21 research outputs found

    Immunogenicity and protective efficacy of recombinant vaccine based on the receptor-binding domain of the Plasmodium vivax duffy binding protein in Aotus monkeys

    Get PDF
    Invasion of human erythrocytes by Plasmodium vivax requires interaction between Duffy binding protein (PvDBP) and the Duffy blood group antigen. The receptor-binding domain of PvDBP lies in a conserved N-terminal, cysteine-rich region, region II (PvRII). PvRII is a valuable malaria subunit vaccine candidate for asexual blood stages. We have evaluated in Aotus monkeys the immunogenicity and protective efficacy of recombinant PvRII formulated in Freund's and Montanide ISA720 adjuvants. Specific antibody titers were determined by an enzyme-linked immunosorbent assay after each of three doses of 50µg of protein administered by the subcutaneous route. Immunization with PvRII formulated in Freund's adjuvant yielded higher antibody titers than immunization with the Montanide ISA720 formulation and offered partial protection. Although the Montanide ISA720 formulation was immunogenic, it did not provide any protection. Given the immunogenicity and partial protection observed, further studies are needed to optimize the PvRII vaccine formulation with adjuvants suitable for human use

    Evaluation of immune responses elicited in mice against a recombinant malaria vaccine based on Plasmodium vivax duffy binding protein

    No full text
    Plasmodium vivax Duffy binding protein (PvDBP) binds the Duffy blood group antigen as the obligate receptor for erythrocyte invasion. We have tested in mice the immunogenicity of recombinant P. vivax region II (PvRII), the receptor-binding domain of PvDBP, formulated with five adjuvants, namely, Montanide ISA720, AS02A, alum, QS21 and MF59. All the formulations elicited high titer antibodies, with Montanide ISA720 and AS02A yielding the highest titers followed by MF59, QS21 and alum. Sera raised against PvRII formulated with AS02A and Montanide ISA720 followed by alum were most effective at blocking PvRII binding to erythrocytes in a functional assay. Analysis of cellular immune responses indicated that all adjuvant groups induced significant interferon-γ, with alum being the highest interferon-γ inducer. These results suggest that recombinant PvRII formulated with human compatible adjuvants is immunogenic in small animal models and that Montanide ISA720, AS02A and alum perform better than MF59 and QS21 in terms of their ability to elicit high titer binding inhibitory antibodies

    Immunogenicity of Plasmodium vivax combination subunit vaccine formulated with human compatible adjuvants in mice

    No full text
    An effective malaria vaccine will probably require the delivery of multiple antigens that induce several layers of immunity. Malaria antigens expressed on the surface and in apical organelles of blood-stage merozoites are potential vaccine candidates given their importance in the invasion of erythrocytes. The present study examined the kinetics of humoral response in BALB/c mice following immunization with combination of two blood-stage Plasmodium vivax invasion related molecules, the N-terminal, cysteine-rich region II of P. vivax Duffy binding protein (PvRII) and the 19 kDa C-terminal region of merozoite surface protein 1 (PvMSP119) formulated with Montanide ISA 720 and alhydrogel. Immunization with combination of recombinant PvRII and PvMSP119 formulated with the Montanide ISA 720 elicited higher antibody titer compared to the alhydrogel formulation. In case of both the adjuvants tested, combination of PvRII and PvMSP119 did not result in suppression of antibody response against either antigen when compared to immunization with individual antigens alone. Analysis of IgG subclasses showed that combination of both the recombinant proteins induced a mixed Th1/Th2-type response with almost all IgG subtypes being expressed in equivalent amount. Antibodies elicited against PvRII showed significant inhibitory effect on the binding of PvRII to recombinant Duffy antigen receptor for chemokines (DARC) in an in vitro binding assay. The results of the present study provide a rationale for a combination vaccine against P. vivax malaria based on PvMSP119 and PvRII

    Antibody responses to Plasmodium vivax Duffy binding and Erythrocyte binding proteins predict risk of infection and are associated with protection from clinical Malaria

    Get PDF
    International audienceBackground: The Plasmodium vivax Duffy Binding Protein (PvDBP) is a key target of naturally acquired immunity. However, region II of PvDBP, which contains the receptor-binding site, is highly polymorphic. The natural acquisition of antibodies to different variants of PvDBP region II (PvDBPII), including the AH, O, P and Sal1 alleles, the central region III-V (PvDBPIII-V), and P. vivax Erythrocyte Binding Protein region II (PvEBPII) and their associations with risk of clinical P. vivax malaria are not well understood.Methodology: Total IgG and IgG subclasses 1, 2, and 3 that recognize four alleles of PvDBPII (AH, O, P, and Sal1), PvDBPIII-V and PvEBPII were measured in samples collected from a cohort of 1 to 3 year old Papua New Guinean (PNG) children living in a highly endemic area of PNG. The levels of binding inhibitory antibodies (BIAbs) to PvDBPII (AH, O, and Sal1) were also tested in a subset of children. The association of presence of IgG with age, cumulative exposure (measured as the product of age and malaria infections during follow-up) and prospective risk of clinical malaria were evaluated.Results: The increase in antigen-specific total IgG, IgG1, and IgG3 with age and cumulative exposure was only observed for PvDBPII AH and PvEBPII. High levels of total IgG and predominant subclass IgG3 specific for PvDBPII AH were associated with decreased incidence of clinical P. vivax episodes (aIRR = 0.56–0.68, P≤0.001–0.021). High levels of total IgG and IgG1 to PvEBPII correlated strongly with protection against clinical vivax malaria compared with IgGs against all PvDBPII variants (aIRR = 0.38, P<0.001). Antibodies to PvDBPII AH and PvEBPII showed evidence of an additive effect, with a joint protective association of 70%.Conclusion: Antibodies to the key parasite invasion ligands PvDBPII and PvEBPII are good correlates of protection against P. vivax malaria in PNG. This further strengthens the rationale for inclusion of PvDBPII in a recombinant subunit vaccine for P. vivax malaria and highlights the need for further functional studies to determine the potential of PvEBPII as a component of a subunit vaccine for P. vivax malaria

    Upregulation of ZIP14 and Altered Zinc Homeostasis in Muscles in Pancreatic Cancer Cachexia

    No full text
    Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer type in which the mortality rate approaches the incidence rate. More than 85% of PDAC patients experience a profound loss of muscle mass and function, known as cachexia. PDAC patients with this condition suffer from decreased tolerance to anti-cancer therapies and often succumb to premature death due to respiratory and cardiac muscle wasting. Yet, there are no approved therapies available to alleviate cachexia. We previously found that upregulation of the metal ion transporter, Zip14, and altered zinc homeostasis are critical mediators of cachexia in metastatic colon, lung, and breast cancer models. Here, we show that a similar mechanism is likely driving the development of cachexia in PDAC. In two independent experimental metastasis models generated from the murine PDAC cell lines, Pan02 and FC1242, we observed aberrant Zip14 expression and increased zinc ion levels in cachectic muscles. Moreover, in advanced PDAC patients, high levels of ZIP14 in muscles correlated with the presence of cachexia. These studies underscore the importance of altered ZIP14 function in PDAC-associated cachexia development and highlight a potential therapeutic opportunity for improving the quality of life and prolonging survival in PDAC patients
    corecore