Human vaccination against Plasmodium vivax Duffy-binding protein induces strain-transcending antibodies

Ruth O Payne, Sarah E Silk, Sean C Elias, Kathryn H Milne, Thomas A Rawlinson, David Llewellyn, A Rushdi Shakri, Jing Jin, Geneviève M Labbé, Nick J Edwards, Ian D Poulton, Rachel Roberts, Ryan Farid, Thomas Jørgensen, Daniel Gw Alanine, Simone C de Cassan, Matthew K Higgins, Thomas D Otto, James S McCarthy, Willem A de Jongh, Alfredo Nicosia, Sarah Moyle, Adrian Vs Hill, Eleanor Berrie, Chetan E Chitnis, Alison M Lawrie, Simon J Draper, Ruth O Payne, Sarah E Silk, Sean C Elias, Kathryn H Milne, Thomas A Rawlinson, David Llewellyn, A Rushdi Shakri, Jing Jin, Geneviève M Labbé, Nick J Edwards, Ian D Poulton, Rachel Roberts, Ryan Farid, Thomas Jørgensen, Daniel Gw Alanine, Simone C de Cassan, Matthew K Higgins, Thomas D Otto, James S McCarthy, Willem A de Jongh, Alfredo Nicosia, Sarah Moyle, Adrian Vs Hill, Eleanor Berrie, Chetan E Chitnis, Alison M Lawrie, Simon J Draper

Abstract

Background: Plasmodium vivax is the most widespread human malaria geographically; however, no effective vaccine exists. Red blood cell invasion by the P. vivax merozoite depends on an interaction between the Duffy antigen receptor for chemokines (DARC) and region II of the parasite's Duffy-binding protein (PvDBP_RII). Naturally acquired binding-inhibitory antibodies against this interaction associate with clinical immunity, but it is unknown whether these responses can be induced by human vaccination.

Methods: Safety and immunogenicity of replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and modified vaccinia virus Ankara (MVA) viral vectored vaccines targeting PvDBP_RII (Salvador I strain) were assessed in an open-label dose-escalation phase Ia study in 24 healthy UK adults. Vaccines were delivered by the intramuscular route in a ChAd63-MVA heterologous prime-boost regimen using an 8-week interval.

Results: Both vaccines were well tolerated and demonstrated a favorable safety profile in malaria-naive adults. PvDBP_RII-specific ex-vivo IFN-γ T cell, antibody-secreting cell, memory B cell, and serum IgG responses were observed after the MVA boost immunization. Vaccine-induced antibodies inhibited the binding of vaccine homologous and heterologous variants of recombinant PvDBP_RII to the DARC receptor, with median 50% binding-inhibition titers greater than 1:100.

Conclusion: We have demonstrated for the first time to our knowledge that strain-transcending antibodies can be induced against the PvDBP_RII antigen by vaccination in humans. These vaccine candidates warrant further clinical evaluation of efficacy against the blood-stage P. vivax parasite.

Trial registration: Clinicaltrials.gov NCT01816113.

Funding: Support was provided by the UK Medical Research Council, UK National Institute of Health Research Oxford Biomedical Research Centre, and the Wellcome Trust.

Keywords: Infectious disease; Vaccines.

Conflict of interest statement

Conflict of interest: S.C. de Cassan, M.K. Higgins, A.V.S. Hill, and S.J. Draper are named inventors on patent applications (patent nos. GB1413530.5, GB1016471.3, and WO/2008/122811) covering malaria vaccines and immunization regimens. A. Nicosia was an employee of and shareholder in Okairòs (since acquired by GlaxoSmithKline), which is developing vectored vaccines for a number of diseases. T. Jørgensen and W.A. de Jongh are employees of, and W.A. de Jongh is a shareholder in, ExpreS2ion Biotechnologies, which has developed and is marketing the ExpreS2 cell expression platform. C.E. Chitnis is a named inventor on a patent covering PvDBP_RII (patent no. WO/1996/040766).

Figures

Figure 1. VAC051 flow chart of study…
Figure 1. VAC051 flow chart of study design and volunteer recruitment.
Recruitment for the VAC051 study took place between May 2013 and February 2014. The final follow-up visit took place in July 2014. All immunizations were administered intramuscularly, with sequential vaccines administered into the deltoid of alternating arms.
Figure 2. Solicited AEs following vaccination with…
Figure 2. Solicited AEs following vaccination with ChAd63 and MVA PvDBP_RII.
The solicited local and systemic adverse events (AEs) recorded for 14 days following ChAd63 PvDBP_RII and for 7 days following MVA PvDBP_RII are shown at the maximum severity reported by all volunteers. (A) Four volunteers received 5 × 109 viral particles (vp) ChAd63 PvDBP_RII (group 1), and (B) 20 received 5 × 1010 vp (group 2). (C) Seven of the group 2 volunteers went on to receive MVA PvDBP_RII 1 × 108 PFU (group 2B), and (D) 8 received 2 × 108 PFU (group 2C). ChAd63, replication-deficient chimpanzee adenovirus serotype 63; MVA, modified vaccinia virus Ankara; PvDBP_RII, region II of the P. vivax Duffy-binding protein.
Figure 3. Ex-vivo IFN-γ T cell response…
Figure 3. Ex-vivo IFN-γ T cell response to vaccination.
(A) Median ex vivo IFN-γ ELISPOT responses in peripheral blood mononuclear cells (PBMCs) to the PvDBP_RII insert (summed response across all the individual peptide pools) shown for all groups. Individual responses are shown in Supplemental Figure 1. Median and individual responses are shown at (B) day 14, (C) day 63, and (D) day 140. Symbols are coded according to group. *P < 0.05. Responses between groups 1 (n = 4) and 2 (n = 20) at day 14, and between groups 2B (n = 7) and 2C (n = 8) at day 140 were assessed by Mann-Whitney test (B and D); responses between groups 2A (n = 4), 2B (n = 7), and 2C (n = 8) at day 63 were assessed by Kruskal-Wallis test with Dunn’s multiple comparison test (C). SFU, spot-forming units; PvDBP_RII, region II of the P. vivax Duffy-binding protein.
Figure 4. Serum antibody response to vaccination.
Figure 4. Serum antibody response to vaccination.
(A) Median anti–PvDBP_RII serum total IgG responses shown for all groups over time. Individual responses are shown in Supplemental Figure 3. Median and individual responses are shown at (B) day 28, (C) day 84, and (D) day 140. The horizontal dotted line indicates the limit of detection of the assay. (E) Isotype profiles of serum antibody responses were assessed by ELISA. Responses are shown at baseline (d0) and for all groups at day 84. Individual and median responses are shown for IgG1 and IgG3; results for IgG2, IgG4, IgA, and IgM are shown in Supplemental Figure 6. (F) Avidity of serum IgG responses at day 84 was assessed by NaSCN-displacement PvDBP_RII ELISA and is reported as the molar (M) concentration of NaSCN required to reduce the starting OD in the ELISA by 50% (IC50). Symbols are coded according to group. *P < 0.05, **P < 0.01. Responses in groups 2A (n = 4), 2B (n = 7), and 2C (n = 8) were assessed by Kruskal-Wallis test with Dunn’s multiple comparison test; responses between groups 2B and 2C were assessed by Mann-Whitney test (C). PvDBP_RII, region II of the P. vivax Duffy-binding protein.
Figure 5. B cell response to vaccination.
Figure 5. B cell response to vaccination.
(A) PvDBP_RII–specific antibody-secreting cell (ASC) responses were assessed by ex-vivo ELISPOT using PvDBP_RII protein and frozen peripheral blood mononuclear cells (PBMCs) from the day 63 time point. Individual and median responses are shown for each group and reported as PvDBP_RII–specific ASCs per million PBMCs used in the assay. (B) Correlation of the ASC response versus the concentration of serum anti–PvDBP_RII IgG measured at day 84. (C) PvDBP_RII–specific memory B cell (mBC) responses were assessed by ELISPOT assay using PvDBP_RII protein. Frozen PBMCs were thawed and underwent a 6-day polyclonal restimulation during which ASCs were derived from mBCs, before testing in the assay. Individual and median responses are shown from the day 84 time point and are reported as mBC-derived PvDBP_RII–specific ASCs per million cultured PBMCs or as (D) percentage of total number of IgG-secreting ASCs. (E and F) Correlations of the mBC response versus the concentration of serum anti–PvDBP_RII IgG at day 84. For all correlations, Spearman’s rank correlation coefficient (rs) and P value are shown. *P < 0.05. Responses between groups 2B (n = 7) and 2C (n = 8) were assessed by Mann-Whitney test. PvDBP_RII, region II of the P. vivax Duffy-binding protein.
Figure 6. PvDBP_RII–DARC in vitro binding inhibition.
Figure 6. PvDBP_RII–DARC in vitro binding inhibition.
(A) Day 84 sera from volunteers in groups 1 (n = 4), 2A (n = 4), 2B (n = 7), and 2C (n = 8) were tested for their ability to inhibit binding of recombinant PvDBP_RII (SalI) to the Duffy antigen receptor for chemokines (DARC) using an ELISA-based assay in Oxford. Samples were titrated starting at 1:5 dilution down to 1:640 (Supplemental Figure 7A). Data show the interpolated dilution for each sample that gave 50% binding inhibition. (B) For positive samples in A (n = 16), the concentration of anti–PvDBP_RII (SalI) serum IgG that gives 50% binding inhibition (EC50) was calculated by dividing the serum ELISA μg/ml by the 50% binding-inhibition serum titer. The result is reported in ng/ml. (CF) Day 0 and day 84 sera were assessed as in A using the assay established at ICGEB, India, using 4 recombinant alleles of PvDBP_RII: SalI, PvAH, PvO, and PvP. In all panels, the individual and median results are shown for each group. The dashed line shows an arbitrary cutoff below which negative samples are plotted. PvDBP_RII, region II of the P. vivax Duffy-binding protein; SalI, Salvador I reference strain.
Figure 7. Binding inhibition of the P.…
Figure 7. Binding inhibition of the P. vivax HMP013 strain DBP_RII.
(A) The location of polymorphic residues in PvDBP_RII (HMP013 strain) have been marked on a structure of the PvDBP_RII (SalI strain) dimer bound to the Duffy antigen receptor for chemokines (DARC) aa 19–30 (PDB code 4NVU) (24). Two views of the dimer are shown, rotated by 90 degrees around the horizontal axis. One molecule of PvDBP_RII is shown in gray surface representation with polymorphic residues colored in red. The second molecule of PvDBP_RII is in blue cartoon representation with SD3 in a darker blue. The 2 helices from DARC are shown in green and cyan, respectively. (B) Day 84 sera from volunteers in groups 1 (n = 4), 2A (n = 4), 2B (n = 7), and 2C (n = 8) were tested for their ability to inhibit binding of recombinant PvDBP_RII (HMP013) to DARC using the ELISA-based assay in Oxford. Samples were titrated starting at 1:5 dilution down to 1:640. Dashed line indicates 50% binding inhibition. Groups coded by color and symbol. (C) Data show the interpolated dilution for each sample that gave 50% binding inhibition. One sample in group 2B did not reach 50% binding inhibition by 1:640 dilution and is plotted at this final titer with open triangle symbol. (D) Correlation of 50% binding-inhibition titers for the SalI and HMP013 alleles of PvDBP_RII measured using the assay in Oxford. Spearman’s rank correlation coefficient (rs) and P value are shown (n = 19). PvDBP_RII, region II of the P. vivax Duffy-binding protein.

References

    1. Halbroth BR, Draper SJ. Recent developments in malaria vaccinology. Adv Parasitol. 2015;88:1–49.
    1. Gething PW, et al. A long neglected world malaria map: Plasmodium vivax endemicity in 2010. PLoS Negl Trop Dis. 2012;6(9):e1814. doi: 10.1371/journal.pntd.0001814.
    1. Howes RE, et al. Plasmodium vivax transmission in Africa. PLoS Negl Trop Dis. 2015;9(11):e0004222. doi: 10.1371/journal.pntd.0004222.
    1. Baird JK. Evidence and implications of mortality associated with acute Plasmodium vivax malaria. Clin Microbiol Rev. 2013;26(1):36–57. doi: 10.1128/CMR.00074-12.
    1. Moorthy VS, Newman RD, Okwo-Bele JM. Malaria vaccine technology roadmap. Lancet. 2013;382(9906):1700–1701.
    1. Herrera S, et al. Phase I safety and immunogenicity trial of Plasmodium vivax CS derived long synthetic peptides adjuvanted with montanide ISA 720 or montanide ISA 51. Am J Trop Med Hyg. 2011;84(2 Suppl):12–20.
    1. Bennett JW, et al. Phase 1/2a trial of Plasmodium vivax malaria vaccine candidate VMP001/AS01B in malaria-naive adults: safety, immunogenicity, and efficacy. PLoS Negl Trop Dis. 2016;10(2):e0004423. doi: 10.1371/journal.pntd.0004423.
    1. Vanloubbeeck Y, et al. Comparison of the immune responses induced by soluble and particulate Plasmodium vivax circumsporozoite vaccine candidates formulated in AS01 in rhesus macaques. Vaccine. 2013;31(52):6216–6224. doi: 10.1016/j.vaccine.2013.10.041.
    1. Malkin EM, et al. Phase 1 vaccine trial of Pvs25H: a transmission blocking vaccine for Plasmodium vivax malaria. Vaccine. 2005;23(24):3131–3138. doi: 10.1016/j.vaccine.2004.12.019.
    1. Wu Y, et al. Phase 1 trial of malaria transmission blocking vaccine candidates Pfs25 and Pvs25 formulated with montanide ISA 51. PLoS One. 2008;3(7):e2636. doi: 10.1371/journal.pone.0002636.
    1. Mueller I, Shakri AR, Chitnis CE. Development of vaccines for Plasmodium vivax malaria. Vaccine. 2015;33(52):7489–7495. doi: 10.1016/j.vaccine.2015.09.060.
    1. Tham WH, Healer J, Cowman AF. Erythrocyte and reticulocyte binding-like proteins of Plasmodium falciparum. Trends Parasitol. 2012;28(1):23–30. doi: 10.1016/j.pt.2011.10.002.
    1. Malleret B, et al. Plasmodium vivax: restricted tropism and rapid remodeling of CD71-positive reticulocytes. Blood. 2015;125(8):1314–1324. doi: 10.1182/blood-2014-08-596015.
    1. Chitnis CE, Sharma A. Targeting the Plasmodium vivax Duffy-binding protein. Trends Parasitol. 2008;24(1):29–34. doi: 10.1016/j.pt.2007.10.004.
    1. Miller LH, Mason SJ, Clyde DF, McGinniss MH. The resistance factor to Plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy. N Engl J Med. 1976;295(6):302–304. doi: 10.1056/NEJM197608052950602.
    1. Payne RO, Griffin PM, McCarthy JS, Draper SJ. Plasmodium vivax controlled human malaria infection - progress and prospects. Trends Parasitol. 2017;33(2):141–150. doi: 10.1016/j.pt.2016.11.001.
    1. Singh AP, Ozwara H, Kocken CH, Puri SK, Thomas AW, Chitnis CE. Targeted deletion of Plasmodium knowlesi Duffy binding protein confirms its role in junction formation during invasion. Mol Microbiol. 2005;55(6):1925–1934. doi: 10.1111/j.1365-2958.2005.04523.x.
    1. Zimmerman PA, Ferreira MU, Howes RE, Mercereau-Puijalon O. Red blood cell polymorphism and susceptibility to Plasmodium vivax. Adv Parasitol. 2013;81:27–76.
    1. Tham WH, Beeson JG, Rayner JC. Plasmodium vivax vaccine research - we’ve only just begun. Int J Parasitol. 2017;47(2–3):111–118.
    1. Menard D, et al. Whole genome sequencing of field isolates reveals a common duplication of the Duffy binding protein gene in Malagasy Plasmodium vivax strains. PLoS Negl Trop Dis. 2013;7(11):e2489. doi: 10.1371/journal.pntd.0002489.
    1. Hester J, et al. De novo assembly of a field isolate genome reveals novel Plasmodium vivax erythrocyte invasion genes. PLoS Negl Trop Dis. 2013;7(12):e2569. doi: 10.1371/journal.pntd.0002569.
    1. Hostetler JB, et al. Independent origin and global distribution of distinct Plasmodium vivax Duffy binding protein gene duplications. PLoS Negl Trop Dis. 2016;10(10):e0005091. doi: 10.1371/journal.pntd.0005091.
    1. Ntumngia FB, Thomson-Luque R, Torres Lde M, Gunalan K, Carvalho LH, Adams JH. A Novel erythrocyte binding protein of Plasmodium vivax suggests an alternate invasion pathway into Duffy-positive reticulocytes. MBio. 2016;7(4):e01261.
    1. Batchelor JD, Malpede BM, Omattage NS, DeKoster GT, Henzler-Wildman KA, Tolia NH. Red blood cell invasion by Plasmodium vivax: structural basis for DBP engagement of DARC. PLoS Pathog. 2014;10(1):e1003869. doi: 10.1371/journal.ppat.1003869.
    1. Batchelor JD, Zahm JA, Tolia NH. Dimerization of Plasmodium vivax DBP is induced upon receptor binding and drives recognition of DARC. Nat Struct Mol Biol. 2011;18(8):908–914. doi: 10.1038/nsmb.2088.
    1. Moreno A, et al. Preclinical assessment of the receptor-binding domain of Plasmodium vivax Duffy-binding protein as a vaccine candidate in rhesus macaques. Vaccine. 2008;26(34):4338–4344. doi: 10.1016/j.vaccine.2008.06.010.
    1. de Cassan SC, et al. Preclinical assessment of viral vectored and protein vaccines targeting the Duffy-binding protein region II of Plasmodium vivax. Front Immunol. 2015;6:348.
    1. Wiley SR, et al. Targeting TLRs expands the antibody repertoire in response to a malaria vaccine. Sci Transl Med. 2011;3(93):93ra69.
    1. Singh AP, Puri SK, Chitnis CE. Antibodies raised against receptor-binding domain of Plasmodium knowlesi Duffy binding protein inhibit erythrocyte invasion. Mol Biochem Parasitol. 2002;121(1):21–31. doi: 10.1016/S0166-6851(02)00017-8.
    1. King CL, et al. Naturally acquired Duffy-binding protein-specific binding inhibitory antibodies confer protection from blood-stage Plasmodium vivax infection. Proc Natl Acad Sci USA. 2008;105(24):8363–8368. doi: 10.1073/pnas.0800371105.
    1. Nicolete VC, Frischmann S, Barbosa S, King CL, Ferreira MU. Naturally acquired binding-inhibitory antibodies to Plasmodium vivax Duffy binding protein and clinical immunity to malaria in rural Amazonians. J Infect Dis. 2016;214(10):1539–1546. doi: 10.1093/infdis/jiw407.
    1. de Cassan SC, Draper SJ. Recent advances in antibody-inducing poxviral and adenoviral vectored vaccine delivery platforms for difficult disease targets. Expert Rev Vaccines. 2013;12(4):365–378. doi: 10.1586/erv.13.11.
    1. Draper SJ, et al. Effective induction of high-titer antibodies by viral vector vaccines. Nat Med. 2008;14(8):819–821. doi: 10.1038/nm.1850.
    1. Douglas AD, et al. A PfRH5-based vaccine is efficacious against heterologous strain blood-stage Plasmodium falciparum infection in aotus monkeys. Cell Host Microbe. 2015;17(1):130–139. doi: 10.1016/j.chom.2014.11.017.
    1. O’Hara GA, et al. Clinical assessment of a recombinant simian adenovirus ChAd63: a potent new vaccine vector. J Infect Dis. 2012;205(5):772–781. doi: 10.1093/infdis/jir850.
    1. de Barra E, et al. A phase Ia study to assess the safety and immunogenicity of new malaria vaccine candidates ChAd63 CS administered alone and with MVA CS. PLoS One. 2014;9(12):e115161. doi: 10.1371/journal.pone.0115161.
    1. Sheehy SH, et al. Phase Ia clinical evaluation of the Plasmodium falciparum blood-stage antigen MSP1 in ChAd63 and MVA vaccine vectors. Mol Ther. 2011;19(12):2269–2276. doi: 10.1038/mt.2011.176.
    1. Sheehy SH, et al. Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors. PLoS ONE. 2012;7(2):e31208. doi: 10.1371/journal.pone.0031208.
    1. Sedegah M, et al. Adenovirus 5-vectored P. falciparum vaccine expressing CSP and AMA1. Part A: safety and immunogenicity in seronegative adults. PLoS One. 2011;6(10):e24586. doi: 10.1371/journal.pone.0024586.
    1. Ewer K, et al. A monovalent chimpanzee adenovirus ebola vaccine boosted with MVA. N Engl J Med. 2016;374(17):1635–1646. doi: 10.1056/NEJMoa1411627.
    1. Elias SC, et al. Analysis of human B-cell responses following ChAd63-MVA MSP1 and AMA1 immunization and controlled malaria infection. Immunology. 2014;141(4):628–644. doi: 10.1111/imm.12226.
    1. Hodgson SH, et al. Combining viral vectored and protein-in-adjuvant vaccines against the blood-stage malaria antigen AMA1: report on a phase 1a clinical trial. Mol Ther. 2014;22(12):2142–2154. doi: 10.1038/mt.2014.157.
    1. Cole-Tobian JL, et al. Strain-specific duffy binding protein antibodies correlate with protection against infection with homologous compared to heterologous plasmodium vivax strains in Papua New Guinean children. Infect Immun. 2009;77(9):4009–4017. doi: 10.1128/IAI.00158-09.
    1. Ceravolo IP, et al. Naturally acquired inhibitory antibodies to Plasmodium vivax Duffy binding protein are short-lived and allele-specific following a single malaria infection. Clin Exp Immunol. 2009;156(3):502–510. doi: 10.1111/j.1365-2249.2009.03931.x.
    1. Sheehy SH, et al. ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol Ther. 2012;20(12):2355–2368. doi: 10.1038/mt.2012.223.
    1. Afolabi MO, et al. Safety and immunogenicity of ChAd63 and MVA ME-TRAP in West African children and infants. Mol Ther. 2016;24(8):1470–1477. doi: 10.1038/mt.2016.83.
    1. Green CA, et al. Chimpanzee adenovirus- and MVA-vectored respiratory syncytial virus vaccine is safe and immunogenic in adults. Sci Transl Med. 2015;7(300):300ra126. doi: 10.1126/scitranslmed.aac5745.
    1. Swadling L, et al. A human vaccine strategy based on chimpanzee adenoviral and MVA vectors that primes, boosts, and sustains functional HCV-specific T cell memory. Sci Transl Med. 2014;6(261):261ra153. doi: 10.1126/scitranslmed.3009185.
    1. Gilbert SC. Clinical development of modified vaccinia virus Ankara vaccines. Vaccine. 2013;31(39):4241–4246. doi: 10.1016/j.vaccine.2013.03.020.
    1. Xainli J, Baisor M, Kastens W, Bockarie M, Adams JH, King CL. Age-dependent cellular immune responses to Plasmodium vivax Duffy binding protein in humans. J Immunol. 2002;169(6):3200–3207. doi: 10.4049/jimmunol.169.6.3200.
    1. Bijker EM, et al. Protection against malaria after immunization by chloroquine prophylaxis and sporozoites is mediated by preerythrocytic immunity. Proc Natl Acad Sci U S A. 2013;110(19):7862–7867. doi: 10.1073/pnas.1220360110.
    1. Burel JG, Apte SH, McCarthy JS, Doolan DL. Plasmodium vivax but not Plasmodium falciparum blood-stage infection in humans is associated with the expansion of a CD8+ T cell population with cytotoxic potential. PLoS Negl Trop Dis. 2016;10(12):e0005031. doi: 10.1371/journal.pntd.0005031.
    1. Biswas S, et al. Assessment of humoral immune responses to blood-stage malaria antigens following ChAd63-MVA immunization, controlled human malaria infection and natural exposure. PLoS One. 2014;9(9):e107903. doi: 10.1371/journal.pone.0107903.
    1. Tran TM, et al. Comparison of IgG reactivities to Plasmodium vivax merozoite invasion antigens in a Brazilian Amazon population. Am J Trop Med Hyg. 2005;73(2):244–255.
    1. Zakeri S, Babaeekhou L, Mehrizi AA, Abbasi M, Djadid ND. Antibody responses and avidity of naturally acquired anti-Plasmodium vivax Duffy binding protein (PvDBP) antibodies in individuals from an area with unstable malaria transmission. Am J Trop Med Hyg. 2011;84(6):944–950. doi: 10.4269/ajtmh.2011.11-0001.
    1. Sampath S, et al. Glycan masking of Plasmodium vivax Duffy Binding Protein for probing protein binding function and vaccine development. PLoS Pathog. 2013;9(6):e1003420. doi: 10.1371/journal.ppat.1003420.
    1. Kano FS, et al. The presence, persistence and functional properties of Plasmodium vivax Duffy binding protein ii antibodies are influenced by HLA class II allelic variants. PLoS Negl Trop Dis. 2016;10(12):e0005177. doi: 10.1371/journal.pntd.0005177.
    1. Chootong P, et al. Mapping epitopes of the Plasmodium vivax Duffy binding protein with naturally acquired inhibitory antibodies. Infect Immun. 2010;78(3):1089–1095. doi: 10.1128/IAI.01036-09.
    1. Chen E, et al. Broadly neutralizing epitopes in the Plasmodium vivax vaccine candidate Duffy binding protein. Proc Natl Acad Sci U S A. 2016;113(22):6277–6282. doi: 10.1073/pnas.1600488113.
    1. Ntumngia FB, et al. Conserved and variant epitopes of Plasmodium vivax Duffy binding protein as targets of inhibitory monoclonal antibodies. Infect Immun. 2012;80(3):1203–1208. doi: 10.1128/IAI.05924-11.
    1. Russell B, et al. A reliable ex vivo invasion assay of human reticulocytes by Plasmodium vivax. Blood. 2011;118(13):e74–e81. doi: 10.1182/blood-2011-04-348748.
    1. Payne RO, et al. Demonstration of the blood-stage Plasmodium falciparum controlled human malaria infection model to assess efficacy of the P. falciparum apical membrane antigen 1 vaccine, FMP2.1/AS01. J Infect Dis. 2016;213(11):1743–1751. doi: 10.1093/infdis/jiw039.
    1. Hjerrild KA, et al. Production of full-length soluble Plasmodium falciparum RH5 protein vaccine using a Drosophila melanogaster Schneider 2 stable cell line system. Sci Rep. 2016;6:30357.
    1. Shakri AR, Rizvi MM, Chitnis CE. Development of quantitative receptor-ligand binding assay for use as a tool to estimate immune responses against Plasmodium vivax Duffy binding protein region II. J Immunoassay Immunochem. 2012;33(4):403–413. doi: 10.1080/15321819.2012.659781.

Source: PubMed

3
Abonneren