Preserved antiviral adaptive immunity following polyclonal antibody ...

1 downloads 0 Views 2MB Size Report
Jul 6, 2016 - hyperimmune intravenous immunoglobulin (IVIG) prepared from donors exposed to pandemic H1N1 influ- enza significantly reduced viral load ...
www.nature.com/scientificreports

OPEN

received: 11 November 2015 accepted: 15 June 2016 Published: 06 July 2016

Preserved antiviral adaptive immunity following polyclonal antibody immunotherapy for severe murine influenza infection Natalie E. Stevens1, Antoinette Hatjopolous1, Cara K. Fraser2, Mohammed Alsharifi3, Kerrilyn R. Diener1,4 & John D. Hayball1,4 Passive immunotherapy may have particular benefits for the treatment of severe influenza infection in at-risk populations, however little is known of the impact of passive immunotherapy on the formation of memory responses to the virus. Ideally, passive immunotherapy should attenuate the severity of infection while still allowing the formation of adaptive responses to confer protection from future exposure. In this study, we sought to determine if administration of influenza-specific ovine polyclonal antibodies could inhibit adaptive immune responses in a murine model of lethal influenza infection. Ovine polyclonal antibodies generated against recombinant PR8 (H1N1) hemagglutinin exhibited potent prophylactic capacity and reduced lethality in an established influenza infection, particularly when administered intranasally. Surviving mice were also protected against reinfection and generated normal antibody and cytotoxic T lymphocyte responses to the virus. The longevity of ovine polyclonal antibodies was explored with a half-life of over two weeks following a single antibody administration. These findings support the development of an ovine passive polyclonal antibody therapy for treatment of severe influenza infection which does not affect the formation of subsequent acquired immunity to the virus. Influenza infection claims approximately 250,000 lives worldwide annually and remains a significant burden on public health systems1. The majority of deaths from influenza infection occur in at-risk populations which include the immunocompromised, infants and the elderly, all which exhibit suboptimal responses to vaccination2,3 and are at higher risk of ‘severe’ influenza infection4. Severe influenza is defined as influenza infection accompanied by complications that require hospitalisation, and is attributed to over 3.4% of all critical hospitalisations during influenza season1. Current treatment for severe influenza generally incorporates supportive care and antiviral medications such as oseltamivir (TamifluTM) or zanamivir (RelenzaTM)5, however viral resistance to such medications is increasing, and is most often acquired by infective viruses during hospitalisation and treatment6, which is a significant risk for immunocompromised patients hospitalised for extended durations7. Vaccination is the most widely-used strategy to combat the morbidity and economic burden of influenza, with severe infection generally associated with failure to vaccinate8 or reduced efficacy of vaccination in immunocompromised populations2,9. Effective seasonal influenza vaccines elicit neutralising antibodies (Abs) against strain-specific glycoproteins, in particular haemagglutinin (HA) which is considered the major neutralising determinant of influenza10. However differences between the predicted HA incorporated into seasonal vaccines and the actual HA expressed by circulating strains can impact vaccine efficacy resulting in increased transmission and a higher burden of severe influenza infections3,11. It is clear therefore, that strategies beyond seasonal

1 Experimental Therapeutics Laboratory, Hanson Institute, and Sansom Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, Australia. 2Preclinical, Imaging and Research Laboratories, South Australian Health and Medical Research Institute, Gilles Plains, Adelaide, SA, Australia. 3Vaccine Research Group, Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia. 4Robinson Research Institute, Discipline of Obstetrics and Gynaecology, School of Medicine, The University of Adelaide, Adelaide, SA, Australia. Correspondence and requests for materials should be addressed to K.R.D. (email: [email protected]) or J.D.H. (email: [email protected])

Scientific Reports | 6:29154 | DOI: 10.1038/srep29154

1

www.nature.com/scientificreports/ vaccination and antiviral medications should be explored to combat the morbidity and mortality of influenza infection, particularly during critical hospitalisation periods12. Passive immunotherapy using neutralising Ab may be the ideal rapid treatment strategy for influenza infection that functions independently of the individual’s immunocompetency12 as highlighted by previous animal and human studies13–15. Furthermore, a recent multicentre double-blind randomised controlled trial has found that hyperimmune intravenous immunoglobulin (IVIG) prepared from donors exposed to pandemic H1N1 influenza significantly reduced viral load in infected individuals compared to IVIG prepared from donations received before the 2009 pandemic16. In a similar manner, further studies have shown reduced mortality in patients treated with convalescent plasma17. Animal studies have shown efficacy of anti-influenza enriched IVIG preparations in protecting immunodeficient mice from morbidity and mortality following influenza infection18, which indicates that this may be an effective strategy for use in immunocompromised patients. Despite clinical efficacy, the use of human-derived Ab therapies have significant logistical challenges including lengthy time periods for identification and screening of potential serum donors, and validation of sera and subsequent Ab products19. Consequently, current guidelines do not recommend IVIG or convalescent sera as a therapy for severe influenza5. Alternatively, passive immunotherapies utilising humanised monoclonal Abs (mAbs) have been developed for influenza prophylaxis and treatment, and whilst mAbs targeting HA have potent neutralising capacity, they are often not cross-reactive against multiple strains and may prompt the development of resistant mutants20. In contrast, mAb strategies that target highly conserved residues on the M2 ion channel protein are cross-reactive and demonstrate in vivo efficacy in experimental models21 however these are generally thought to function through the instigation of antibody-mediated cell cytotoxicity rather than direct viral neutralisation20,22. As such, the efficacy of these mAbs in immunocompromised individuals may be reduced. Polyclonal Abs (pAbs) can overcome these deficiencies by their intrinsic ability to target multiple epitopes, which can increase cross-reactivity and reduce the development of resistant strains23,24. Approaches using pAbs sourced from animal sera are advantageous as large amounts of pAb can be collected and batched for reduced variability. Furthermore, pAbs have simplified screening procedures compared to human-derived Abs. Ovine pAbs are particularly efficient and cost-effective to produce, and exhibit reduced immunogenicity compared to those sourced from horses or other large mammals25. Though ovine-origin products carry a risk of contamination with transmissible prion proteins, sourcing of ovine serum from countries free from known prion disease such as Australia and New Zealand can remove this risk, and thus commercially available ovine pAb products are commonly sourced from regions such as these. Ovine pAb fragments have been used with success in the clinic, and are currently available as critical care therapies for life-threatening digoxin toxicity (DigiFab ​, DIGIBIND ​)26 and crotalid snake envenomation (CroFab ​)27. We have previously shown that ovine serum pAbs elicited against HA have potent neutralising capacity in vitro and are protective in an in vivo murine influenza model13. However, any Ab administered to prevent or treat influenza must not prevent the formation of adaptive immunity necessary for enduring protection against reinfection with future circulating strains, and this has not been explored in the context of foreign pAb delivery. Therefore the aims of this study were to explore the role of ovine anti-HA pAb therapy in a mouse model of severe influenza and to assess the effect on subsequent influenza-specific adaptive responses. It was demonstrated that ovine purified pAb treatment can effectively reduce mortality in the murine model and does not impede the development of protective humoral or cell-mediated responses to influenza.







Methods

Animals.  Female 6–8 week old BALB/c mice were purchased from Laboratory Animal Services, The

University of Adelaide and housed in IVCs under specific pathogen-free facility conditions with food and water provided ad libitum. All animal experiments were approved by the University of South Australia Animal Ethics Committee, and conducted in accordance with National and Institutional ethical and regulatory guidelines.

Purification of ovine polyclonal anti-HA from serum.  Sheep were immunised with baculovirus-produced recombinant HA (rHA) derived from PR8 influenza with serum stored at −​20 °C as previously described13. Thawed serum was filtered through glass wool and pAb precipitated by addition of saturated ammonium sulfate solution to 45% (v/v). After centrifugation (20 min, 10,000 ×​ g) pelleted protein precipitate was dissolved in milliQ water and dialysed into PBS before application to a Protein G agarose column (Thermo Fisher Scientific). Bound IgG was eluted with 0.1 M glycine (pH 2.5) and dialysed into PBS before concentration determination via BCA assay (Thermo Fisher Scientific) according to manufacturer’s instructions. Concentrated IgG was subsequently analysed for purity via SDS-PAGE. Control serum was sourced from non-immunised sheep and total IgG purified as described. Murine model of influenza infection.  Groups of mice were administered purified pAbs (25 mg/kg) or PBS either via intraperitoneal (IP) injection (500 μ​L) or intranasal (IN) inhalation (32 μ​L) under anaesthetic. This dose was selected empirically from a pilot study (data not shown). For severe influenza infection, mice received IN challenge (32 μ​L) with PR8 H1N1 (500 TCID50) or A/PC H3N2 (3000 TCID50) influenza virus that had been purified from hen egg amniotic fluid as previously described28. Viral inocula were diluted on the day of use from freshly thawed viral stocks and untreated controls were run in parallel with all treated groups to ensure that the dose of virus administered caused an infection requiring euthanasia within 6–10 days. All challenged mice were provided with soaked food and sunflower seeds to support them through the associated pain and distress of infection, and were assessed daily for clinical symptoms of infection which included weight loss, ruffled coat, wheezing or abnormal respiration, hunched posture and reluctance to move. A clinical score of 5 or 20% weight loss was used to identify those mice reaching predetermined endpoints requiring euthanasia. Weight loss was the primary indicator for euthanasia in this study as most mice did not reach clinical thresholds. Scientific Reports | 6:29154 | DOI: 10.1038/srep29154

2

www.nature.com/scientificreports/

Detected Analyte

Sample

Coating protein

Coating Concentration

Detection Ab

Concentration Depicted in

500 TCID50/mL in HRP-linked rabbit anti-murine 100 mM NaHCO3 IgG (Sigma; Cat #A9044)

1:10,000 in 1% BSA PBS-T

Figure 3c

Purified donkey antiOvine IgG in circulation Murine sera sheep IgG (Sigma; Cat # SAB3700721)

5 μ​g/mL in 100 mM NaHCO3

HRP-linked anti-sheep IgG (Sigma; Cat # A3415)

1:15,000 in 1% BAS PBS-T

Figure 4a

Murine anti-ovine IgG

Murine sera

Ovine pAb

10 μ​g/mL in 100 mM NaHCO3

HRP-linked rabbit anti-murine IgG (Sigma; Cat # A9044)

1:10,000 in 1% BSA PBS-T

Figure 4b

Ovine anti-PR8 IgG

Ovine pAb

PR8 virus

500 TCID50/mL in 100 mM NaHCO3

HRP-linked anti-sheep IgG (Sigma; Cat # A3415)

1:15,000 in 1% BAS PBS-T

Figure 5a

Ovine anti-A/PC IgG

Ovine pAb

A/PC virus

500 TCID50/mL in 100 mM NaHCO3

HRP-linked anti-sheep IgG (Sigma; Cat # A3415)

1:15,000 in 1% BAS PBS-T

Figure 5a

Murine anti-PR8 IgG

Murine sera

PR8 virus

Table 1.  Antibodies used for ELISA assays. ELISA protocols were developed to assess the levels of murine anti-PR8 IgG, circulating ovine IgG or murine anti-ovine IgG in sera from infected or treated mice, or to assess the ability of ovine IgG to bind to PR8 and A/PC viruses.

Haemagglutination-inhibition assay (HAI).  All HAI assays were performed as previously described13.

Briefly, purified pAb (500 μ​g/mL; 30  μ​L) was pipetted into duplicate wells of a round-bottom 96-well plate and serially diluted two-fold in PBS before the addition of either PR8 influenza virus (5 haemagglutination (HA) units in 30 μ​L) to all wells. After 30 minutes incubation at room temperature, 0.5% (v/v) chicken red blood cells (cRBC) in PBS (30 μ​L) was added to each well and gently mixed. Plates were visualised over a light box after 45 minutes. The endpoint HAI titre was recorded as the highest dilution of test sample that was able to completely inhibit the agglutination of cRBC by virus in duplicate wells.

ELISA analysis.  Specific ELISA methods were employed to measure binding of anti-HA Abs against PR8

or A/PC virus and to quantify circulating anti-influenza Abs, anti-ovine Abs or ovine pAbs in mouse sera. Combinations of coating and detection Ab are summarised in Table 1. Briefly, EIA/RIA high-binding ELISA plates (Corning) were coated with 100 μ​L of the relevant coating protein diluted in 100 mM NaHCO3 buffer (pH9) and incubated overnight at 4 °C. Plates were blocked with 2% (w/v) BSA in PBS (1 hour, 37 °C) and the relevant samples diluted 1:2 for Ab quantification, or serially diluted for endpoint analysis in 1% (w/v) BSA in PBS with 0.05% Tween (PBS-T). After 3 washes with PBS-T (200 μ​L), diluted samples or serially diluted ovine pAb standards (32,000–2 ng/mL in PBS-T) were added to duplicate wells (100 μ​L) before incubation (2 hour, 37 °C). Plates were washed as before and bound serum Abs were detected by addition of the relevant detection Ab (100 μ​L) followed by incubation for 1 hour (37 °C). The plates were subsequently developed with OPD substrate (Sigma; 100 μ​L), the reaction stopped with 3 M HCl (50 μ​L), and the absorbance read at 490 nm. For endpoint ELISA methods, endpoint absorbance values were defined as the mean absorbance plus two standard deviations of negative control samples diluted 1:100.

In vivo cytotoxic lymphocyte (CTL) assay.  The ability of mice to mount a cell-mediated response to influ-

enza infection was assessed using an in vivo cytotoxicity assay. Splenocytes from a donor mouse were harvested and RBC subsequently lysed. Washed splenocytes were pulsed with Kd-restricted influenza nucleoprotein-derived peptide TYQRTRALV (NPP) before two washes in endotoxin-free PBS followed by CellTrace ​Far Red staining (0.1 μ​M) for 20 minutes at 37 °C. Cells were subjected to two more washes and mixed at a 1:1 ratio with unpulsed control cells stained with CFSE (3 μ​M; 10 minutes at 37 °C). Optimal staining was confirmed by flow cytometry (BD FACSCanto II) before transfer into infected or control mice via tail vein injection (8–10 million cells per mouse). Spleens from recipient mice were harvested 18 hours later and single cell preparations were assessed for percentage of FarRed and CFSE cells remaining via flow cytometry and FACSDiva software. Percent cell specific lysis was calculated using the following equation: % specific lysis =​  [1−(infected mouse %CFSEpos: %FarRedpos/ control mouse %CFSEpos: %FarRedpos)] × 100.



Statistical analyses.  Statistical analyses were performed using GraphPad Prism V5 software. Survival

curves were compared using the Mantel-Cox (log-rank) test. ELISA endpoints and in vivo CTL data were compared using two-tailed unpaired T-tests. Statistical significance was defined as P ≤​  0.05.

Results

Prophylactic administration of ovine anti-HA pAbs protects mice against lethal influenza infection. 

Previous studies have established the neutralisation capacity of generated whole ovine anti-HA antisera in vitro and in vivo13, however it remained necessary to confirm the potent neutralising capacity of the now purified IgG in vivo. To assess this, groups of mice were intraperitoneally administered purified ovine anti-HA pAbs, control pAbs, or PBS, and twenty-four hours later challenged intranasally with a lethal dose of PR8 influenza. Mice that had received anti-HA pAbs exhibited reduced weight loss (Fig. 1a) and mild signs of infection from days 2–6 (Supplementary Figure S1) and variable weight loss after day 7. However, all resumed weight gain after day 11, and none required euthanasia. In contrast, administration of control ovine pAbs or PBS could not control influenza infection which led to rapid weight loss from day 3 and required euthanasia by day 6 (Fig. 1b,c respectively). Thus a statistically significant survival advantage was conferred to the group of mice that received anti-HA ovine

Scientific Reports | 6:29154 | DOI: 10.1038/srep29154

3

www.nature.com/scientificreports/

Figure 1.  Administration of ovine anti-HA pAbs protects against subsequent lethal influenza infection. Mice were prophylactically administered (25 mg/kg in 500 μ​L) ovine anti-HA pAbs (a), control pAbs (b) or PBS (c) via the intraperitoneal route. Twenty-four hours later mice were challenged with 500 TCID50 (32 μ​L) PR8 influenza. The mice were closely monitored for weight loss (a–c) and euthanized according to predetermined humane endpoints (indicated by arrowheads). Data is presented as the weight loss of individual mice from infection day (n =​ 5). Corresponding survival curves (d) were analysed using the Mantel-Cox test to compare anti-HA treated to PBS treated and control treated mice. Significance is denoted as thus: *​*​  =​  P