology. PLoS Med. 2020;17 doi: 10.1371/journal.pmed.1003252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Whitmer SLM, Lo MK, Sazzad HMS, et al. Inference of Nipah virus evolution, 1999—2015. Virus Evol. 2021;7 doi: 10.1093/ve/veaa062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chen C, Fang J, Chen S, et al. The efficacy and safety of remdesivir alone and in combination with other drugs for the treatment of COVID-19: a systematic review and meta-analysis. BMC Infect Dis. 2023;23:672. doi: 10.1186/s12879-023-08525-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nguyen THT, Guedj J, Anglaret X, et al. Favipiravir pharmacokinetics in Ebola-Infected patients of the JIKI trial reveals concentrations lower than targeted. PLoS Negl Trop Dis. 2017;11 doi: 10.1371/journal.pntd.0005389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wang Y, Zhong W, Salam A, et al. Phase 2a, open-label, dose-escalating, multi-center pharmacokinetic study of favipiravir (T-705) in combination with oseltamivir in patients with severe influenza. EBioMedicine. 2020;62 doi: 10.1016/j.ebiom.2020.103125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Madelain V, Guedj J, Mentré F, et al. Favipiravir pharmacokinetics in nonhuman primates and insights for future efficacy studies of hemorrhagic fever viruses. Antimicrob Agents Chemother. 2017;61 doi: 10.1128/AAC.01305-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bixler SL, Bocan TM, Wells J, et al. Efficacy of favipiravir (T-705) in nonhuman primates infected with Ebola virus or Marburg virus. Antiviral Res. 2018;151:97–104. doi: 10.1016/j.antiviral.2017.12.021. [DOI] [PubMed] [Google Scholar]
  • 76.Batool S, Vuthaluru K, Hassan A, et al. Efficacy and safety of favipiravir in treating COVID-19 patients: a meta-analysis of randomized control trials. Cureus. 2023;15 doi: 10.7759/cureus.33676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cheng HY, French CE, Salam AP, et al. Lack of evidence for ribavirin treatment of Lassa fever in systematic review of published and unpublished studies. Emerg Infect Dis. 2022;28:1559–1568. doi: 10.3201/eid2808.211787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Salam AP, Duvignaud A, Jaspard M, et al. Ribavirin for treating Lassa fever: a systematic review of pre-clinical studies and implications for human dosing. PLoS Negl Trop Dis. 2022;16 doi: 10.1371/journal.pntd.0010289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Central Team, Ministry of Health and Family Welfare, Government of India Clinical Management Protocol for Nipah Virus Disease. 2018. https://cghealth.nic.in/cghealth17/Information/content/NipahVirus/Treatment.pdf
  • 80.Government of Kerala Department of Health and Family Welfare Nipah virus infection guidelines for surveillance, diagnosis, treatment, prevention and control. 2021. https://dhs.kerala.gov.in/wp-content/uploads/2021/09/Nipah-Guidelines-9-04-21-2-1.pdf
  • 81.Directorate General of Health Services, Ministry of Health & Family Welfare, Government of the People's Republic of Bangladesh. National Guideline for Management Prevention and Control of Nipah Virus Infection including Encephalitis. 2018. https://www.iedcr.org/pdf/files/nipah/National_Nipah.pdf
  • 82.Haeusler IL, Chan XHS, Guérin PJ, White NJ. The arrhythmogenic cardiotoxicity of the quinoline and structurally related antimalarial drugs: a systematic review. BMC Med. 2018;16:200. doi: 10.1186/s12916-018-1188-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Drell W, Welch AD. Azaribine-homocystinemia-thrombosis in historical perspective. Pharmacol Ther. 1989;41:195–206. doi: 10.1016/0163-7258(89)90106-x. [DOI] [PubMed] [Google Scholar]
  • 84.Oey A, McClure M, Symons JA, et al. Lumicitabine, an orally administered nucleoside analog, in infants hospitalized with respiratory syncytial virus (RSV) infection: safety, efficacy, and pharmacokinetic results. PLoS One. 2023;18 doi: 10.1371/journal.pone.0288271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host-directed therapies for bacterial and viral infections. Nat Rev Drug Discov. 2018;17:35–56. doi: 10.1038/nrd.2017.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nikolay B, Ribeiro Dos Santos G, Lipsitch M, et al. Assessing the feasibility of Nipah vaccine efficacy trials based on previous outbreaks in Bangladesh. Vaccine. 2021;39:5600–5606. doi: 10.1016/j.vaccine.2021.08.027. [DOI] [PubMed] [Google Scholar]
  • 87.International Severe Acute Respiratory and emerging Infection Consortium West Africa Lassa fever Consortium (WALC) 2023. https://isaric.org/research/lassa-fever-resources/walc/
  • 88.US Food and Drug Administration Animal rule approvals. 2002. https://www.fda.gov/drugs/nda-and-bla-approvals/animal-rule-approvals
  • 89.European Medicines Agency Exceptional circumstances. 2024. https://www.ema.europa.eu/en/glossary/exceptional-circumstances
  • 90.Mire CE, Satterfield BA, Geisbert TW. Nonhuman primate models for Nipah and Hendra virus countermeasure evaluation. Methods Mol Biol. 2023;2682:159–173. doi: 10.1007/978-1-0716-3283-3_12. [DOI] [PubMed] [Google Scholar]
  • 91.Rockx B, Mire CE. Ferret models for henipavirus infection. Methods Mol Biol. 2023;2682:205–217. doi: 10.1007/978-1-0716-3283-3_15. [DOI] [PubMed] [Google Scholar]
  • 92.Juelich T, Smith J, Freiberg AN. Syrian golden hamster model for Nipah virus infection. Methods Mol Biol. 2023;2682:219–229. doi: 10.1007/978-1-0716-3283-3_16. [DOI] [PubMed] [Google Scholar]
  • 93.Centre for Epidemic Preparedness Innovations Nipah virus assays and animal models for vaccine development. Jan 9, 2021. https://media.tghn.org/medialibrary/2021/02/Nipah_Virus_Assays_and_Animal_Models_for_Vaccine_Development_final.pdf
  • 94.Moorthy V, Abubakar I, Qadri F, et al. The future of the global clinical trial ecosystem: a vision from the first WHO Global Clinical Trials Forum. Lancet. 2024;403:124–126. doi: 10.1016/S0140-6736(23)02798-8. [DOI] [PubMed] [Google Scholar]
  • 95.Williamson PR, Altman DG, Blazeby JM, et al. Developing core outcome sets for clinical trials: issues to consider. Trials. 2012;13:132. doi: 10.1186/1745-6215-13-132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.ISARIC Clinical Characterisation Protocol Investigators Group ISARIC/WHO clinical characterisation protocol (CCP) 2023. https://isaric.net/ccp/
  • 97.Dunning JW, Merson L, Rohde GGU, et al. Open source clinical science for emerging infections. Lancet Infect Dis. 2014;14:08–09. doi: 10.1016/S1473-3099(13)70327-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lee KE, Umapathi T, Tan CB, et al. The neurological manifestations of Nipah virus encephalitis, a novel paramyxovirus. Ann Neurol. 1999;46:428–432. [PubMed] [Google Scholar]
  • 99.Tan CT, Goh KJ, Wong KT, et al. Relapsed and late-onset Nipah encephalitis. Ann Neurol. 2002;51:703–708. doi: 10.1002/ana.10212. [DOI] [PubMed] [Google Scholar]
  • 100.Johnson T, Jamrozik E, Hurst T, Cheah PY, Parker MJ. Ethical issues in Nipah virus control and research: addressing a neglected disease. J Med Ethics. 2024;50:612–617. doi: 10.1136/jme-2023-109469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.WHO Emergency use of unproven clinical interventions outside clinical trials: ethical considerations. Apr 12, 2022. https://www.who.int/publications/i/item/9789240041745
  • 102.Meskell P, Biesty LM, Dowling M, et al. Factors that impact on recruitment to vaccine trials in the context of a pandemic or epidemic: a qualitative evidence synthesis. Cochrane Database Syst Rev. 2023;9:MR000065. doi: 10.1002/14651858.MR000065.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.WHO Target product profiles. 2023. https://www.who.int/observatories/global-observatory-on-health-research-and-development/analyses-and-syntheses/target-product-profile/who-target-product-profiles

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary appendix
mmc1.pdf (1.9MB, pdf)

RESOURCES