We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleFree Access

Specific polyclonal F(ab’)2 neutralize a large panel of highly pathogenic avian influenza A viruses (H5N1) and control infection in mice

    ,
    Frédéric Jacquot

    INSERM Jean Mérieux BSL4 Laboratory, Lyon, France

    ,
    Sareth Rith

    Institut Pasteur du Cambodge, Phnom Penh, Cambodia

    , , ,
    Caroline Carbonnelle

    INSERM Jean Mérieux BSL4 Laboratory, Lyon, France

    ,
    Vincent Lotteau

    CIRI, Centre de Recherche en Infectiologie, Lyon, France

    INSERM U1111, Lyon, France

    Université de Lyon, France

    , ,
    Hervé Raoul

    INSERM Jean Mérieux BSL4 Laboratory, Lyon, France

    ,
    Philippe Buchy‡,

    Institut Pasteur du Cambodge, Phnom Penh, Cambodia

    Authors contributed equally

    Search for more papers by this author

    &
    Published Online:https://doi.org/10.2217/imt.14.40

    Abstract

    Aim: There is still no specific therapy for infection with the highly pathogenic avian influenza A virus (HPAI) H5N1, which caused 39 human cases with a 64% fatality rate in 2013. Materials & methods: We prepared highly purified specific equine polyclonal immunoglobulin fragments (F(ab’)2) against H5N1 and tested them for efficacy in vitro and with different administration schedules in H5N1-challenged BALB/c mice. Results:in vitro, F(ab’)2 neutralized 21 different H5N1 strains from different areas, representative of 11 different clades and sub-clades and 9 years of evolution of the virus. In vivo mouse experiments identified that the most efficient administration protocol consists of five consecutive daily injections after infection; 10 mg/kg giving a 60% increase in survival. Conclusion: These data demonstrate the ability of anti-H5N1 F(ab’)2 to markedly reduce the mortality and morbidity associated with infection of mice with HPAI H5N1 virus, and their potential for human therapy.

    Figure 1.  Phylogenetic analysis of influenza A(H5N1) virus strains isolated worldwide from 2004 to 2013.

    The multiple sequence alignment of the 1659 nucleotide sequence (position: 49 to 1707) of the hemagglutinin (HA) genes was conducted using ClustalW, version 2. The phylogenetic analysis was carried as a distance-based neighbor-joining using the Jukes–Cantor model. The phylogenetic tree was generated using MEGA v.5.05 software with 1000 bootstrap replicates (values ≥70 shown on branch). Scale bar indicates number of nucleotide substitutions per site.

    Figure 2.  Characterization of purified equine anti-H5N1 F(ab’)2 (FBF001) by high performance liquid chromatography (HPLC) analysis.

    A total of 120 µg of protein contained in 30 µl was injected in a TSK G3000 SW column packed with silica support (length 60 cm – ID 7.5 mm), and the separation was performed at a flow rate of 0.5 ml/min to determine the molecular size distribution of anti-H5N1 immunoglobulin fragments at different production stages. Peak identification is presented in the table.

    TR: Time of retention.

    Figure 3.  In vivo neutralization efficiency of F(ab’)2 anti-H5N1 (FBF001) against 10 × LD50 of influenza H5N1 virus.

    10 × lethal dose 50% (LD50) of viruses were incubated 1 h at 34°C in the presence of variable amounts of F(ab’)2 anti-H5N1 (µg). These mixture preparations were injected intraperitoneally in different groups of mice (n = 10) at Day 0. Mice were monitored for clinical signs and survival. Survival analysis was performed on Day+14 after challenge.

    Figure 4.  Efficacy of FBF001 injected via intraperitoneal route to control H5N1 infection in mouse.

    40 mg/kg of FBF001 were injected intraperitoneally to BALB/c mice 1 day after or on Days+1, +2, +3, +4 and +5 after intranasal challenge with 10 × LD50 of the A/Vietnam/1194/2004 H5N1 influenza virus strain (n = 8/group). Mice were monitored for clinical signs and survival. Significance was evaluated for each treated group in comparison with virus control (group 3), using the Gehan–Breslow–Wilcoxon test.

    ***p < 0.001.

    Figure 5.  Dose-effect of FBF001 on mouse survival rate.

    The indicated amount of FBF001 was injected intraperitoneally into mice (10 per group) on Day+1, Day+2, Day+3, Day+4 and Day+5 and the mice were challenged intranasally on Day 0 with A/Vietnam/1194/2004 (H5N1) virus. Mice were monitored daily for clinical signs and survival during 14 days. Significance was evaluated for each treated group in comparison with virus control group, using the Gehan–Breslow–Wilcoxon test. (A) Survival rate of mice after viral challenge with 1 × lethal dose 50% (LD50) of H5N1 influenza virus. (B) Survival rate of mice after viral challenge with 10 × LD50 of H5N1 influenza virus. (C) Survival rate of mice after viral challenge with 100 × LD50 of H5N1 influenza virus.

    *p < 0.1.

    **p < 0.01.

    ***p < 0.001.

    ****p < 0.0001.

    Since 2003, highly pathogenic avian influenza (HPAI) A/H5N1 (H5N1) viruses have been associated with almost 650 reported infections to the WHO, with a high mortality rate (59%) [1]. Human infection with influenza H5N1 virus often occurs after direct handling or after close contact with infected poultry [2,3]. So far, very few cases of human-to-human transmission have been identified [3,4]. However, the continuing occurrence of human infections since 2003 provide the H5N1 virus opportunities for reassortment and/or mutations associated with better person-to-person transmission.

    In humans, severe H5N1 influenza infection is usually associated with viral loads and hypercytokinemia [3]. In one study, infectious virus was recovered from the blood of 40% of severely ill patients [5]. Specific antibodies can be detected after approximately 2–3 weeks [6,7]. The combination of the direct damage caused by the virus and the intense inflammatory reaction of the patients is probably responsible for the high severity of H5N1 infection [8,9].

    Since 2003, the fatality rate associated with H5N1 infection has not improved. Although several new antiviral drugs have been developed recently (e.g., RNA polymerase inhibitors such as Favipiravir, and inhibitors of the hemagglutinin-mediated viral entry) [10,11], the antiviral therapies currently marketed are limited to adamantanes (amantadine and rimantadine), which inhibit the M2 ion channel, and to the neuraminidase inhibitors (oseltamivir, peramivir, zanamivir and lanimavir). Owing to increased resistance to adamantanes in clade 1 viruses isolated in South-East Asian countries until 2012, the treatment mainly relies on neuraminidase inhibitors, such as oseltamivir. Reduced sensitivity of some H5N1 virus strains to neuraminidase inhibitors has been observed occasionally [12–14] and appearance of resistant mutations has also been reported in patients during the course of the treatment [15,16].

    Passive immunotherapy could be an interesting alternative or an additional strategy when used in combination with antiviral drugs for the treatment of H5N1 infections. Some studies in mice have already demonstrated the efficacy of monoclonal [17–19] or polyclonal antibodies on H5N1 infections [20–22]. Human convalescent plasma was also reported to efficiently help in controlling the infection in severely ill patients [23,24].

    H5N1 virus is rapidly evolving, which poses serious public health concerns [25]. This fast evolution makes prophylactic vaccine development complicated, and in this context the use of polyclonal immunoglobulins appears to have many advantages, limiting the risk of viral escape mutations and allowing rapid development and production of a novel efficient treatment. Moreover, such products are particularly well suited for emergency conditions, as is currently the case for rabies [26,27] or envenomation [28–30]. While in the past equine antisera were sometimes the cause of significant side effects due to anaphylactic shock, mainly due to the presence of Fc fragments, the new generation of processed and purified polyclonal immunoglobulins containing highly purified F(ab’)2 fragments, are well tolerated [26].

    Here we report the development and the evaluation in a mouse model of specific polyclonal anti-H5N1 immunoglobulins to investigate their therapeutic potential for the clinical management of humans exposed to HPAI H5N1 virus.

    Material & methods

    Equine hyperimmune sera

    Four healthy French trotter horses were immunized with an anti-H5N1 split vaccine containing the seed-inactivated A/Vietnam/1194/2004 H5N1 virus. Horses had no detectable antibodies against H5N1 virus before immunization and were strictly controlled for several viruses. Blood samples were collected regularly up to 4 weeks after immunization. Plasma were prepared and stored at -20°C.

    Preparation of equine F(ab’)2 fragments

    Pooled horse plasma was purified as described previously to obtain highly purified F(ab’)2 fragments [31,32]. The main purification steps consist of: anion-exchange chromatography steps, which eliminate proteins including albumin; hydrolysis of whole immunoglobulins into F(ab’)2 fragments in order to eliminate Fc fragments, which are horse specific; and pasteurization (heat treatment) at 60°C for 10 h for the viral safety of the product. The final bulk product (FBF001) is filtered at a level of 0.2 µm and then stored at +5°C ± 3°C until use.

    High-performance liquid chromatography

    High-performance liquid chromatography (HPLC) using a TSK G3000 SW column packed with silica support (length 60 cm; internal diameter [ID] 7.5 mm) was employed to determine the molecular size distribution of anti-H5N1 immunoglobulin fragments at different production stages. A total of 120 µg of protein contained in 30 µl was injected, and the separation was performed at a flow rate of 0.5 ml/min.

    Viruses & animals

    All influenza A H5N1 strains used in this study were amplified using Madin-Darby canine kidney (MDCK) cells. The infectious titer was determined by limit-dilution in MDCK cells cultures, and expressed as the 50% tissue culture infectious dose (TCID50). The H5N1 strains used in this study were selected for their representativeness of H5N1 virus circulation in the world since 2003 and to represent the last 9 years of virus evolution in Cambodia (with viruses from clades 1, 1.1.1 and 1.1.2) (Figure 1).

    All in vitro experiments involving infectious virus were performed in a biosafety level 3 laboratory. In vivo experiments, in 8–10-week-old BALB/c mice (Charles River Laboratories, MA, USA) were performed in a biosafety level 4 laboratory. Mice were housed and manipulated according to the guidelines of Directive 2010/63/UE.

    Mice were anesthetized with 5% isoflurane (1-chloro-2,2,2-trifluoroethyl difluoromethyl ether) in an induction chamber before viral infection and treatment with FBF001.

    The 50% lethal dose (LD50) for A/Vietnam/1194/2004 virus was determined in BALB/c mice by mortality analysis of six groups of five mice infected with six different dilutions of infectious virus (mock or 102 to 106 × TCID50). The titer was calculated by the Reed-Muench method [33]. A/Vietnam/1194/2004 virus was diluted in D-PBS to obtain solutions titrated at respectively 1, 10 and 100 × LD50 (corresponding to 103, 104 and 105 × TCID50, respectively). Mice were inoculated through intranasal route with 16 µl of one of the infectious virus doses.

    Microneutralization & hemagglutination inhibition assays

    For standard microneutralization (MN) assay, 100 × TCID50 of each virus strain tested (Table 1) were incubated with two different serial dilutions of FBF001 product (solution concentration: 29.4 g/l; from 1/20 to 1/18,000) for 1 h at room temperature prior to addition to MDCK cells. Cell monolayers were incubated for a further 3 days and examined for cytopathic effects. Determination of the end point neutralizing antibody titers was performed in four wells per dilution. The neutralizing titer was defined as the reciprocal of the highest dilution of serum at which the infectivity of 100 × TCID50 of H5N1 virus for MDCK cells was completely neutralized in 50% of the wells. The titer was calculated by the Reed-Muench method [33]. Hemagglutination inhibition (HI) testing was performed with human group O red blood cells according to the method described by Rowe [34]. All tests were performed in triplicate.

    Protective efficacy of polyclonal equine F(ab’)2 against A/Vietnam/1194/2004 in mice

    Mixtures of FBF001 and virus were prepared in D-PBS to obtain doses corresponding to 5, 0.5, 0.1, 0.05, 0.025, 0.01 and 0.005 µg FBF001/animal. Each dose also contained 10 × LD50 of virus. After 1 h of incubation at 34°C, mice received an intraperitoneal (i.p.) injection (300 µl) of one of the mixtures.

    In vivo prophylactic & therapeutic efficacy of polyclonal equine F(ab’)2 against A/Vietnam/1194/2004 in mice

    8–10 week-old BALB/c mice were inoculated intranasally (i.n.) with 1, 10 or 100 × LD50 of virus at day 0. 200 µl of FBF001 solutions were injected i.p. to obtain final doses of 2.5, 5, 10, 20 or 40 mg/kg, 1 day after infection or during 5 consecutive days after infection. As negative control, non-infected mice were i.p. inoculated with FBF001 with the highest concentration of FBF001. As positive control, mice were i.p. inoculated with 200 µl of buffer.

    Statistical analyses

    • • in vitro data: all statistical analyses were performed using Stata/SE version 12.0 (StataCorp, TX, USA). Significance was assigned at p = 0.05 for all parameters and the standard deviation (SD) was calculated for mean. Non-parametric test Wilcoxon rank-sum test were used for continuous variables;

    • • In vivo data: all statistical analyses were performed using GraphPad Prism 6 (GraphPad Software, CA, USA). Survival data were analyzed by Gehan-Breslow-Wilcoxon test (treated group vs non-treated group).

    Results

    Preparation of highly purified F(ab’)2 fragments

    The production of highly purified F(ab’)2 fragments was based on hydrolysis and purification of hyperimmune equine plasma, produced by immunization of horses with inactivated A/Vietnam/1194/2004 H5N1 virus. Equine hyperimmune plasma was processed, following a validated process described previously [31,32]. SDS-PAGE showed that digestion with pepsin produces fragments of approximately 100 kDa, corresponding to F(ab’)2 fragments (data not shown). Smaller proteins, including albumin, α- and β-globulins and Fc fragments, which are responsible for the high reactogenicity of the crude preparations are eliminated by the process. At the end of the purification process, HPLC analysis of the bulk solution of immunoglobulins (FBF001) showed the presence of approximately 90% of F(ab’)2 fragments in the product (Figure 2). The proportion of F(ab’)2 plus Fab’ in the product was almost 99%, showing the very high purity level of this polyclonal immunoglobulin fragments solution.

    in vitro activity of FBF001 anti-H5N1 F(ab’)2

    FBF001 was tested against the homologous strain A/Vietnam/1194/2004 and titers of 1/2560 and 1/4000 were obtained by HI and MN tests, respectively.

    Cross-neutralization activity of FBF001

    A study was designed to confirm the neutralizing activity of FBF001 on 21 different H5N1 strains isolated over a 9-year period in different parts of the world, and representative of 11 different clades/sub-clades that emerged worldwide following H5N1 virus natural evolution (Figure 1). in vitro neutralization titers ranged from 1/1000 to 1/8000 for all clades tested (Table 1). These neutralization results were confirmed by HI assay with titers ranging from 1/960 to 1/2560 (Table 1). The mean MN and HI titers were respectively 1/4416 and 1/1600 for clade 1 viruses, 1/3666 and 1/1600 for clade 2 viruses, and 1/8000 and 1/1920 for clade 4, 7 and 9 viruses. None of the differences of MN and HI titers between clade 1, 2, 4, 7 and 9 viruses were significant.

    Protective efficacy of FBF001 in vivo

    To confirm the neutralizing activity of FBF001 in vivo, specific experiments were performed using a BALB/c mice model. This mouse model has already been shown susceptible to non-adapted H5N1 strains [35], with evidence of virus replication in the lungs.

    As shown in Figure 3, 0.1 µg of FBF001 was sufficient to prevent the lethal effects of 10 × LD50 of H5N1 viruses i.n. injected in mice and 5 ng protected 60% of animals against death by avian influenza infection.

    In vivo proof-of-concept of efficacy of FBF001 to treat H5N1 infection in mice

    The BALB/c i.n. mouse model was also used to assess the potential therapeutic efficacy of FBF001 against H5N1 virus, using various bodyweight doses of FBF001. One or multiple doses of FBF001 at 40 mg/kg were i.p. injected in mice inoculated with 10 × LD50 of A/Vietnam/1194/2004 H5N1 influenza virus. Mice received FBF001 24 h after virus challenge, or at days +1, +2, +3, +4 and +5 after challenge.

    When treatment was administered for 5 consecutive days after viral challenge (days +1 to +5; group 2 [Figure 4]) 100% protection against H5N1 challenge was observed. One dose administered 24 h after viral challenge resulted in a 48-h delay in mortality, but ultimately no significant effect on mouse survival rate was observed in comparison with the control group.

    Determination of the optimal therapeutic dose in mice

    The same model was used to perform a dose-ranging study with multiple conditions of experimental infection, designed to evaluate the optimal dose of FBF001 to be used in a Day+1 to Day+5 protocol. Doses varying from 2.5 to 10 mg/kg were tested in animals infected with 1 × LD50 of A/Vietnam/1194/2004 virus H5N1 virus (Figure 5A).

    A mortality rate of 80% was observed after 8 days in the control group (Figure 5A; group 5). A better survival rate was observed with doses of 5 mg/kg or more (groups 2 and 3). Treatment with doses at 10 mg/kg resulted in a 60% decrease in mortality compared with untreated animals (Figure 5A, group 3).

    When mice were challenge with 10 × LD50 of A/Vietnam/1194/2004 virus H5N1 virus (Figure 5B), the survival rate did significantly increase in comparison with untreated animals (group 5) at 14 days after virus challenge when a 5-day protocol designed at 20 mg/kg dose (group 2) was used. A 10% increase in survival was observed in animals treated with 10, 5 or 2.5 mg/kg of FBF001 and mortality was delayed in all treated groups (Figure 5B).

    In the animal groups challenged with 100 × LD50 of virus (Figure 5C), a 48-h delay in occurrence of the first death associated with a 10% mortality rate was obtained in groups that received 2.5 and 5 mg/kg. An increase in 30% of the survival rate combined with a 48-h delay in first death occurrence was obtained with a dose of 10 mg/kg (group 2).

    Discussion

    The HPAI H5N1 virus, which was first detected in 1997 in Asia, remains a major source of public health concern. Although the number of cases is limited on a global scale, H5N1 infection is consistently associated with a very high fatality rate of almost 59%. Only a few mutations in the receptor-binding site or a reassortment between H5N1 and a seasonal influenza virus could lead to the emergence of a new influenza strain with pandemic potential.

    Usually, the disease develops after a short incubation period and a peak of viremia is reached in only a few days [8]. The presence of virus in extrapulmonary tissues of some patients suggests that HPAI H5N1 virus has, as in animals, the potential to disseminate to other organs and tissues in humans [7]. Therefore, the use of post-exposure vaccination does not seem a suitable therapeutic option in exposed individuals. Additionally, current antiviral drugs such as oseltamivir have only a limited impact on mortality, even at twice the recommended dose, particularly if they are not administered during the very early phase of the disease [3]. It is therefore prudent to explore other therapeutic options.

    In clinical emergency situations, drugs must provide a broad spectrum of action and instant activity to stop, as quickly as possible, the progression of the infection. By its mechanism of action, passive immunotherapy has the advantage over the orally administrated drugs, thanks to an immediate effect on the virus. Some authors have proposed the development of monoclonal human or humanized antibodies against HPAI H5N1 [17,19,36]. However, despite encouraging results, this strategy presents major limitations, such as the possibility of emergence of escape mutants, the long research and development phase and specific process development that are incompatible with a clinical emergency, and the high cost of such treatment.

    By contrast, polyclonal immunoglobulins may offer many advantages in terms of cross-reactivity, rapidity and cost of development. Unlike antiviral drugs, they could provide specific and instant protection and may eventually reduce the risk of transmission through droplets if the correct epitopes are neutralized [37].

    Convalescent human plasma already demonstrated promising results in the treatment of H5N1 infections, most probably by decreasing the viral load [24]. A meta-analysis on the use of convalescent plasma during the 1918 influenza pandemic has shown that this strategy was effective in reducing mortality if administered early [38].

    Polyclonal immunoglobulins have been used for decades in the management of other clinical emergencies, such as snakebite envenomation or severe poisoning (e.g., tetanus toxin, botulin toxin, digoxin) [39]. Severe infectious diseases, such as rabies, are also treated by intramuscular injection of specific polyclonal immunoglobulins, in combination with classical anti-rabies vaccines [27].

    Equine rabies polyglobulin production methods were followed to produce these equine anti-H5N1 polyclonal immunoglobulins that consist of highly purified F(ab’)2, free of the Fc fragment, which holds the species determinant. While historically equine serum could be responsible for significant side effects, there is now an extensive history of use of purified equine polyglobulin fragments (F(ab’)2) demonstrating their efficacy and safety in humans, with no serious adverse reaction or serum sickness reported [26,40].

    Our data surprisingly demonstrate that the equine anti-H5N1 preparation, FBF001, not only has a high neutralization activity (titer of 1/6000) against the homologous A/Vietnam/1194/04 strain (Table 1), but also a very broad spectrum of neutralizing activity against heterologous strains from the same or different clades, while a previous study demonstrated only slight cross-neutralization activity of equine F(ab’)2 on heterologous strains [22]. This suggests that FBF001 may recognize a wide range of epitopes, including conserved epitopes, such as those located in regions like the stalk domain of the HA protein [41]. The protective efficacy of FBF001 was confirmed in vivo(Figure 3) even at concentrations lower than 0.1 µg (still 60% protection with 5 ng of FBF001), suggesting a potential indication for use as prophylactic treatment in persons recently exposed to the virus.

    Previous studies have already demonstrated the efficacy of equine anti-H5N1 F(ab’)2 in reducing viremia and mortality in mice [20,22]. The experiments we performed with FBF001 are the first proof of concept for efficacy of the product in mice that also allowed the design of a therapeutic protocol that could be eventually used in humans (Figures 4 & 5). In a post-exposure protocol a single injection 24 h after exposure was not sufficient to prevent the development of infection, and only led to delay by 48 h of the first case of death. This study demonstrates the necessity of a multiple injection protocol to manage HPAI H5N1 virus in infected mice. Previously published pharmacokinetic data indicated a half-life of 13.7 h after intravenous administration of 10 mg/kg of equine F(ab’)2 in mice [42]. Since the peak of viremia is reached approximately 5 days after H5N1 infection in humans [8], a 5-day protocol is justified to maintain the availability of adequate concentrations of neutralizing F(ab’)2 just before and during the peak of viremia in order to control the virus replication and reduce the risks of systemic spread. The use of a repeated dose protocol may not impact the efficacy and safety of the product, such as previously described for anti-SARS-CoV equine F(ab’)2 tested in macaques in a 4-week daily injection protocol [43].

    Two key observations in the mouse model may suggest a potential efficacy of FBF001 in humans: an increase in the survival rate observed in animals by day 14 after infection; and the induction of a delay in infection-related death (in patients a delay of a few days could give an additional time window for other treatments such as antiviral drugs to reach the tissues with optimal concentrations and for the patient to develop its own immune response).

    Additionally, although a regimen of 10 mg/kg of FBF001 administered daily for 5 consecutive days was sufficient to obtain an optimal efficacy (Figure 5), a good laboratory practice toxicity test performed in mice using seven repeated 100 mg/kg dose injections did not show any toxicity (data not shown), confirming the good safety potential of such product.

    Conclusion

    These results demonstrate a real potential of FBF001 (proposed trade name Fabenflu®, Fab’entech, Lyon, France) for the prophylaxis of individuals exposed to a wide range of HPAI H5N1 viruses, as well as for the treatment of infected patients, especially in association with antiviral drugs, a combined therapy that remains to be evaluated in the mouse model. The optimal therapeutic protocol developed in mice and consisting of five consecutive injections of 10 mg/kg or more of FBF001 on days 1–5 after viral challenge could easily be transposed into treatment for humans after clinical development of this product.

    Table 1.  Neutralization and hemagglutination inhibition titers measured with FBF001 on various isolates belonging to several clades and sub-clades of H5N1 virus.
    H5N1 virus cladeVirus strainGenbank accession number (HA segment)Average MN titer (SD)Average HI titer (SD)
    1A/Vietnam/1194/2004AY6513331/4000 (0)1/2560 (0)
    1A/chicken/Cambodia/022LC2b/2005HG6649451/4000 (0)1/960 (452)
    1A/Cambodia/duck/D14AL/2006HQ2004501/3000 (1414)1/960 (452)
    1.1.1A/duck/Cambodia/D3PV/2006HQ2004751/3000 (1414)1/1920 (905)
    1.1.1A/duck/Cambodia/67F8/2008JN5889441/4000 (2828)1/1920 (905)
    1.1.1A/Cambodia/R0405050/2007FJ2254721/2000 (0)1/1600 (1337)
    1.1.1A/chicken/Cambodia/TLC1/2009JN5888111/3000 (1414)1/960 (452)
    1.1.2A/duck/Cambodia/PV027D1/2010JN5888211/3000 (1414)1/1920 (905)
    1.1.2A/chicken/Cambodia/008LC1/2011JN5888241/4000 (2828)1/1280 (0)
    1.1.2A/Cambodia/W0526301/2012KF3692141/8000 (0)1/1920 (905)
    1.1.2A/Cambodia/X0125302/2013KF0013881/8000 (0)1/1280 (0)
    1.1.2A/chicken/Cambodia/X0124310/2013KF0014861/7000 (1414)1/1920 (905)
    2.1.3.2A/Indonesia/5/2005 (H5N1)-PR8-IBCDC-RG2EF5413941/5000 (1414)1/1920 (905)
    2.2A/bar-headed goose/Qinghai /1A/2005DQ1378731/5000 (1414)1/1920 (905)
    2.2.1A/Egypt/N03072/2010 (H5N1)-PR8-IDCDC-RG29CY0624841/4000 (0)1/1920 (905)
    2.3.2.1A/Hubei/1/2010 (H5N1)-PR8-IDCDC-RG30Cy0987581/1000 (0)1/1600 (1357)
    2.3.4A/chicken/Bangladesh/IIRS1984-30/2011JN7959241/1000 (0)1/960 (452)
    2.3.4A/Anhui/1/2005 (H5N1)-PR8-IBCDC-RG6HM1721041/6000 (0)1/1280 (0)
    4A/goose/Guiyang/1175/2006DQ9927711/8000 (0)1/1920 (905)
    7A/chicken/Shanxi/2/2006DQ9148141/8000 (0)1/1920 (905)
    9A/chicken/Henan/12/2004AY950232.11/8000 (0)1/1920 (905)

    The FBF001 solution (29.4 g/l) was diluted in Dulbecco Modified Eagle’s Medium to obtain final dilution of 1/20, 1/40, 1/80, 1/160, 1/320, 1/640, 1/1280, 1/2560, 1/5120 and 1/10,240. All these solutions were tested for MN and HI on 21 different HPAI virus (H5N1).

    HI: Hemagglutination inhibition; MN: Microneutralization.

    Executive summary

    • • Infection with highly pathogenic avian influenza A virus (H5N1) results in a high proportion of fatalities, with increasing evidence of resistance to antivirals.

    • • We prepared highly purified equine immunoglobulin F(ab’)2 fragments against H5N1.

    • • The final product was highly purified, consisting of almost 99% F(ab’)2 + Fab’, with no Fc fragments which elicit ‘serum sickness’.

    • • The F(ab’)2 preparation was tested in vitro for viral neutralization, and In vivo in BALB/c mice against challenge with H5N1 virus.

    • •  in vitro the F(ab’)2 preparation efficiently neutralized a broad panel of H5N1 isolates.

    • • In vivo the F(ab’)2 preparation protected mice against challenge with fatal doses of H5N1 virus, the most efficient protocol being five consecutive daily injections of 10 mg/kg.

    • • Administration of tenfold the effective dose of F(ab’)2 did not produce any toxicity in mice.

    • • Anti-H5N1 F(ab’)2 is a potentially important tool in the treatment of H5N1 infection.

    Acknowledgements

    The in vitro seroneutralization assays and hemagglutination inhibition assays were performed at the Institut Pasteur in Cambodia. We are extremely grateful to Li-Mei Chen and Ruben Donis (Influenza Division, CDC, GA, USA), Richard Webby (St Jude Hospital, TN, USA) and Jimmy Kwang (Temasek Life Sciences Laboratory, Singapore) for kindly providing some reference H5N1 strains used in this study. All in vivo experiments using pathogenic H5N1 influenza virus were performed in the Jean Mérieux BSL-4 laboratory in Lyon, France. We especially want to thank Delphine Pannetier, Aurélie Duthey and Stéphane Mély for their support. We also thank Keith Veitch for his support in final editorial reviewing.

    Financial & competing interests disclosure

    This research was partly financed by OSEO innovation. The authors declare a consultancy agreement between Fab’entech and Vincent Lotteau. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations.

    Papers of special note have been highlighted as: • of interest •• of considerable interest

    References

    • WHO. Cumulative number of confirmed human cases for avian influenza A(H5N1) reported to WHO, 2003–2013. www.who.int/influenza/human_animal_interface/EN_GIP_20140124CumulativeNumberH5N1cases.pdf?ua=1
    • Tran TH, Nguyen TL, Nguyen TD et al. Avian influenza A (H5N1) in 10 patients in Vietnam. N. Engl. J. Med.350(12),1179–1188 (2004).
    • Beigel JH, Farrar J, Han AM et al. Avian influenza A (H5N1) infection in humans. N. Engl. J. Med.353(13),1374–1385 (2005).
    • Nguyen TH, Farrar J, Horby P. Person-to-person transmission of influenza A (H5N1). Lancet371(9622),1392–1394 (2008).
    • Buchy P, Mardy S, Vong S et al. Influenza A/H5N1 virus infection in humans in Cambodia. J. Clin. Virol.39(3),164–168 (2007).
    • Katz JM, Lim W, Bridges CB et al. Antibody response in individuals infected with avian influenza A (H5N1) viruses and detection of anti-H5 antibody among household and social contacts. J. Infect. Dis.180(6),1763–1770 (1999).
    • Buchy P, Vong S, Chu S et al. Kinetics of neutralizing antibodies in patients naturally infected by H5N1 virus. PLoS One5(5),e10864 (2010).• Study about the kinetics of anti-H5 neutralizing antibodies in naturally H5N1-infected patients with severe clinical illness or asymptomatic infection.
    • Gambotto A, Barratt-Boyes SM, De Jong MD, Neumann G, Kawaoka Y. Human infection with highly pathogenic H5N1 influenza virus. Lancet371(9622),1464–1475 (2008).• Comprehensive review on H5N1 physiopathology in human.
    • Peiris JS, Cheung CY, Leung CY, Nicholls JM. Innate immune responses to influenza A H5N1: friend or foe? Trends Immunol.30(12),574–584 (2009).
    • 10  Basu A, Antanasijevic A, Wang M et al. New small molecule entry inhibitors targeting hemagglutinin-mediated influenza A virus fusion. J. Virol.88(3),1447–1460 (2014).
    • 11  Furuta Y, Gowen BB, Takahashi K, Shiraki K, Smee DF, Barnard DL. Favipiravir (T-705), a novel viral RNA polymerase inhibitor. Antiviral Res.100(2),446–454 (2013).
    • 12  Naughtin M, Dyason JC, Mardy S, Sorn S, Von Itzstein M, Buchy P. Neuraminidase inhibitor sensitivity and receptor-binding specificity of Cambodian clade 1 highly pathogenic H5N1 influenza virus. Antimicrob. Agents Chemother.55(5),2004–2010 (2011).
    • 13  Mckimm-Breschkin JL, Barrett S, Pudjiatmoko et al. I222 Neuraminidase mutations further reduce oseltamivir susceptibility of Indonesian Clade 2.1 highly pathogenic avian influenza A(H5N1) viruses. PLoS One8(6),e66105 (2013).
    • 14  Mckimm-Breschkin JL, Selleck PW, Usman TB, Johnson MA. Reduced sensitivity of influenza A (H5N1) to oseltamivir. Emerg. Infect. Dis.13(9),1354–1357 (2007).
    • 15  De Jong MD, Bach VC, Phan TQ et al. Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by coma. N. Engl. J. Med.352(7),686–691 (2005).
    • 16  Le QM, Kiso M, Someya K et al. Avian flu: isolation of drug-resistant H5N1 virus. Nature437(7062),1108 (2005).
    • 17  Hanson BJ, Boon AC, Lim AP, Webb A, Ooi EE, Webby RJ. Passive immunoprophylaxis and therapy with humanized monoclonal antibody specific for influenza A H5 hemagglutinin in mice. Respir. Res.7,126 (2006).
    • 18  Prabhu N, Prabakaran M, Hongliang Q et al. Prophylactic and therapeutic efficacy of a chimeric monoclonal antibody specific for H5 haemagglutinin against lethal H5N1 influenza. Antivir. Ther.14(7),911–921 (2009).
    • 19  Sakabe S, Iwatsuki-Horimoto K, Horimoto T et al. A cross-reactive neutralizing monoclonal antibody protects mice from H5N1 and pandemic (H1N1) 2009 virus infection. Antiviral Res.88(3),249–255 (2010).
    • 20  Lu J, Guo Z, Pan X et al. Passive immunotherapy for influenza A H5N1 virus infection with equine hyperimmune globulin F(ab’)2 in mice. Respir. Res.7,43 (2006).
    • 21  Nguyen HH, Tumpey TM, Park HJ et al. Prophylactic and therapeutic efficacy of avian antibodies against influenza virus H5N1 and H1N1 in mice. PLoS One5(4),e10152 (2010).
    • 22  Zhao Z, Yan F, Chen Z et al. Cross clade prophylactic and therapeutic efficacy of polyvalent equine immunoglobulin F(ab’)2 against highly pathogenic avian influenza H5N1 in mice. Int. Immunopharmacol.11(12),2000–2006 (2011).
    • 23  Kong LK, Zhou BP. Successful treatment of avian influenza with convalescent plasma. Hong Kong Med. J.12(6),489 (2006).
    • 24  Zhou B, Zhong N, Guan Y. Treatment with convalescent plasma for influenza A (H5N1) infection. N. Engl. J. Med.357(14),1450–1451 (2007).•• Provides interesting clinical data on the use of convalescent plasma to treat H5N1 patients.
    • 25  Gutierrez RA, Naughtin MJ, Horm SV, San S, Buchy P. A(H5N1) virus evolution in South East Asia. Viruses1(3),335–361 (2009).
    • 26  Lang J, Attanath P, Quiambao B et al. Evaluation of the safety, immunogenicity, and pharmacokinetic profile of a new, highly purified, heat-treated equine rabies immunoglobulin, administered either alone or in association with a purified, Vero-cell rabies vaccine. Acta Trop.70(3),317–333 (1998).
    • 27  Quiambao BP, Dytioco HZ, Dizon RM, Crisostomo ME, Laot TM, Teuwen DE. Rabies post-exposure prophylaxis in the Philippines: health status of patients having received purified equine F(ab’)2 fragment rabies immunoglobulin (Favirab). PLoS Negl. Trop. Dis.2(5),e243 (2008).
    • 28  Chippaux JP, Lang J, Amadi-Eddine S, Fagot P, Le Mener V. Short report: treatment of snake envenomations by a new polyvalent antivenom composed of highly purified F(ab)2: results of a clinical trial in northern Cameroon. Am. J. Trop. Med. Hyg.61(6),1017–1018 (1999).
    • 29  Chippaux JP, Lang J, Eddine SA et al. Clinical safety of a polyvalent F(ab’)2 equine antivenom in 223 African snake envenomations: a field trial in Cameroon. VAO (Venin Afrique de l’Ouest) Investigators. Trans. R Soc. Trop. Med. Hyg.92(6),657–662 (1998).
    • 30  Chippaux JP, Massougbodji A, Stock RP, Alagon A. Clinical trial of an F(ab’)2 polyvalent equine antivenom for African snake bites in Benin. Am. J. Trop. Med. Hyg.77(3),538–546 (2007).
    • 31  Nguyen L. Production d’immunoglobulines thérapeutiques hautement purifiées (ITHP): analyse d’un procédé de purification. Biologie Aujourd’hui204,55–59 (2010).
    • 32  Grandgeorge M, Véron JL, Lutsch C et al. Preparation of improved F(ab’)2 antivenoms. An example: new polyvalent European viper antivenom (equine). In: Envenomings and Their Treatments. Fondation Mérieux, France, 161–172 (1996).
    • 33  Reed L, Muench H. A simple method of estimating fifty percent endpoints. Am. J. Hyg.27,493–497 (1938).
    • 34  Rowe T, Abernathy RA, Hu-Primmer J et al. Detection of antibody to avian influenza A (H5N1) virus in human serum by using a combination of serologic assays. J. Clin. Microbiol.37(4),937–943 (1999).
    • 35  Lu X, Tumpey TM, Morken T, Zaki SR, Cox NJ, Katz JM. A mouse model for the evaluation of pathogenesis and immunity to influenza A (H5N1) viruses isolated from humans. J. Virol.73(7),5903–5911 (1999).
    • 36  Masalova OV, Klimova RR, Chichev EV et al. Development of monoclonal antibodies to highly pathogenic avian influenza H5N1 virus and their application to diagnostics, prophylaxis, and therapy. Acta Virol.55(1),3–14 (2011).
    • 37  Thornburg NJ, Nannemann DP, Blum DL et al. Human antibodies that neutralize respiratory droplet transmissible H5N1 influenza viruses. J. Clin. Invest.123(10),4405–4409 (2013).
    • 38  Luke TC, Kilbane EM, Jackson JL, Hoffman SL. Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment? Ann. Intern. Med.145(8),599–609 (2006).•• Meta-analysis of studies from the Spanish influenza era reported that transfusion of influenza-convalescent human blood products reduced mortality in patients with influenza complicated by pneumonia.
    • 39  Chippaux JP, Goyffon M. Venoms, antivenoms and immunotherapy. Toxicon36(6),823–846 (1998).• Interesting review on envenomation and the use of immunotherapy in humans.
    • 40  De Haro L, Lang J, Bedry R et al. [Snake bite by European vipers. A multicenter study of tolerance to Viperfav, a new intravenous antivenom]. Ann. Fr. Anesth. Reanim.17(7),681–687 (1998).
    • 41  Steel J, Lowen AC, Wang TT et al. Influenza virus vaccine based on the conserved hemagglutinin stalk domain. MBio1(1),e00018-10 (2010).
    • 42  Bazin-Redureau M, Pepin S, Hong G, Debray M, Scherrmann JM. Interspecies scaling of clearance and volume of distribution for horse antivenom F(ab’)2. Toxicol. Appl. Pharmacol.150(2),295–300 (1998).•• An allometric approach among three mammalian species to predict F(ab’)2 pharmacokinetic parameters in humans.
    • 43  Xu Y, Jia Z, Zhou L et al. Evaluation of the safety, immunogenicity and pharmacokinetics of equine anti-SARS-CoV F(ab’)(2) in macaque. Int. Immunopharmacol.7(13),1834–1840 (2007).