Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 14(8): e1007273, 2018 08.
Article in English | MEDLINE | ID: mdl-30153307

ABSTRACT

Human whipworm (Trichuris trichiura) infects approximately 1 in 15 people worldwide, representing the leading infectious cause of colitis and subsequent, inflammatory bowel disease (IBD). Current control measures focused on mass deworming have had limited success due to low drug efficacies. Vaccination would be an ideal, cost-effective strategy to induce protective immunity, leading to control of infection and transmission. Here we report the identification of whey acidic protein, a whipworm secretory protein, as a strong immunogen for inducing protective efficacy in a surrogate mouse T. muris infection model. The recombinant WAP protein (rTm-WAP49), as well as a single, highly conserved repeat within WAP (fragment 8) expressed as an Na-GST-1 fusion protein (rTm-WAP-F8+Na-GST-1), generate a strong T helper type 2 (Th2) immune response when delivered as subcutaneous vaccines formulated with Montanide ISA 720. Oral challenge with T. muris infective eggs following vaccination led to a significant reduction in worm burden of 48% by rTm-WAP49 and 33% by rTm-WAP-F8+Na-GST-1. The cellular immune correlates of protection included significant antigen-specific production of Th2 cytokines IL-4, IL-9, and IL-13 by cells isolated from the vaccine-draining inguinal lymph nodes, parasite-draining mesenteric lymph nodes, and spleen in mice vaccinated with either rTm-WAP49 or rTm-WAP-F8+Na-GST-1. The humoral immune correlates included a high antigen-specific ratio of IgG1 to IgG2a, without eliciting an IgE-mediated allergic response. Immunofluorescent staining of adult T. muris with WAP antisera identified the worm's pathogenic stichosome organ as the site of secretion of native Tm-WAP protein into the colonic mucosa. Given the high sequence conservation for the WAP proteins from T. muris and T. trichiura, the results presented here support the WAP protein to be further evaluated as a potential human whipworm vaccine candidate.


Subject(s)
Immunity , Milk Proteins/immunology , Trichuriasis/prevention & control , Trichuris/immunology , Animals , Antibodies, Helminth/metabolism , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Antigens, Helminth/pharmacology , Immunity/drug effects , Immunity/genetics , Male , Mice , Mice, Inbred AKR , Mice, Knockout , Mice, SCID , Milk Proteins/genetics , Milk Proteins/pharmacology , Trichuriasis/immunology , Trichuris/genetics , Vaccination/methods
2.
Vaccine ; 36(14): 1853-1862, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29496347

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) has infected at least 2040 patients and caused 712 deaths since its first appearance in 2012, yet neither pathogen-specific therapeutics nor approved vaccines are available. To address this need, we are developing a subunit recombinant protein vaccine comprising residues 377-588 of the MERS-CoV spike protein receptor-binding domain (RBD), which, when formulated with the AddaVax adjuvant, it induces a significant neutralizing antibody response and protection against MERS-CoV challenge in vaccinated animals. To prepare for the manufacture and first-in-human testing of the vaccine, we have developed a process to stably produce the recombinant MERS S377-588 protein in Chinese hamster ovary (CHO) cells. To accomplish this, we transfected an adherent dihydrofolate reductase-deficient CHO cell line (adCHO) with a plasmid encoding S377-588 fused with the human IgG Fc fragment (S377-588-Fc). We then demonstrated the interleukin-2 signal peptide-directed secretion of the recombinant protein into extracellular milieu. Using a gradually increasing methotrexate (MTX) concentration to 5 µM, we increased protein yield by a factor of 40. The adCHO-expressed S377-588-Fc recombinant protein demonstrated functionality and binding specificity identical to those of the protein from transiently transfected HEK293T cells. In addition, hCD26/dipeptidyl peptidase-4 (DPP4) transgenic mice vaccinated with AddaVax-adjuvanted S377-588-Fc could produce neutralizing antibodies against MERS-CoV and survived for at least 21 days after challenge with live MERS-CoV with no evidence of immunological toxicity or eosinophilic immune enhancement. To prepare for large scale-manufacture of the vaccine antigen, we have further developed a high-yield monoclonal suspension CHO cell line.


Subject(s)
Antigens, Viral/genetics , Antigens, Viral/immunology , Coronavirus Infections/immunology , Gene Expression , Genetic Engineering , Middle East Respiratory Syndrome Coronavirus/immunology , Viral Vaccines/immunology , Animals , CHO Cells , Coronavirus Infections/prevention & control , Cricetulus , Epitopes/chemistry , Epitopes/immunology , Genetic Vectors/genetics , Immunogenicity, Vaccine , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Mice , Protein Processing, Post-Translational , Recombinant Proteins/genetics , Recombinant Proteins/immunology
3.
J Pharm Sci ; 107(5): 1468-1473, 2018 05.
Article in English | MEDLINE | ID: mdl-29274820

ABSTRACT

Chagas disease due to chronic infection with Trypanosoma cruzi is a neglected cause of heart disease, affecting approximately 6-10 million individuals in Latin America and elsewhere. T. cruzi Tc24, a calcium-binding protein in the flagellar pocket of the parasite, is a candidate antigen for an injectable therapeutic vaccine as an alternative or a complement to chemotherapy. Previously, we reported that a genetically engineered construct from which all cysteine residues had been eliminated (Tc24-C4) yields a recombinant protein with reduced aggregation and improved analytical purity in comparison to the wild-type form, without compromising antigenicity and immunogenicity. We now report that the established process for producing Escherichia coli-expressed Tc24-C4 protein is robust and reproducibly yields protein lots with consistent analytical characteristics, freeze-thaw, accelerated, and long-term stability profiles. The data indicate that, like most proteins, Tc24-C4 should be stable at -80°C, but also at 4°C and room temperature for at least 30 days, and up to 7-15 days at 37°C. Thus, the production process for recombinant Tc24-C4 is suitable for Current Good Manufacturing Practice production and clinical testing, based on process robustness, analytical characteristics, and stability profile.


Subject(s)
Antigens, Protozoan/chemistry , Calcium-Binding Proteins/chemistry , Protozoan Proteins/chemistry , Protozoan Vaccines/chemistry , Trypanosoma cruzi/chemistry , Antigens, Protozoan/immunology , Calcium-Binding Proteins/immunology , Chagas Disease/immunology , Chagas Disease/prevention & control , Freezing , Humans , Protein Stability , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Temperature , Trypanosoma cruzi/immunology
4.
J Biomed Mater Res B Appl Biomater ; 106(4): 1587-1594, 2018 05.
Article in English | MEDLINE | ID: mdl-28804955

ABSTRACT

It is believed that an effective vaccine against leishmaniasis will require a T helper type 1 (TH 1) immune response. In this study, we investigated the adjuvanticity of the Toll-like receptor (TLR) 7/8 agonist 3M-052 in combination with the Leishmania donovani 36-kDa nucleoside hydrolase recombinant protein antigen (NH36). NH36 and 3M-052 were encapsulated in separate batches of poly(lactic-co-glycolic acid) (PLGA) microparticles (MPs). The loading efficiency for NH36 was 83% and for 3M-052 was above 95%. In vitro stimulation of bone marrow-derived dendritic cells, measured by IL-12 secretion, demonstrated that 3M-052 (free or MP-formulated) had a concentration-dependent immunostimulatory effect with an optimum concentration of 2 µg/mL. In immunogenicity studies in BALB/c mice, MP-formulated NH36 and 3M-052 elicited the highest serum titers of TH 1-associated IgG2a and IgG2b antibodies and the highest frequency of IFNγ-producing splenocytes. No dose dependency was observed among MP/NH36/3M-052 groups over a dose range of 4-60 µg 3M-052 per injection. The ability of MP-formulated NH36 and 3M-052 to elicit a TH 1-biased immune response indicates the potential for PLGA MP-formulated 3M-052 to be used as an adjuvant for leishmaniasis vaccines. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1587-1594, 2018.


Subject(s)
Antigens, Protozoan , Heterocyclic Compounds, 3-Ring , Leishmania donovani/immunology , Leishmaniasis Vaccines , Leishmaniasis, Visceral , Polylactic Acid-Polyglycolic Acid Copolymer , Protozoan Proteins , Stearic Acids , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/pharmacology , Dose-Response Relationship, Immunologic , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Immunogenicity, Vaccine , Leishmaniasis Vaccines/chemistry , Leishmaniasis Vaccines/pharmacology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/pathology , Leishmaniasis, Visceral/prevention & control , Mice , Mice, Inbred BALB C , Molybdoferredoxin , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Protozoan Proteins/chemistry , Protozoan Proteins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Stearic Acids/chemistry , Stearic Acids/pharmacology
5.
J Parasitol ; 103(5): 579-583, 2017 10.
Article in English | MEDLINE | ID: mdl-28581897

ABSTRACT

B-cell superantigens (BC-SAgs) are immunoevasins that have evolved in response to innate catalytic IgM antibodies; germ-line encoded immunoglobulins present in the preimmune repertoire independent of prior antigen exposure. Catalysis is the result of a 2-step process that involves first the formation of a non-covalent bond between the BC-SAg and the immunoglobulin followed by covalent bond formation at the catalytic site resulting in target hydrolysis. Tc24 is a recently described Trypanosoma cruzi BC-SAg hypothesized to play a role in evading the humoral response early in the infection period. We previously demonstrated that exposure to Tc24 following immunization or infection resulted in the depletion of the catalytic IgM response, leaving a gap in the catalytic IgM repertoire. The present report compares the BC-SAg properties of wild-type Tc24 (Tc24-WT) to that of 2 recombinant Tc24 isoforms: Tc24-C2 (Cys to Ser mutations in the 2 most-proximal Cys residues) and Tc24-C4 (Cys to Ser mutations in all 4 Cys residues present). BC-SAg activity was assessed by immunizing mice with the respective isoforms and examining the ability of IgM purified from the respective groups to hydrolyze the 3 Tc24 isoforms. In addition, the ability of IgM purified from naive mice to hydrolyze the Tc24 isoforms was also assessed. Immunization with Tc24-WT, Tc24-C2, or Tc24-C4 resulted in loss of IgM-mediated hydrolysis of Tc24-WT. However, the ability of IgM purified from naive mice (previously shown to hydrolyze Tc24-WT) was less effective in hydrolyzing the 2 Tc24 isoforms. These data demonstrate that although the BC-SAg site in the mutants remained intact, their reduced susceptibility to IgM-mediated hydrolysis suggested that structural changes resulting from the Cys to Ser mutations altered accessibility to the catalytic site in the 2 isoforms.


Subject(s)
Cysteine/genetics , Immunoglobulin M/immunology , Superantigens/genetics , Trypanosoma cruzi/genetics , Trypanosoma cruzi/immunology , Animals , Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Calcium-Binding Proteins/immunology , Chagas Disease/immunology , Cysteine/chemistry , Hydrolysis , Immunity, Innate/immunology , Immunization , Mice , Mutation , Protein Isoforms/immunology , Protozoan Vaccines , Sequence Alignment , Serine/chemistry , Serine/genetics , Superantigens/chemistry , Superantigens/immunology
6.
J Pharm Sci ; 106(8): 1961-1970, 2017 08.
Article in English | MEDLINE | ID: mdl-28456726

ABSTRACT

From 2002 to 2003, a global pandemic of severe acute respiratory syndrome (SARS) spread to 5 continents and caused 8000 respiratory infections and 800 deaths. To ameliorate the effects of future outbreaks as well as to prepare for biodefense, a process for the production of a recombinant protein vaccine candidate is under development. Previously, we reported the 5 L scale expression and purification of a promising recombinant SARS vaccine candidate, RBD219-N1, the 218-amino acid residue receptor-binding domain (RBD) of SARS coronavirus expressed in yeast-Pichia pastoris X-33. When adjuvanted with aluminum hydroxide, this protein elicited high neutralizing antibody titers and high RBD-specific antibody titers. However, the yield of RBD219-N1 (60 mg RBD219-N1 per liter of fermentation supernatant; 60 mg/L FS) still required improvement to reach our target of >100 mg/L FS. In this study, we optimized the 10 L scale production process and increased the fermentation yield 6- to 7-fold to 400 mg/L FS with purification recovery >50%. A panel of characterization tests indicated that the process is reproducible and that the purified, tag-free RBD219-N1 protein has high purity and a well-defined structure and is therefore a suitable candidate for production under current Good Manufacturing Practice and future phase-1 clinical trials.


Subject(s)
Pichia/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Spike Glycoprotein, Coronavirus/genetics , Vaccines, Synthetic/genetics , Viral Vaccines/genetics , Cloning, Molecular/methods , Fermentation , Humans , Industrial Microbiology/methods , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Severe acute respiratory syndrome-related coronavirus/chemistry , Severe Acute Respiratory Syndrome/prevention & control , Severe Acute Respiratory Syndrome/virology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/isolation & purification , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/isolation & purification , Viral Vaccines/chemistry , Viral Vaccines/isolation & purification
7.
PLoS Negl Trop Dis ; 11(3): e0005374, 2017 03.
Article in English | MEDLINE | ID: mdl-28278244

ABSTRACT

BACKGROUND: Immunity to the sand fly salivary protein SALO (Salivary Anticomplement of Lutzomyia longipalpis) protected hamsters against Leishmania infantum and L. braziliensis infection and, more recently, a vaccine combination of a genetically modified Leishmania with SALO conferred strong protection against L. donovani infection. Because of the importance of SALO as a potential component of a leishmaniasis vaccine, a plan to produce this recombinant protein for future scale manufacturing as well as knowledge of its structural characteristics are needed to move SALO forward for the clinical path. METHODOLOGY/PRINCIPAL FINDINGS: Recombinant SALO was expressed as a soluble secreted protein using Pichia pastoris, rSALO(P), with yields of 1g/L and >99% purity as assessed by SEC-MALS and SDS-PAGE. Unlike its native counterpart, rSALO(P) does not inhibit the classical pathway of complement; however, antibodies to rSALO(P) inhibit the anti-complement activity of sand fly salivary gland homogenate. Immunization with rSALO(P) produces a delayed type hypersensitivity response in C57BL/6 mice, suggesting rSALO(P) lacked anti-complement activity but retained its immunogenicity. The structure of rSALO(P) was solved by S-SAD at Cu-Kalpha to 1.94 Å and refined to Rfactor 17%. SALO is ~80% helical, has no appreciable structural similarities to any human protein, and has limited structural similarity in the C-terminus to members of insect odorant binding proteins. SALO has three predicted human CD4+ T cell epitopes on surface exposed helices. CONCLUSIONS/SIGNIFICANCE: The results indicate that SALO as expressed and purified from P. pastoris is suitable for further scale-up, manufacturing, and testing. SALO has a novel structure, is not similar to any human proteins, is immunogenic in rodents, and does not have the anti-complement activity observed in the native salivary protein which are all important attributes to move this vaccine candidate forward to the clinical path.


Subject(s)
Psychodidae/chemistry , Recombinant Proteins/immunology , Salivary Proteins and Peptides/immunology , Animals , Gene Expression , Mice, Inbred C57BL , Pichia/genetics , Pichia/metabolism , Protein Conformation , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Salivary Proteins and Peptides/administration & dosage , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/genetics
8.
Hum Vaccin Immunother ; 13(3): 621-633, 2017 03 04.
Article in English | MEDLINE | ID: mdl-27737611

ABSTRACT

A therapeutic vaccine for human Chagas disease is under development by the Sabin Vaccine Institute Product Development Partnership. The aim of the vaccine is to significantly reduce the parasite burden of Trypanosoma cruzi in humans, either as a standalone product or in combination with conventional chemotherapy. Vaccination of mice with Tc24 formulated with monophosphoryl-lipid A (MPLA) adjuvant results in a Th1 skewed immune response with elevated IgG2a and IFNγ levels and a statistically significant decrease in parasitemia following T. cruzi challenge. Tc24 was therefore selected for scale-up and further evaluation. During scale up and downstream process development, significant protein aggregation was observed due to intermolecular disulfide bond formation. To prevent protein aggregation, cysteine codons were replaced with serine codons which resulted in the production of a non-aggregated and soluble recombinant protein, Tc24-C4. No changes to the secondary structure of the modified molecule were detected by circular dichroism. Immunization of mice with wild-type Tc24 or Tc24-C4, formulated with E6020 (TLR4 agonist analog to MPLA) emulsified in a squalene-oil-in-water emulsion, resulted in IgG2a and antigen specific IFNγ production levels from splenocytes that were not significantly different, indicating that eliminating putative intermolecular disulfide bonds had no significant impact on the immunogenicity of the molecule. In addition, vaccination with either formulated wild type Tc24 or Tc24-C4 antigen also significantly increased survival and reduced cardiac parasite burden in mice. Investigations are now underway to examine the efficacy of Tc24-C4 formulated with other adjuvants to reduce parasite burden and increase survival in pre-clinical studies.


Subject(s)
Chagas Disease/prevention & control , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Recombinant Proteins/immunology , Trypanosoma cruzi/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Cysteine/genetics , Disease Models, Animal , Female , Heart/parasitology , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice, Inbred BALB C , Mutagenesis , Parasite Load , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Survival Analysis , Trypanosoma cruzi/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
9.
Protein Expr Purif ; 130: 129-136, 2017 02.
Article in English | MEDLINE | ID: mdl-27773761

ABSTRACT

The nucleoside hydrolase gene from Leishmania donovani was cloned and expressed in Escherichia coli as a full length 36-kDa protein (LdNH36). Following lysis and extraction, the protein was purified by anion exchange and gel filtration chromatography. The purified protein had a molecular mass of approximately 36-kDa and was confirmed to be >99% pure. Using a nucleoside hydrolase assay, the protein was found to exhibit a Km of 741 ± 246 µM. Protein integrity was confirmed by lithium dodecyl sulfate polyacrylamide gel electrophoresis (LDS-PAGE), mass spectrometry (MS), and enzymatic assay. Analysis of antibody levels from immunized mice indicated that LdNH36 alone or in a stable emulsion with the Toll-like receptor-4 ligand glucopyranosyl lipid adjuvant (GLA-SE) as immunostimulant induced high levels of antigen-specific IgG antibodies. The cellular immune response indicated a Th1 response in mice immunized with LdNH36, but only when formulated with GLA-SE. Mice immunized with the LdNH36 antigen in combination with the GLA-SE adjuvant and challenged with Leishmania mexicana showed significant reductions (>20 fold) in parasite burden, confirming the protective efficacy of this vaccine candidate.


Subject(s)
Immunogenicity, Vaccine , Leishmania donovani , Leishmaniasis Vaccines , Leishmaniasis, Cutaneous , N-Glycosyl Hydrolases , Protozoan Proteins , Animals , Female , Leishmania donovani/enzymology , Leishmania donovani/genetics , Leishmania donovani/immunology , Leishmaniasis Vaccines/biosynthesis , Leishmaniasis Vaccines/immunology , Leishmaniasis Vaccines/isolation & purification , Leishmaniasis Vaccines/pharmacokinetics , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Mice , Mice, Inbred BALB C , N-Glycosyl Hydrolases/biosynthesis , N-Glycosyl Hydrolases/immunology , N-Glycosyl Hydrolases/isolation & purification , N-Glycosyl Hydrolases/pharmacology , Protozoan Proteins/biosynthesis , Protozoan Proteins/immunology , Protozoan Proteins/isolation & purification , Protozoan Proteins/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
10.
Hum Vaccin Immunother ; 12(7): 1707-20, 2016 07 02.
Article in English | MEDLINE | ID: mdl-26839079

ABSTRACT

Leishmania donovani is the major cause of visceral leishmaniasis (kala-azar), now recognized as the parasitic disease with the highest level of mortality second only to malaria. No human vaccine is currently available. A 36 kDa L. donovani nucleoside hydrolase (LdNH36) surface protein has been previously identified as a potential vaccine candidate antigen. Here we present data on the expression of LdNH36 in Pichia pastoris and its purification at the 20 L scale to establish suitability for future pilot scale manufacturing. To improve efficiency of process development and ensure reproducibility, 4 N-linked glycosylation sites shown to contribute to heterogeneous high-mannose glycosylation were mutated to glutamine residues. The mutant LdNH36 (LdNH36-dg2) was expressed and purified to homogeneity. Size exclusion chromatography and light scattering demonstrated that LdNH36-dg2 existed as a tetramer in solution, similar to the wild-type recombinant L. major nucleoside hydrolase. The amino acid mutations do not affect the tetrameric interface as confirmed by theoretical modeling, and the mutated amino acids are located outside the major immunogenic domain. Immunogenic properties of the LdNH36-dg2 recombinant protein were evaluated in BALB/c mice using formulations that included a synthetic CpG oligodeoxynucleotide, together with a microparticle delivery platform (poly(lactic-co-glycolic acid)). Mice exhibited high levels of IgG1, IgG2a, and IgG2b antibodies that were reactive to both LdNH36-dg2 and LdNH36 wild-type. While the point mutations did affect the hydrolase activity of the enzyme, the IgG antibodies elicited by LdNH36-dg2 were shown to inhibit the hydrolase activity of the wild-type LdNH36. The results indicate that LdNH36-dg2 as expressed in and purified from P. pastoris is suitable for further scale-up, manufacturing, and testing in support of future first-in-humans phase 1 clinical trials.


Subject(s)
Antigens, Protozoan/immunology , Gene Expression , Leishmania donovani/immunology , Mutant Proteins/immunology , N-Glycosyl Hydrolases/immunology , Recombinant Proteins/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Chromatography, Gel , Dynamic Light Scattering , Female , Immunoglobulin G/blood , Leishmania donovani/genetics , Mice, Inbred BALB C , Models, Molecular , Molecular Weight , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/isolation & purification , N-Glycosyl Hydrolases/genetics , Pichia/genetics , Pichia/metabolism , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
11.
Vaccine ; 33(36): 4505-12, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26192358

ABSTRACT

The Tc24 calcium binding protein from the flagellar pocket of Trypanosoma cruzi is under evaluation as a candidate vaccine antigen against Chagas disease. Previously, a DNA vaccine encoding Tc24 was shown to be an effective vaccine (both as a preventive and therapeutic intervention) in mice and dogs, as evidenced by reductions in T. cruzi parasitemia and cardiac amastigotes, as well as reduced cardiac inflammation and increased host survival. Here we developed a suitable platform for the large scale production of recombinant Tc24 (rTc24) and show that when rTc24 is combined with a monophosphoryl-lipid A (MPLA) adjuvant, the formulated vaccine induces a Th1-biased immune response in mice, comprised of elevated IgG2a antibody levels and interferon-gamma levels from splenocytes, compared to controls. These immune responses also resulted in statistically significant decreased T. cruzi parasitemia and cardiac amastigotes, as well as increased survival following T. cruzi challenge infections, compared to controls. Partial protective efficacy was shown regardless of whether the antigen was expressed in Escherichia coli or in yeast (Pichia pastoris). While mouse vaccinations will require further modifications in order to optimize protective efficacy, such studies provide a basis for further evaluations of vaccines comprised of rTc24, together with alternative adjuvants and additional recombinant antigens.


Subject(s)
Antigens, Protozoan/immunology , Chagas Disease/prevention & control , Protozoan Vaccines/immunology , Trypanosoma cruzi/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Chagas Disease/immunology , Cloning, Molecular , Disease Models, Animal , Escherichia coli/genetics , Female , Gene Expression , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Lipid A/administration & dosage , Mice, Inbred BALB C , Parasite Load , Parasitemia/prevention & control , Pichia/genetics , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Spleen/immunology , Survival Analysis , Th1 Cells/immunology , Trypanosoma cruzi/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
12.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 7): 925-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26144240

ABSTRACT

Leishmaniasis is a neglected vector-borne disease with a global prevalence of over 12 million cases and 59,000 annual deaths. Transmission of the parasite requires salivary proteins, including LJL143 from the New World sandfly Lutzomyia longipalpis. LJL143 is a known marker of sandfly exposure in zoonotic hosts. LJL143 was crystallized from soluble protein expressed using Pichia pastoris. X-ray data were collected to 2.6 Šresolution from orthorhombic crystals belonging to space group P2(1)2(1)2(1), with average unit-cell parameters a = 57.39, b = 70.24, c = 79.58 Å. The crystals are predicted to have a monomer in the asymmetric unit, with an estimated solvent content of 48.5%. LJL143 has negligible homology to any reported structures, so the phases could not be determined by molecular replacement. All attempts at S-SAD failed and future studies include experimental phase determination using heavy-atom derivatives.


Subject(s)
Psychodidae , Salivary Proteins and Peptides/biosynthesis , Salivary Proteins and Peptides/chemistry , Amino Acid Sequence , Animals , Crystallization , Crystallography, X-Ray/methods , Gene Expression Regulation , Molecular Sequence Data , Salivary Proteins and Peptides/genetics
13.
Hum Vaccin Immunother ; 11(6): 1474-88, 2015.
Article in English | MEDLINE | ID: mdl-25905574

ABSTRACT

Over 400 million people living in the world's poorest developing nations are infected with hookworms, mostly of the genus Necator americanus. A bivalent human hookworm vaccine composed of the Necator americanus Glutathione S-Transferase-1 (Na-GST-1) and the Necator americanus Aspartic Protease-1 (Na-APR-1 (M74)) is currently under development by the Sabin Vaccine Institute Product Development Partnership (Sabin PDP). Both monovalent vaccines are currently in Phase 1 trials. Both Na-GST-1 and Na-APR-1 antigens are expressed as recombinant proteins. While Na-GST-1 was found to express with high yields in Pichia pastoris, the level of expression of Na-APR-1 in this host was too low to be suitable for a manufacturing process. When the tobacco plant Nicotiana benthamiana was evaluated as an expression system, acceptable levels of solubility, yield, and stability were attained. Observed expression levels of Na-APR-1 (M74) using this system are ∼300 mg/kg. Here we describe the achievements and obstacles encountered during process development as well as characterization and stability of the purified Na-APR-1 (M74) protein and formulated vaccine. The expression, purification and analysis of purified Na-APR-1 (M74) protein obtained from representative 5 kg reproducibility runs performed to qualify the Na-APR-1 (M74) production process is also presented. This process has been successfully transferred to a pilot plant and a 50 kg scale manufacturing campaign under current Good Manufacturing Practice (cGMP) has been performed. The 50 kg run has provided a sufficient amount of protein to support the ongoing hookworm vaccine development program of the Sabin PDP.


Subject(s)
Antigens, Helminth/isolation & purification , Antigens, Helminth/metabolism , Aspartic Acid Endopeptidases/isolation & purification , Aspartic Acid Endopeptidases/metabolism , Necator americanus/enzymology , Vaccines, Synthetic/isolation & purification , Vaccines, Synthetic/metabolism , Animals , Antigens, Helminth/genetics , Aspartic Acid Endopeptidases/genetics , Biotechnology/methods , Gene Expression , Necator americanus/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Technology, Pharmaceutical/methods , Nicotiana/genetics , Nicotiana/metabolism , Vaccines, Synthetic/genetics
14.
Hum Vaccin Immunother ; 10(7): 1914-25, 2014.
Article in English | MEDLINE | ID: mdl-25424799

ABSTRACT

Infection by the human hookworm Necator americanus is a leading cause of anemia and disability in the developing countries of Africa, Asia, and the Americas. In order to prevent childhood hookworm disease in resource poor settings, a recombinant vaccine is under development by the Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, a Product Development Partnership (PDP). Previously, we reported on the expression and purification of a highly promising hookworm vaccine candidate, Na-GST-1, an N. americanus glutathione s-transferase expressed in Pichia pastoris (yeast), which led to production of 1.5 g of 95% pure recombinant protein at a 20L scale. (1) (,) (2) (,) (3) This yield and purity of Na-GST-1 was sufficient for early pilot manufacturing and initial phase 1 clinical testing. However, based on the number of doses which would be required to allow mass vaccination and a potential goal to deliver a vaccine as inexpensively as possible, a higher yield of expression of the recombinant antigen at the lowest possible cost is highly desirable. Here we report on modifications to the fermentation (upstream process) of the antigen expressed in P. pastoris, and to the purification (downstream process) of the recombinant protein that allowed for a 2-3-fold improvement in the final yield of Na-GST-1 purified protein. The major improvements included upstream process changes such as the addition of a sorbitol pulse and co-feed during methanol induction as well as an extension of the induction stage to approximately 96 hours; downstream process changes included modifying the UFDF to flat sheet with a 10 kDa Molecular Weight cut-off (MWCO), adjusting the capacity of an ion-exchange chromatography step utilizing a gradient elution as opposed to the original step elution, and altering the hydrophobic interaction chromatography conditions. The full process, as well as the purity and stability profiles of the target Na-GST-1, and its formulation on Alhydrogel(®), is described.


Subject(s)
Antigens, Helminth/isolation & purification , Glutathione Transferase/isolation & purification , Hookworm Infections/prevention & control , Necator americanus/enzymology , Technology, Pharmaceutical/methods , Vaccines, Synthetic/isolation & purification , Animals , Antigens, Helminth/genetics , Biotechnology/methods , Chemistry, Pharmaceutical , Chromatography, Liquid/methods , Culture Media/chemistry , Drug Stability , Glutathione Transferase/genetics , Hookworm Infections/immunology , Humans , Necator americanus/immunology , Pichia/genetics , Pichia/growth & development , Pichia/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Transcriptional Activation , Vaccines, Synthetic/genetics
15.
Hum Vaccin Immunother ; 10(3): 648-58, 2014.
Article in English | MEDLINE | ID: mdl-24355931

ABSTRACT

Development of vaccines for preventing a future pandemic of severe acute respiratory syndrome (SARS) caused by SARS coronavirus (SARS-CoV) and for biodefense preparedness is urgently needed. Our previous studies have shown that a candidate SARS vaccine antigen consisting of the receptor-binding domain (RBD) of SARS-CoV spike protein can induce potent neutralizing antibody responses and protection against SARS-CoV challenge in vaccinated animals. To optimize expression conditions for scale-up production of the RBD vaccine candidate, we hypothesized that this could be potentially achieved by removing glycosylation sites in the RBD protein. In this study, we constructed two RBD protein variants: 1) RBD193-WT (193-aa, residues 318-510) and its deglycosylated forms (RBD193-N1, RBD193-N2, RBD193-N3); 2) RBD219-WT (219-aa, residues 318-536) and its deglycosylated forms (RBD219-N1, RBD219-N2, and RBD219-N3). All constructs were expressed as recombinant proteins in yeast. The purified recombinant proteins of these constructs were compared for their antigenicity, functionality and immunogenicity in mice using alum as the adjuvant. We found that RBD219-N1 exhibited high expression yield, and maintained its antigenicity and functionality. More importantly, RBD219-N1 induced significantly stronger RBD-specific antibody responses and a higher level of neutralizing antibodies in immunized mice than RBD193-WT, RBD193-N1, RBD193-N3, or RBD219-WT. These results suggest that RBD219-N1 could be selected as an optimal SARS vaccine candidate for further development.


Subject(s)
Severe acute respiratory syndrome-related coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , Female , Gene Expression , Glycosylation , Mice, Inbred BALB C , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spike Glycoprotein, Coronavirus/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...