Nipah Virus Therapeutics: A Systematic Review to Support Prioritisation for Clinical Trials =========================================================================================== * Xin Hui S Chan * Ilsa L Haeusler * Bennett J K Choy * Md Zakiul Hassan * Junko Takata * Tara P Hurst * Luke M Jones * Shanghavie Loganathan * Elinor Harriss * Jake Dunning * Joel Tarning * Miles W Carroll * Peter W Horby * Piero L Olliaro ## Abstract Nipah virus disease is a bat-borne zoonosis with person-to-person transmission, a case fatality rate of 38-75%, and recognised pandemic potential. The first reported outbreak occurred in Malaysia and Singapore in 1998, since followed by multiple outbreaks in Bangladesh and India. No therapeutics or vaccines have been licensed to date, and only few candidates are in development. This systematic review aimed to assess the evidence for the safety and efficacy of therapeutic options (monoclonal antibodies and small molecules) for Nipah virus and other henipaviral diseases in order to support candidate prioritisation for further evaluation in clinical trials. At present, there is sufficient evidence to trial only m102.4 and remdesivir (singly and/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 to optimise selection and dosing of therapeutic candidates in animal challenge and natural human infection are needed. **Evidence before this study** Nipah virus infection is a bat-borne zoonosis with person-to-person transmission, a case fatality rate of 38-75%, and recognised pandemic potential. No therapeutics or vaccines have been licensed to date, and only few candidates are in development. We conducted this systematic review to assess the evidence for the safety and efficacy of therapeutic options (monoclonal antibodies and small molecules) for Nipah virus and other henipaviral diseases to support candidate prioritisation for further evaluation in clinical trials. We searched bibliographic databases 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 title, abstract, and subject heading keywords, with synonyms and variant spellings as additional search terms. We searched trial registries 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. We searched the Trip database and WHO website for guidelines and reports. All searches were conducted on 30 May 2022. We did not apply language or publication date limits. Studies were included if they contained primary data on the safety and/or efficacy of monoclonal antibodies (*in vivo)* or small molecules (*in vivo* or *in vitro*) for the treatment and/or prophylaxis of Nipah, Hendra, and related *Henipaviridae*. Almost all had critical or high risk of bias. **Added value of this study** This is the most detailed systematic review and analysis of the Nipah virus therapeutics landscape to date, including all available *in vivo* and related *in vitro* data on the safety, efficacy, and pharmaco-kinetics of monoclonal antibodies and small molecules with the specific aim of supporting prioritisation for clinical trials. We also present a roadmap for how *in vivo* development of Nipah therapeutics could be strengthened to achieve greater equity, efficiency, and effectiveness. **Implications of the available evidence** At present, there is sufficient evidence to trial only m102.4 and remdesivir for prophylaxis and early treatment of Nipah virus infection. Well-designed clinical efficacy trials as well as *in vivo* pharmacokinetic-pharmacodynamic studies to optimise selection and dosing of therapeutic candidates in animal challenge and natural human infection are needed. ## Introduction Nipah virus disease is a zoonotic infection acquired through contact with or ingestion of contaminated body fluids of infected mammals1–3. Pteropid fruit bats (flying foxes) are the primary reservoir of Nipah virus. Secondary hosts include domestic animals4,5 (pigs, horses, cows) and humans. There is person-to-person transmission. Its clinical presentation ranges from asymptomatic6 to an acute respiratory syndrome and fatal encephalitis1,7. After an incubation period of four to 14 days, fever, headache, and myalgia may be followed by shortness of breath and cough or confusion and seizures which can rapidly progress to coma within 24 to 48 hours1,7. Disease occurs in all age groups1,7. The case fatality ratio (CFR) is estimated to be between 38 to 75% and debilitating long-term neurological complications, such as paralysis, are common in Nipah survivors1,8. Nipah virus is part of the genus *Henipavirus* along with Hendra virus which also causes fatal encephalitis and respiratory disease in horses and humans. Both Nipah and Hendra viruses are biosafety level 4 (BSL-4) pathogens requiring 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 disease9. First identified in 1998, following an outbreak among pig farmers and abattoir workers in Malaysia10 and Singapore11, Nipah virus is named after the Malaysian village from which the virus was first isolated. 283 cases of encephalitis and 109 deaths were recorded, a CFR of 38.3%12. This outbreak was halted with mass culling of more than one million pigs and comprehensive modernisation of pig farming practice, including the separation of fruit tree plantations from pig farms13. There have been no further Nipah cases in Malaysia and Singapore in the subsequent 25 years, and only one further outbreak of the Nipah virus Malaysia (NiV-M) strain in the Philippines in 201414 related to horse slaughter and consumption. Outbreaks of the Nipah virus Bangladesh (NiV-B) strain have been reported in Bangladesh3 and India (West Bengal15, Kerala16), with healthcare workers15 and family17 caring for infected patients emerging as another important risk group. The highest mortality rates have been recorded in Bangladesh where outbreaks occur almost annually in the winter following harvesting and consumption of contaminated raw date palm sap2, a local delicacy. Since 2001, there have been 335 cases with 237 deaths in Bangladesh, a CFR of 70.7%3. The 2023 outbreak in Bangladesh was the largest since 2015 with 14 cases and 10 deaths. A second outbreak occurred less than 6 months later in Kerala, India with six cases and two fatalities18. Patient outcomes have not improved in 25 years since the first reported outbreaks due in part to the market failure typical of counter-measure development for a high-consequence pathogen19. There are no licensed vaccines or therapeutics for Nipah virus infection, and only a few candidates are currently in development20. In recognition of the need for vaccines and therapeutics, Nipah has been a priority disease in the World Health Organization (WHO) Research & Development Blueprint since 201821. Clinical evaluation is limited by the infeasibility of a controlled human infection model and the small number of patients in sporadic outbreaks. Assessment of efficacy is currently reliant on animal challenge studies conducted in BSL-4 facilities. We conducted this systematic review to assess the evidence for the safety and efficacy of therapeutic options (monoclonal antibodies [mAbs] and small molecules) for Nipah virus and other *Henipaviridae* causing human disease in order to support candidate prioritisation for further 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). ### Search Strategy We conducted an electronic literature search of the following bibliographic databases 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 title, abstract, and subject heading keywords, with synonyms and variant spellings as additional search terms. We searched the following trial registries 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. We searched the Trip database and WHO website for guidelines and reports. Full search strategies are in the appendix. All searches were conducted on 30 May 2022. We did not apply language or publication date limits. References were imported into EndNote and de-duplicated then screened against eligibility criteria. Reference lists of eligible records were checked for additional relevant studies. ### Eligibility Criteria Studies were included if they contained primary data on the safety and/or efficacy of mAbs (*in vivo)* or small molecules (*in vivo* or *in vitro*) for the treatment and/or prophylaxis of Nipah and/or Hendra infections. Studies on candidates without therapeutic applications (e.g. mAbs for diagnostics) or with only *in silico* data were excluded. ### Data Extraction We extracted data on the viruses studied, study characteristics (funder, year, location, design), intervention characteristics (drug, dose, route, administration timepoints), efficacy outcomes (all measures, all timepoints), and safety outcomes (all measures, all timepoints). Study investigators and experts were contacted for further information if necessary. ### Data Analysis The review pilot identified significant heterogeneity in study designs, outcome measures, and reporting. Quantitative data synthesis was deemed not possible. All available data were therefore prespecified to be summarised in tabular format by individual therapeutic candidate as a narrative synthesis prioritising clinical and animal studies. ### Quality Assessment Risk of bias assessment was undertaken for the study designs for which standardised tools exist: Risk of Bias 2 (RoB 222) for randomised clinical trials (RCTs), Risk of Bias in Non-randomized Studies of Interventions (ROBINS-I23) for non-randomised clinical studies, and Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE24) for animal studies. ### Review Team & Tools At least two independent reviewers performed screening (titles and abstracts, then full texts), agreed study eligibility, extracted data, and undertook risk of bias assessment using Covidence (Veritas Health Innovation Ltd, Melbourne, Australia). ### Role of the Funding Source Our funders had no role in study design, data collection, data analysis, data interpretation, or writing of the report. All authors have access to the data in the study and accept responsibility to submit for publication. ## Results ### Included Studies We identified 56 eligible studies (Figure 1): 12 of mAbs with clinical and/or animal data25–36 (Table 1 & Supplementary Table I), 25 of small molecules with clinical and/or animal data (Table 2 & Supplementary Tables II-III), and 19 of small molecules with *in vitro* data only (Supplementary Table IV). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/15/2024.03.11.24304091/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2024/03/15/2024.03.11.24304091/F1) Figure 1: PRISMA Flow Chart View this table: [Table 1:](http://medrxiv.org/content/early/2024/03/15/2024.03.11.24304091/T1) Table 1: Nipah and Hendra Virus Therapeutic Monoclonal Antibodies View this table: [Table 2:](http://medrxiv.org/content/early/2024/03/15/2024.03.11.24304091/T2) Table 2: Nipah and Hendra Virus Therapeutic Small Molecules (Clinical and Animal Studies) There was only one clinical trial, a first-in-human phase 1 study in healthy volunteers of m102.426, a mAb targeting the Hendra virus (HeV) envelope G glycoprotein, conducted in Australia. Of the eight reports of compassionate use for treatment or post-exposure prophylaxis during Hendra or Nipah virus outbreaks, seven were case series of fewer than 10 patients in Australia37, India (Kerala)25,38–41, and Singapore11. The remaining outbreak report was from the two centres where 194 of the 283 cases in the 1998 Malaysia outbreak were treated, the majority with ribavirin42. Of the 23 animal studies, there were seven studies in non-human primates (African green monkeys [AGMs])27–29,43–46, five in ferrets30–32,34,47, and 12 in Syrian golden hamsters33,35,36,46,48–55. All except one involved infectious challenge with Nipah and/or Hendra virus (Supplementary Table V). Nipah virus Malaysia (NiV-M) was the most common challenge strain used in 12 studies28,30,32,36,46–50,52,53,55, followed by six studies using Nipah virus Bangladesh (NiV-B)27,33,34,43,44,51 and five HeV29,32,35,45,49. All animal challenge studies reported death, and time of death, as outcome measures. The majority also reported clinical outcomes (all: signs and symptoms; AGMs only: radiological changes, blood test abnormalities) with day of onset, and a smaller majority reported pathology and virology (detection of RNA, antigen, or live virus by culture) at necropsy. A minority assessed correlation between drug concentrations and survival. ### Monoclonal Antibodies The 12 articles on mAbs reported data on six sets of antibodies from three research groups. Seven articles were on m102.4, an anti-HeV-G glycoprotein antibody, the most advanced candidate in clinical development (Table 1 & Supplementary Table I). m102.4 was the only Nipah drug candidate with clinical data from an RCT26 (phase 1) and with *in vivo* data from more than one animal species (AGMs27–29 and ferrets30,31) challenged with different henipaviruses (NiV-B27, NiV-M28,30, HeV29) (Table 1). Currently available data in humans support its safety. In a first-in-human dose-escalation randomised placebo-controlled trial of intravenous (IV) m102.4 (single doses of 1-20mg/kg + two doses of 20mg/kg 72 hours apart) in 40 healthy adult volunteers followed up for ∼4 months between 2015 and 2016, no serious adverse events were reported26. The frequency of adverse events, of which headache was the most common, were similar between the different treatment and placebo groups26. No anti-m102.4 antibodies were detected26. Prior to this trial, 14 individuals aged 8-59 years had received m102.4 as post-exposure prophylaxis on compassionate grounds for Hendra (n=13) and Nipah virus (n=1) infections in Australia and the USA respectively26. Of these, two individuals experienced infusion-related febrile reactions that were attributed to an early production process of the antibody26. There was also one outbreak report describing a single patient receiving m102.4 as post-exposure prophylaxis in Kerala, India, in 201825. The patient was reported to have recovered completely, but no further details were provided. The four animal challenge studies of m102.4 provide evidence of its efficacy in preventing death and severe disease in all treated animals when administered as a single dose in ferrets 10 hours after oronasal NiV-M inoculation30 (n=3) or in a two-dose regimen given 48 hours apart in monkeys starting within five days after intratracheal NiV-M28 (n=12) or HeV29 (n=12) challenge. In comparison, all control animals died within 8-10 days in these three studies. However, the treatment window for the two-dose regimen after NiV-B challenge in monkeys27 was shorter than that for NiV-M and HeV, with only the animals treated within three days (n=6) from inoculation surviving to the end of the study while the monkeys treated on days 5 and 7 (n=2) had similar outcomes to controls (n=2). In these four animal challenge studies, protection from disease was supported by the absence of pathological changes in treated animals on necropsy versus gross pathology in control animals, as well as the correlation of antibody levels with survival, including on day 3 post-challenge. Additionally, the developers of m102.4 are now also developing h5B.3, an anti-NiV-F glycoprotein antibody. When administered intraperitoneally (IP) to ferrets in a regimen of two 20mg/kg doses given 48 hours apart starting within three days of intranasal challenge with NiV-M (n=6) or HeV (n=3), h5B.3 protected all treated ferrets from severe disease32. Two articles33,34 from 2020 and 2021, from Vanderbilt University in the United States, describe two sets of anti-HeV receptor binding protein antibodies. HENV-26 (n=5) and HENV-32 (n=5) administered as two doses of 15mg/kg IP on days 3 and 5 post-intranasal NIV-B challenge each protected ferrets from death and severe disease compared to controls (n=3)34. 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)33. A further two articles35,36 from Institut National de la Santé et de la Recherche Médicale (INSERM), France dating from the late 2000s, describe two groups of anti-NiV-F and anti-NiV-G protein antibodies. These were detailed studies of protection, dose titration, and therapeutic window involving 12436 and 5435 hamsters respectively. However, there have been no further studies of these candidates in the subsequent two decades. ### Small Molecules The 25 articles on small molecules with *in vivo* (Table 2 & Supplementary Table II) and *in vitro* (Supplementary Table III) data described 10 potential therapeutics and one group of syndrome-directed broad-spectrum empirical antimicrobials. #### Ribavirin Ten articles were on ribavirin, a repurposed nucleoside analogue prodrug. These were six clinical case series for treatment37,39–42 and post-exposure prophylaxis37,38 for Nipah38–42 and Hendra37 outbreaks (Table 2), three animal challenge studies of AGMs (HeV45 only) and hamsters (both HeV49 and NiV-M49,50) (Table 2), plus three sets of *in vitro* experiments with HeV49,56 and NiV-M49,50 (Supplementary Table III). The small numbers of patients treated with ribavirin (n<10 in all except the Malaysia Nipah outbreak42) and pragmatic observational designs of case series precluded definitive statements about clinical efficacy. Dose regimens were different between the four publications37,38,40,42 reporting this information and were not reported in the remaining two39,41. All eight healthcare workers in the only post-exposure prophylaxis case series38 of ribavirin did not complete the prescribed course due to adverse effects: six of eight had symptoms (such as fatigue or headache) or transient laboratory abnormalities (increased bilirubin and/or decreased haemoglobin levels). In the three animal studies, when administered IP49,50 or subcutaneously45,50 (SC) 24 hours pre-challenge or within 12 hours post-challenge, ribavirin 50-150mg/kg/day delayed time to but did not prevent death or symptoms after NiV-M inoculation in hamsters49,50 (n=17) or HeV inoculation in AGMs45 (n=6), compared to untreated controls. Ribavirin 60mg/kg/day neither delayed nor prev-ented death in HeV-challenged hamsters (n=5)49. Systemic toxicity from high dose 200mg/kg/day of ribavirin IP was seen in both infected and uninfected (control) hamsters necessitating euthanasia49. *In vitro* experiments of ribavirin assessed viral replication through virus yield reduction56, cytopathic effect50, and dose response49 assays in NiV-M and HeV infected Vero50,56 and HeLa49 cells. Ribavirin doses used to achieve 58-fold56 or 100% reductions49,50 in viral yield were high (50-409μM) compared to half-maximal inhibitory concentrations49 (IC50) for NiV-M (4.18μM) and HeV (4.96μM). #### Chloroquine Three articles were on the widely used 4-aminoquinoline antimalarial chloroquine47,49,57: two animal challenge studies47,49 (Table 2) and two sets of *in vitro* experiments49,57 (Supplementary Table III). Ferrets administered chloroquine 25mg/kg/day IV 24 hours before (n=3) and 10 hours after (n=3) NiV-M challenge had disease courses identical to controls47. NiV-M and HeV inoculated hamsters treated six hours after challenge with chloroquine 50mg/kg IP on alternate days as monotherapy (n=5 per virus) or in combination with ribavirin 30mg/kg IP twice a day (n=5 per virus) died earlier or at the same time respectively as untreated controls49. Chloroquine 50mg/kg/day IP was also ineffective49. Higher doses of 100 and 150mg/kg/day of chloroquine IP were consistently lethal by day 2 in both infected and uninfected hamsters49. #### Remdesivir The three articles and one abstract on remdesivir, a nucleoside analogue, reported two AGM NiV-B challenge studies assessing IV remdesivir43,44 (Table 2) and *in vitro* data from multiple assays on both the IV58 and oral59 formulations (Supplementary Table III). Remdesivir 10mg/kg/day given from one day post-challenge protected all four AGMs from death, with mild transient respiratory signs in two and detectable viral RNA in brain tissue of one43. Controls all died after respiratory symptoms with detectable viraemia43. Reporter virus, cytopathic effect, and virus yield reduction assays for remdesivir IV (GS573458) and oral (GS44152459) as well as viral antigen reduction and minigenome assays for GS573458 were performed in cell types including HeLa and human small airway epithelial cells. Mean 50% maximal effective concentration (EC50) values were sub-micromolar for both and an order of magnitude lower for GS573458 (0.029-0.066μM) than GS44152459 (0.19-0.95μM). #### Favipiravir The single article on favipiravir48, a nucleoside analogue prodrug, contained data from an NiV-M hamster challenge study (Table 2) and *in vitro* assays (Supplementary Table III). Hamsters loaded with 600mg/kg SC immediately post-challenge then given a maintenance dose of 300mg/kg orally twice a day (n=5) or SC daily (n=5) for 13 days all survived without clinical signs or detectable pathology or viral antigen on necropsy while controls all died by day 5-6. The doses used in virus yield (100μM) and delayed treatment (250μM) assays to attain 100% and 10-fold (at one-hour post-infection) viral reductions, respectively, were high. EC50 values for HeV, NiV-B, and NiV-M were 11.7μM, 14.8μM, and 44.2μM respectively48. #### Others Six other groups of small molecules were studied, none of which provided complete protection from death at the doses used in animal challenge models. 6-azauridine, the nucleoside analogue metabolite of previously licensed azaribine, delayed mean time to death by ∼1 day, but did not prevent death, when given immediately prior to full-dose NiV-M challenge (350 x median lethal dose [LD50]) as a 175mg/kg/day continuous SC infusion for 14 days in hamsters50. Rintatolimod, a toll-like receptor 3 agonist, provided partial protection at 3mg/kg/day IP for 10 days, administered from 2 hours after low-dose (35 x LD50) NiV-M inoculation50 (Table 2 & Supplementary Table II). Periodate heparin, an experimental glycosaminoglycan competitive inhibitor of *trans*-infection, protected one of five NiV-M challenged hamsters at a dose of 10mg/kg/day SC for 12 days from day of infection52. Despite promising *in vitro* results, experimental cell entry inhibitors like the lectin griffithsin (oxidation resistant and trimeric monomer)51 and fusion inhibitory lipopeptides (cholesterol and tocopherol-based)46,54 administered at 10mg/kg/day intranasally (hamsters46,53,54) or intratracheally (AGMs46) prevented death in up to half of each group of NiV-B51 or NiV-M46,53 challenged animals (Table 2). Defective interfering virus particles given IP or intranasal also had partial efficacy in NiV-M challenged hamsters55 while virus yield reduction assays had up to an order of magnitude greater reduction in NiV-M than NiV-B infected Vero cells60 (Supplementary Table III). ### Risk of Bias Almost all the *in vivo* studies had critical (six of eight case series) or high (18 of 23 animal studies) risks of bias. Only three studies were assessed to have low risk of bias: the one RCT (of healthy volunteers)26, an outbreak report of a single case41, and an NiV-B challenge study in AGMs43. The remaining five25,30,32,52,61 studies had unclear risk of bias (Supplementary Figures I-V). ## Discussion To our knowledge, this is the most detailed review to date of the therapeutics landscape for Nipah and Hendra virus disease with the specific aim of supporting candidate prioritisation for clinical trials. We did not identify any ongoing or completed therapeutic efficacy RCTs for Nipah or Hendra virus infection. There were no data on *in vivo* drug resistance. ### Drugs The pipeline of therapeutics with the potential to be deployed rapidly at the outset of a henipavirus outbreak is currently limited to a few mAbs and repurposed small molecules with efficacy data from animal challenge models (Table 3). The comparative advantages of mAbs and small molecules are summarised in Table 4. View this table: [Table 3:](http://medrxiv.org/content/early/2024/03/15/2024.03.11.24304091/T3) Table 3: Nipah and Hendra Therapeutic Candidates Clinical Prioritisation View this table: [Table 4:](http://medrxiv.org/content/early/2024/03/15/2024.03.11.24304091/T4) Table 4: Comparative Advantages of Monoclonal Antibodies and Small Molecules Of the mAbs, only m102.4 has been studied in humans, with safety and pharmacokinetic (PK) data from a phase 1 RCT in healthy adults26. m102.4 is also the only mAb with efficacy (from challenge with NiV-B27, NiV-M28,30, and HeV29) and PK data (without challenge29,31) from two animal species. Further PK studies of mAb candidates to determine 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 negative for chloroquine. Remdesivir was the only small molecule with *in vivo* data from challenge with NiV-B43, the strain closely related to those causing recent Nipah outbreaks in Bangladesh and India62, and has accumulated acceptable safety data from its widespread intravenous use in COVID-1963. While favipiravir prevented death in NiV-M challenged hamsters after a subcutaneous loading dose followed by subcutaneous or oral maintenance doses48, and could be an attractive choice for post-exposure prophylaxis with a licensed oral formulation64, its non-linear clinical pharmacokinetics seen in Ebola65, influenza66, and COVID-1967 necessitate further dose optimisation prior to inclusion in Nipah trials. This non-linearity is thought to be explained by concentration-dependent aldehyde oxidase inhibition reproducible in non-human primates68, and it is unclear if there is an additional infection-specific contribution. PK studies of parenteral (including intravenous69) favipiravir in NiV-B inoculated non-human primates would be a key next step in favipiravir evaluation. Notably, favipiravir is associated with teratogenicity in four animal species64 and further data on its safety in humans are needed70,71. Ribavirin prolongs survival but does not prevent death in monkeys and hamsters challenged with HeV45 and NiV-M49,50, and is toxic to hamsters at high doses49. Issues with clinical tolerability (fatigue, anaemia, and hyperbilirubinaemia)38 are further likely to reduce adherence to a ribavirin-containing post-exposure prophylaxis regimen. Clinical reports of ribavirin in Nipah and Hendra outbreaks were all observational with dosing based on that used for Lassa fever42. Recent clinical and PK meta-analyses of ribavirin in Lassa treatment highlight the lack of robust evidence for its efficacy72 and that conventional dosing regimens are unlikely to reliably achieve the serum concentrations required to inhibit Lassa virus replication73. PK modelling is ongoing for available ribavirin dosing regimens for Nipah and Hendra. Ribavirin remains part of the Nipah treatment guidelines in India74,75 but not Bangladesh76. Consultation with Nipah stakeholders would establish whether the potential to delay death by hours to days justifies further use. Chloroquine did not protect ferrets47 or hamsters49 from NiV-M and was lethal at higher doses in hamsters49. The narrow therapeutic window of chloroquine is well-established in clinical practice, where it is a safe and effective antimalarial but has also been employed for rapid self-poisoning in deliberate overdose77,78. Chloroquine should not be used for the treatment or prevention of Nipah or Hendra infection. Promising *in vitro* efficacy has yet to translate into convincing *in vivo* protection for the experimental small molecules in Table 2. The parent drugs of 6-azauridine (azaribine) and ALS-8112 (lumicitabine) have been withdrawn from market and development respectively due to safety concerns of thrombosis79 and paediatric neutropenia80. It is unclear whether periodate heparin, fusion lipo-peptides, and defective viral particles can be manufactured at scale or are stable for stockpiling. ### Treatment Indications & Use Cases Antiviral drugs, whether mAbs or small molecules, appear to have a narrow temporal window within which they are likely to have clinically relevant efficacy, limiting their use to prophylaxis (pre- and post-exposure) and possibly early treatment19. They could also play a key role in providing bridging protection prior to vaccine response or availability. The time window for protection post-challenge81 is shorter with NiV-B than NiV-M in monkeys27 although this has yet to be validated in humans. Immunomodulators could be used in combination with pathogen-directed antivirals82 in later phases of infection when immunopathology is thought to dominate19 although there are as yet no data on such combinations. Rintatolimod was the only host-directed agent with *in vivo* efficacy data specifically for henipavirus infection identified from this review, providing only partial protection after low dose NiV-M challenge in hamsters50. ### Drug Evaluation This paucity of drug candidates and high-quality evidence overall underscores the challenges of clinical development of therapeutics for rare but high-threat infections with pandemic potential. Despite further Nipah outbreaks in the past year, there remain insufficient cases under the current epidemiological situation to obtain the human phase 3 RCT efficacy data necessary for licensure83 or to attract substantial commercial investment. Alternative approaches similar to the regionally driven end-to-end West African Lassa fever Consortium84 framework are needed19. The requirement for BSL-4 precautions for pre-clinical studies of NiV and HeV also restricts these to a small number of specialist facilities, few of which are located where Nipah or Hendra outbreaks have occurred. #### Animal Studies In the absence of outbreak RCTs, efficacy evaluation of Nipah and Hendra therapeutics is reliant on controlled animal challenge studies. The variable agreement between *in vitro* and *in vivo* efficacy results for most of the small molecules identified in this review emphasises the importance of animal efficacy data for clinical prioritisation. The United States Food and Drug Administration allows for approval of drugs for conditions which threaten health security under the Animal Rule85 when field trials are not possible, provided four criteria are met: 1) sufficient understanding of the pathophysiology of the condition and mechanism of its reduction by the product; 2) efficacy demonstrated in at least two animal species or one species which is a well-characterised model for predicting the product’s response in humans; 3) animal study endpoint clearly related to the desired outcome in humans, typically reduction in mortality or major morbidity; and 4) PK and pharmacodynamic (PD) data from animals and humans supporting selection of an effective dose in humans. Anti-infective agents approved under the Animal Rule include: raxibacumab and obitoxaximab for anthrax, antibiotics like ciprofloxacin for plague, and tecovirimat and brincidofovir for smallpox. The European Medicines Agency has a similar Exceptional Circumstances86 mechanism for granting marketing authorisation to medicines where collection of comprehensive efficacy and safety data under normal conditions of use is not possible. The clinical and pathological features as well as the strengths and limitations of the major animal models in Nipah (AGMs87, ferrets88, and Syrian Golden Hamsters89) have been reviewed90 by the Coalition for Epidemic Preparedness Innovations (CEPI). AGMs are closest in physiology to humans but have less consistent neurological symptoms than hamsters and ferrets90. CEPI are also improving these models, particularly through standardising the virus challenge stock90. Supporting access to NiV-B strains as well as standardisation of dose and route of challenge for each model could aid comparability across studies. For therapeutics studies, having uninfected controls to assess drug toxicity thresholds, ideally in the context of PK-PD studies to determine *in vivo* EC50 values and concentration-efficacy relationships, is crucial for dose optimisation to derisk human studies. #### Clinical Studies Human data remain essential for safety evaluation85. Phase 1 first-in-human safety data need to be collected for new therapeutic agents and existing experience from repurposed agents critically appraised for any potential exacerbation of adverse effects by the pathophysiology of Nipah or Hendra infection prior to deployment in outbreak settings. Within an outbreak, these therapeutics should be evaluated in well-designed phase 2 clinical trials integrated and sustained in health systems91 using pre-approved standardised protocols maximising statistical and operational efficiency in assessment of internationally-agreed core outcome measures92. Where possible, drug concentrations should be measured at the same time points as efficacy and safety outcomes to characterise and quantify PK-PD relationships, including at different stages of disease. Where RCTs are not possible, observational studies employing enhanced clinical characterisation protocols93 incorporating the same outcome measures could provide higher-quality observational data than is currently available94. The long-term neurological sequelae of Nipah encephalitis95–97 also merit more systematic characterisation and potential inclusion as outcomes. Outbreaks of high-threat infections invoke the ethical duty98,99 to conduct inclusive research with speed and rigor. Community and stakeholder engagement99, including on design and interventions in trials, are key to support genuinely informed consent and maintain trust in the scientific process100. ### Frameworks & Tools It is vital that potential therapeutics and their appropriate dosing regimens are selected, optimised, and stockpiled based on all available clinical and pre-clinical evidence well in advance of any outbreak. This continuous iterative process should be guided by disease-specific, and where appropriate product-specific, target product profiles101 (comprising indication, safety, efficacy, route, stability, and affordability characteristics) developed through consensus among all relevant stakeholders, including regulators, end users, and communities. Systems pharmacology, statistical, mathematical, and economic modelling are powerful tools to support decision-making by providing a formal framework for integration of (typically sparse) data from multiple study types, species, and diseases, as well as informing design efficiency of phase 1 and phase 2 RCTs. ## Conclusion At present, there is sufficient evidence to trial only m102.4 (mAb) and remdesivir (small molecule) (singly and/or in combination) for prophylaxis and early treatment of Nipah virus infection. In addition to well-designed RCTs, *in vivo* PK-PD studies to support drug selection and dose optimisation for all high-threat infections are needed. ## Supporting information Supplementary Appendix [[supplements/304091_file03.docx]](pending:yes) ## Data Availability All data produced in the present work are contained in the manuscript ## Contributors XHSC, JD, PWH, and PLO conceptualised this review. XHSC wrote the study protocol and designed the search strategy with EH who conducted the literature search. XHSC piloted the review and screened studies with ILH and SL. XHSC, ILH, BJKC, MZH, JT, LMJ, and TPH extracted and tabulated data. MZH conducted the risk of bias assessment with BJKC. MZH and XHSC produced the figures. XHSC wrote the initial draft with ILH and BJKC. All authors critically reviewed, edited, and approved the final version of the manuscript. ## Acknowledgements XHSC is a United Kingdom (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 would like to thank all members of the Pandemic Sciences Institute Henipavirus Programme as well as delegates of the International Pandemic Sciences Conference 2023 and the Nipah Virus 25th Anniversary Symposium, both in Oxford, for illuminating discussions. * Received March 11, 2024. * Revision received March 11, 2024. * Accepted March 15, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 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(17): 1229–35. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM200004273421701&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10781618&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000086660600001&link_type=ISI) 2. 2.Luby SP, Rahman M, Hossain MJ, et al. Foodborne transmission of Nipah virus, Bangladesh. Emerg Infect Dis 2006; 12(12): 1888–94. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid1212.060732&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17326940&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000242301900013&link_type=ISI) 3. 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(19): 1804–14. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/nejmoa1805376&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31067370&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 4. 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(11): e3302. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pntd.0003302&link_type=DOI) 5. 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(2): 393–6. 6. 6.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(3): 533–5. 7. 7.Hossain MJ, Gurley ES, Montgomery JM, et al. Clinical presentation of Nipah virus infection in Bangladesh. Clin Infect Dis 2008; 46(7): 977–84. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/529147&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18444812&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000253817800003&link_type=ISI) 8. 8.Sejvar JJ, Hossain J, Sana SK, et al. Long-term neurological and functional outcome in Nipah virus infection. Annals of Neurology 2007; 62(3): 235–42. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.21178&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17696217&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000249937000009&link_type=ISI) 9. 9.Broder CC, Wong KT. Henipaviruses. Neurotropic Viral Infections: Volume 1: Neurotropic RNA Viruses: Springer International Publishing; 2016: 45–83. 10. 10.Chua KB, Goh KJ, Wong KT, et al. Fatal encephalitis due to Nipah virus among pig-farmers in Malaysia. Lancet 1999; 354(9186): 1257–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(99)04299-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10520635&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000083010400012&link_type=ISI) 11. 11.Paton NI, Leo Y, Zaki SR, et al. Outbreak of Nipah-virus infection among abattoir workers in Singapore. Lancet 1999; 354(9186): 1253–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(99)04379-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10520634&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000083010400011&link_type=ISI) 12. 12.Chua KB. Epidemiology, surveillance and control of Nipah virus infections in Malaysia. Malaysian Journal of Pathology 2010; 32(2): 69–73. 13. 13.Suit BY, Hassan L, Krauss SE, et al. Mental Model of Malaysian Pig Farmers in Implementing Disease Prevention and Control Practices. Frontiers in Veterinary Science 2021; 8 (no pagination). 14. 14.Ching PK, de los Reyes VC, Sucaldito MN, et al. Outbreak of henipavirus infection, Philippines, 2014. Emerg Infect Dis 2015; 21(2): 328–31. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2102.141433&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25626011&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 15. 15.Chadha MS, Comer JA, Lowe L, et al. Nipah virus-associated encephalitis outbreak, Siliguri, India. Emerg Infect Dis 2006; 12(2): 235–40. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid1202.051247&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16494748&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000235287400009&link_type=ISI) 16. 16.Arunkumar G, Chandni R, Mourya DT, et al. Outbreak Investigation of Nipah Virus Disease in Kerala, India, 2018. J Infect Dis 2019; 219(12): 1867–78. 17. 17.Hassan MZ, Sazzad HMS, Luby SP, et al. Nipah virus contamination of hospital surfaces during outbreaks, Bangladesh, 2013-2014. Emerging Infectious Disease s 2018; 24(1): 15–21. 18. 18.World Health Organization. Disease Outbreak News: Nipah Virus Infection - India. 2023. [https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON490](https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON490) (accessed 12 February 2024). 19. 19.Hassan MZ, Shirin T, Satter SM, et al. Nipah virus disease: what can we do to improve patient care? Lancet Infect Dis 2024. 20. 20.Gomez Roman R, Tornieporth N, Cherian NG, et al. Medical countermeasures against henipaviruses: a review and public health perspective. Lancet Infect Dis 2022; 22(1): e13–e27. 21. 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: 63–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.antiviral.2018.09.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 22. 22.Sterne JAC, Savovic J, Page MJ, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ 2019; 366: l4898. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE3OiIzNjYvYXVnMjhfMi9sNDg5OCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzE1LzIwMjQuMDMuMTEuMjQzMDQwOTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 23. 23.Sterne JA, Hernan MA, Reeves BC, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016; 355: i4919. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE4OiIzNTUvb2N0MTJfMTEvaTQ5MTkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8xNS8yMDI0LjAzLjExLjI0MzA0MDkxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 24. 24.Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol 2014; 14: 43. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2288-14-43&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24667063&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 25. 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. 26. 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 Infectious Diseases 2020; 20(4): 445–54. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(19)30634-6&link_type=DOI) 27. 27.Mire CE, Satterfield BA, Geisbert JB, et al. Pathogenic Differences between Nipah Virus Bangladesh and Malaysia Strains in Primates: Implications for Antibody Therapy. Scientific Reports 2016; 6. 28. 28.Geisbert TW, Mire CE, Geisbert JB, et al. Therapeutic treatment of Nipah virus infection in nonhuman primates with a neutralizing human monoclonal antibody. Science Translational Medicine 2014; 6(242) (no pagination). 29. 29.Bossart KN, Geisbert TW, Feldmann H, et al. A Neutralizing Human Monoclonal Antibody Protects African Green Monkeys from Hendra Virus Challenge. Science Translational Medicine 2011; 3(105). 30. 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 Pathogens 2009; 5(10) (no pagination). 31. 31.Zhu Z, Bossart KN, Bishop KA, et al. Exceptionally potent cross-reactive neutralization of Nipah and Hendra viruses by a human monoclonal antibody. Journal of Infectious Diseases 2008; 197(6): 846–53. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/528801&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18271743&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 32. 32.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. Journal of Infectious Diseases 2020; 221: S471–S9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiz515&link_type=DOI) 33. 33.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 Reports 2021; 36(9) (no pagination). 34. 34.Dong J, Cross RW, Doyle MP, et al. Potent Henipavirus Neutralization by Antibodies Recognizing Diverse Sites on Hendra and Nipah Virus Receptor Binding Protein. Cell 2020; 183(6): 1536–50.e17. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2020.11.023&link_type=DOI) 35. 35.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(2): 459–65. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.virol.2009.03.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19328514&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265663100025&link_type=ISI) 36. 36.Guillaume V, Contamin H, Loth P, et al. Antibody prophylaxis and therapy against Nipah virus infection in hamsters. Journal of Virology 2006; 80(4): 1972–8. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjk6IjgwLzQvMTk3MiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzE1LzIwMjQuMDMuMTEuMjQzMDQwOTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 37. 37.Playford EG, McCall B, Smith G, et al. Human Hendra virus encephalitis associated with equine outbreak, Australia, 2008. Emerging Infectious Diseases 2010; 16(2): 219–23. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid1602.090552&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20113550&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 38. 38.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. Journal of Infection 2019; 78(6): 491–503. 39. 39.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 Infectious Diseases 2019; 6(Supplement 2): S659–S60. 40. 40.Radhakrishnan C, Renjith TP, Arshad F, 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. Clinical Infectious Diseases 2020; 71(1): 152–7. 41. 41.Warrier A. A single case outbreak of Nipah Encephalitis from India in May-June 2019. International Journal of Infectious Diseases 2020; 101(Supplement 1): 247. 42. 42.Chong HT, Kamarulzaman A, Tan CT, et al. Treatment of acute Nipah encephalitis with ribavirin. Annals of Neurology 2001; 49(6): 810–3. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.1062&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11409437&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000169091400021&link_type=ISI) 43. 43.Lo MK, Feldmann F, Gary JM, et al. Remdesivir (GS-5734) protects African green monkeys from Nipah virus challenge. Sci Transl Med 2019; 11(494). 44. 44.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 Infectious Diseases 2017; 4(Supplement 1): S737. 45. 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. Journal of Virology 2010; 84(19): 9831–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjEwOiI4NC8xOS85ODMxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDMvMTUvMjAyNC4wMy4xMS4yNDMwNDA5MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 46. 46.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(2): 218–27. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiy152&link_type=DOI) 47. 47.Pallister J, Middleton D, Crameri G, et al. Chloroquine administration does not prevent nipah virus infection and disease in ferrets. Journal of Virology 2009; 83(22): 11979–82. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjExOiI4My8yMi8xMTk3OSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzE1LzIwMjQuMDMuMTEuMjQzMDQwOTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 48. 48.Dawes BE, Kalveram B, Ikegami T, et al. Favipiravir (T-705) protects against Nipah virus infection in the hamster model. Scientific Reports 2018; 8(1). 49. 49.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. Journal of General Virology 2010; 91(3): 765–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1099/vir.0.017269-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19889926&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000275587200021&link_type=ISI) 50. 50.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(5): 1768–72. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYWFjIjtzOjU6InJlc2lkIjtzOjk6IjUwLzUvMTc2OCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzE1LzIwMjQuMDMuMTEuMjQzMDQwOTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 51. 51.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. (Special Issue: Twenty years of Nipah virus research.). Journal of Infectious Diseases 2020; 221(Suppl. 4): S480–S92. 52. 52.Mathieu C, Dhondt KP, Chalons M, et al. Heparan sulfate-dependent enhancement of henipavirus infection. Mbio 2015; 6(2) (no pagination). 53. 53.Mathieu C, Augusto MT, Niewiesk S, et al. Broad spectrum antiviral activity for paramyxoviruses is modulated by biophysical properties of fusion inhibitory peptides. Scientific Reports 2017; 7. 54. 54.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 Pathogens 2010; 6(10) (no pagination). 55. 55.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(2): e0329421. 56. 56.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. Archives of Virology 2005; 150(3): 521–32. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00705-004-0417-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15526144&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 57. 57.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. Journal of Virology 2009; 83(10): 5148–55. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjEwOiI4My8xMC81MTQ4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDMvMTUvMjAyNC4wMy4xMS4yNDMwNDA5MS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 58. 58.Lo MK, Jordan R, Arvey A, et al. GS-5734 and its parent nucleoside analog inhibit Filo-, Pneumo-, and Paramyxoviruses. Scientific Reports 2017; 7. 59. 59.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). Microbiology Spectrum 2021; 9(3) (no pagination). 60. 60.Welch SR, Tilston NL, Lo MK, et al. Inhibition of Nipah virus by defective interfering particles. (Special Issue: Twenty years of Nipah virus research.). Journal of Infectious Diseases 2020; 221(Suppl. 4): S460–S70. 61. 61.Wolf MC, Freiberg AN, Zhang T, et al. A broad-spectrum antiviral targeting entry of enveloped viruses. Proceedings of the National Academy of Sciences of the United States of Ameri ca 2010; 107(7): 3157–62. 62. 62.Whitmer SLM, Lo MK, Sazzad HMS, et al. Inference of Nipah virus evolution, 1999-2015. Virus Evol 2021; 7(1): veaa062. 63. 63.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(1): 672. 64. 64.Pharmaceuticals and Medical Devices Agency. Avigan (Favipiravir) Review Report. Tokyo, Japan, 2014. 65. 65.Nguyen TH, 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(2): e0005389. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pntd.0005389&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 66. 66.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: 103125. 67. 67.Irie K, Nakagawa A, Fujita H, et al. Population pharmacokinetics of favipiravir in patients with COVID-19. CPT Pharmacometrics Syst Pharmacol 2021; 10(10): 1161–70. 68. 68.Madelain V, Guedj J, Mentre F, et al. Favipiravir Pharmacokinetics in Nonhuman Primates and Insights for Future Efficacy Studies of Hemorrhagic Fever Viruses. Antimicrob Agents Chemother 2017; 61(1). 69. 69.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. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.antiviral.2017.12.021&link_type=DOI) 70. 70.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(2): 45–51. 71. 71.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(1): e33676. 72. 72.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(1). Emerg Infect Dis 2022; 28(8): 1559–68. 73. 73.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(3): e0010289. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pntd.0010289&link_type=DOI) 74. 74.Government of India Ministry of Health and Family Welfare. Clinical Management Protocol for Nipah Virus Disease. 2018. 75. 75.Government of Kerala Department of Health and Family Welfare. Nipah Virus Infection Guidelines for Surveillance, Diagnosis, Treatment, Prevention and Control. 2021. 76. 76.Government of the People’s Republic of Bangladesh Ministry of Health and Family Welfare. National Guideline for Management, Prevention and Control of Nipah Virus Infection including Encephalitis. 2018. 77. 77.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(9): e1003252. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pmed.1003252&link_type=DOI) 78. 78.Haeusler IL, Chan XHS, Guerin PJ, White NJ. The arrhythmogenic cardiotoxicity of the quinoline and structurally related antimalarial drugs: a systematic review. BMC Med 2018; 16(1): 200. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12916-018-1188-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 79. 79.Drell W, Welch AD. Azaribine-homocystinemia-thrombosis in historical perspective. Pharmacol Ther 1989; 41(1-2): 195–206. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2469090&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 80. 80.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(7): e0288271. 81. 81.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: 105658. 82. 82.Kaufmann SHE, Dorhoi A, Hotchkiss RS, Bartenschlager R. Host-directed therapies for bacterial and viral infections. Nat Rev Drug Discov 2018; 17(1): 35–56. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrd.2017.162&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28935918&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 83. 83.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(39): 5600–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2021.08.027&link_type=DOI) 84. 84.ISARIC. West Africa Lassa fever Consortium (WALC). 2023. [https://isaric.org/research-2/lassa-fever-resources/walc/](https://isaric.org/research-2/lassa-fever-resources/walc/) (accessed 10 January 2024). 85. 85.United States Food and Drug Administration. Animal Rule Approvals. 2002. [https://www.fda.gov/drugs/nda-and-bla-approvals/animal-rule-approvals](https://www.fda.gov/drugs/nda-and-bla-approvals/animal-rule-approvals) (accessed 4 January 2024). 86. 86.European Medicines Agency. Exceptional Circumstances. 2024. [https://www.ema.europa.eu/en/glossary/exceptional-circumstances](https://www.ema.europa.eu/en/glossary/exceptional-circumstances) (accessed 10 January 2024). 87. 87.Mire CE, Satterfield BA, Geisbert TW. Nonhuman Primate Models for Nipah and Hendra Virus Countermeasure Evaluation. Methods Mol Biol 2023; 2682: 159–73. 88. 88.Rockx B, Mire CE. Ferret Models for Henipavirus Infection. Methods Mol Biol 2023; 2682: 205–17. 89. 89.Juelich T, Smith J, Freiberg AN. Syrian Golden Hamster Model for Nipah Virus Infection. Methods Mol Biol 2023; 2682: 219–29. 90. 90.Centre for Epidemic Preparedness Innovations. Nipah Virus Assays and Animal Models for Vaccine Development, 2021. 91. 91.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 2023. 92. 92.Williamson PR, Altman DG, Blazeby JM, et al. Developing core outcome sets for clinical trials: issues to consider. Trials 2012; 13: 132. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1745-6215-13-132&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22867278&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 93. 93.ISARIC Clinical Characterisation Protocol Investigators Group. ISARIC/WHO Clinical Characterisation Protocol. 2023. [https://isaric.net/ccp/](https://isaric.net/ccp/) (accessed 9 January 2024). 94. 94.Dunning JW, Merson L, Rohde GGU, et al. Open source clinical science for emerging infections. Lancet Infect Dis 2014; 14(1): 8–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(13)70327-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24355025&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) 95. 95.Lee KE, Umapathi T, Tan CB, et al. The neurological manifestations of Nipah virus encephalitis, a novel paramyxovirus. Ann Neurol 1999; 46(3): 428–32. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/1531-8249(199909)46:3<428::AID-ANA23>3.0.CO;2-I&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10482278&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000082360000023&link_type=ISI) 96. 96.Tan CT, Goh KJ, Wong KT, et al. Relapsed and late-onset Nipah encephalitis. Ann Neurol 2002; 51(6): 703–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.10212&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12112075&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000175863700007&link_type=ISI) 97. 97.Sejvar JJ, Hossain J, Saha SK, et al. Long-term neurological and functional outcome in Nipah virus infection. Ann Neurol 2007; 62(3): 235–42. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/ana.21178&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17696217&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F15%2F2024.03.11.24304091.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000249937000009&link_type=ISI) 98. 98.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 2023. 99. 99.World Health Organization. Emergency use of unproven clinical interventions outside clinical trials: ethical considerations, 2022. 100.100.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(9): MR000065. 101.101.World Health Organization. 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](https://www.who.int/observatories/global-observatory-on-health-research-and-development/analyses-and-syntheses/target-product-profile/who-target-product-profiles) (accessed 5 January 2024). 102.102.British National Formulary. Remdesivir. 2024. [https://bnf.nice.org.uk/drugs/remdesivir/](https://bnf.nice.org.uk/drugs/remdesivir/) (accessed 6 January 2024).