Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Nat Commun ; 15(1): 5960, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013877

ABSTRACT

Hookworm infection remains a significant public health concern, particularly in low- and middle-income countries, where mass drug administration has not stopped reinfection. Developing a vaccine is crucial to complement current control measures, which necessitates a thorough understanding of host immune responses. By leveraging controlled human infection models and high-dimensional immunophenotyping, here we investigated the immune remodeling following infection with 50 Necator americanus L3 hookworm larvae in four naïve volunteers over two years of follow-up and compared the profiles with naturally infected populations in endemic areas. Increased plasmacytoid dendritic cell frequency and diminished responsiveness to Toll-like receptor 7/8 ligand were observed in both controlled and natural infection settings. Despite the increased CD45RA+ regulatory T cell (Tregs) frequencies in both settings, markers of Tregs function, including inducible T-cell costimulatory (ICOS), tumor necrosis factor receptor 2 (TNFR2), and latency-associated peptide (LAP), as well as in vitro Tregs suppressive capacity were higher in natural infections. Taken together, this study provides unique insights into the immunological trajectories following a first-in-life hookworm infection compared to natural infections.


Subject(s)
Dendritic Cells , Necator americanus , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , Animals , Dendritic Cells/immunology , Necator americanus/immunology , Male , Adult , Necatoriasis/immunology , Hookworm Infections/immunology , Hookworm Infections/parasitology , Female , Endemic Diseases , Young Adult , Immunophenotyping
2.
PLoS One ; 19(6): e0299022, 2024.
Article in English | MEDLINE | ID: mdl-38829836

ABSTRACT

Controlled Human Infection Models (CHIS) involve administering human pathogens to healthy participants in controlled medical settings, which can elicit complex bioethical issues. Understanding how the community perceives such studies can significantly increase the participant's sense of cooperation and increases the researcher's and the participant's transparency. The current study describes the development of an educational intervention to achieve these ends as it aims to (1) analyze perceptions of the Controlled Human Infection Studies (CHIS), and (2) evaluate the participants' comprehension of the CHIS. METHODS: This is a qualitative action research that includes the development of an educational intervention with residents of a rural area in Minas Gerais, Brazil, where there is continuous natural transmission of the human pathogen Necator americanus ("hookworm"). In this area, it is intended to carry out a proposed phase 3 vaccine clinical trial in the future to test the efficacy of hookworm vaccines using controlled human infection. Two data collection strategies were used: an educational intervention and a focus group. RESULTS: The participants' perceptions showed distinct perspectives on CHIS. On one side, they recognized that the investigation is essential for the community, but on the other side, they thought that there would be resistance to its conduct by fear of infection. The idea that the study would generate a benefit for the greater good, contributing to the prevention of hookworm infection, was clearly stated. The participants perceived that the study offered concrete risks that could be reduced by constant monitoring by the researchers. They also mentioned the importance of access to information and the positive influence those who express interest in participating in the study can exert in the community. In relation to comprehension the participants memorized the information, mobilized it to explain everyday situations and created strategies to disseminate the study and engage the community in its development. By repeating and making sense of the information, the participant not only assimilates the knowledge transmitted, but also creates new knowledge. CONCLUSION: We concluded that an educational process of discussion and dialogue around participants' perceptions about the CHIS, promotes understanding and allows ways to disseminate information about the research to be collectively created.


Subject(s)
Necator americanus , Necatoriasis , Humans , Brazil , Animals , Necator americanus/immunology , Female , Necatoriasis/prevention & control , Necatoriasis/transmission , Necatoriasis/immunology , Male , Adult , Hookworm Infections/prevention & control , Hookworm Infections/transmission , Vaccines/immunology , Middle Aged , Community Participation/methods , Young Adult , Focus Groups
3.
Lancet Infect Dis ; 24(7): 760-774, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38513684

ABSTRACT

BACKGROUND: A human hookworm vaccine is being developed to protect children against iron deficiency and anaemia associated with chronic infection with hookworms. Necator americanus aspartic protease-1 (Na-APR-1) and N americanus glutathione S-transferase-1 (Na-GST-1) are components of the blood digestion pathway critical to hookworm survival in the host. Recombinant Na-GST-1 and catalytically inactive Na-APR-1 (Na-APR-1[M74]) adsorbed to Alhydrogel were safe and immunogenic when delivered separately or co-administered to adults in phase 1 trials in non-endemic and endemic areas. We aimed to investigate the safety and immunogenicity of these antigens in healthy children in a hookworm-endemic area. METHODS: This was a randomised, controlled, observer-blind, phase 1, dose-escalation trial, conducted in a clinical research centre, in 60 children aged six to ten years in Lambaréné, a hookworm-endemic region of Gabon. Healthy children (determined by clinical examination and safety laboratory testing) were randomised 4:1 to receive co-administered Na-GST-1 on Alhydrogel plus Na-APR-1(M74) on Alhydrogel and glucopyranosyl lipid A in aqueous formulation (GLA-AF), or co-administered ENGERIX-B hepatitis B vaccine (HBV) and saline placebo, injected into the deltoid of each arm. Allocation to vaccine groups was observer-masked. In each vaccine group, children were randomised 1:1 to receive intramuscular injections into each deltoid on two vaccine schedules, one at months 0, 2, and 4 or at months 0, 2, and 6. 10 µg, 30 µg, and 100 µg of each antigen were administered in the first, second, and third cohorts, respectively. The intention-to-treat population was used for safety analyses; while for immunogenicity analyses, the per-protocol population was used (children who received all scheduled vaccinations). The primary outcome was to evaluate the vaccines' safety and reactogenicity in healthy children aged between six and ten years. The secondary outcome was to measure antigen-specific serum IgG antibody levels at pre-vaccination and post-vaccination timepoints by qualified ELISAs. The trial is registered with ClinicalTrials.gov, NCT02839161, and is completed. FINDINGS: Between Jan 23 and Oct 3, 2017, 137 children were screened, of whom 76 were eligible for this trial. 60 children were recruited, and allocated to either 10 µg of the co-administered antigens (n=8 for each injection schedule), 30 µg (n=8 for each schedule), 100 µg (n=8 for each schedule), or HBV and placebo (n=6 for each schedule) in three sequential cohorts. Co-administration of the vaccines was well tolerated; the most frequent solicited adverse events were mild-to-moderate injection-site pain, observed in up to 12 (75%) of 16 participants per vaccine group, and mild headache (12 [25%] of 48) and fever (11 [23%] of 48). No vaccine-related serious adverse events were observed. Significant anti-Na-APR-1(M74) and anti-Na-GST-1 IgG levels were induced in a dose-dependent manner, with peaks seen 14 days after the third vaccinations, regardless of dose (for Na-APR-1[M74], geometric mean levels [GML]=2295·97 arbitrary units [AU] and 726·89 AU, while for Na-GST-1, GMLs=331·2 AU and 21·4 AU for the month 0, 2, and 6 and month 0, 2, and 4 schedules, respectively). The month 0, 2, and 6 schedule induced significantly higher IgG responses to both antigens (p=0·01 and p=0·04 for Na-APR-1[M74] and Na-GST-1, respectively). INTERPRETATION: Co-administration of recombinant Na-APR-1(M74) and Na-GST-1 to school-aged Gabonese children was well tolerated and induced significant IgG responses. These results justify further evaluation of this antigen combination in proof-of-concept controlled-infection and efficacy studies in hookworm-endemic areas. FUNDING: European Union Seventh Framework Programme.


Subject(s)
Necator americanus , Humans , Male , Child , Female , Gabon , Necator americanus/immunology , Animals , Hookworm Infections/prevention & control , Hookworm Infections/immunology , Antigens, Helminth/immunology , Antibodies, Helminth/blood , Glutathione Transferase/immunology , Glutathione Transferase/genetics , Single-Blind Method , Vaccines/immunology , Vaccines/administration & dosage , Immunogenicity, Vaccine
4.
Lancet Infect Dis ; 21(2): 275-285, 2021 02.
Article in English | MEDLINE | ID: mdl-32926834

ABSTRACT

BACKGROUND: Hookworms cause substantial morbidity in children and women of reproductive age. The control strategy of mass drug administration is suboptimal, hence the need for a vaccine. Necator americanus aspartic protease-1 (Na-APR-1) and N americanus glutathione S-transferase-1 (Na-GST-1) are involved in the digestion and detoxification of haemoglobin in the hookworm digestive tract. In animal models, vaccination against these antigens resulted in protection from challenge infection. Both vaccine candidates were shown to be safe and well tolerated when administered separately to healthy adults. We assessed the safety and immunogenicity of co-administered Na-GST-1 and Na-APR-1 (M74) vaccines in healthy Gabonese adults. METHODS: This randomised, controlled, double-blind, phase 1, dose-escalation trial was done at the Centre de Recherches Médicales de Lambaréné, in a region of Gabon where N americanus and other helminths are prevalent. Healthy adults aged 18-50 years and living in Lambaréné or the surrounding areas were recruited to the study. Participants were enrolled consecutively into two dose cohorts (30 µg or 100 µg of the experimental vaccines) and randomly assigned in blocks (block size four) to receive three doses of either co-administered Na-GST-1 plus Na-APR-1 (M74; 30 µg or 100 µg of each), adjuvanted with Alhydrogel (aluminium hydroxide gel suspension) together with an aqueous formulation of glucopyranosyl lipid A, or hepatitis B vaccine plus saline (control group). Vaccines were administered intramuscularly on days 0, 28, and 180. The primary endpoint was safety, with immunogenicity a secondary endpoint. The intention-to-treat population was used for safety analyses, whereas for immunogenicity analyses, the per-protocol population was used (participants who received all scheduled vaccinations). Control vaccine recipients for both dose cohorts were combined for the analyses. The trial is registered with ClinicalTrials.gov, NCT02126462. FINDINGS: Between Oct 27, 2014, and Jan 31, 2015, 56 individuals were screened for eligibility, of whom 32 were enrolled and randomly assigned to one of the three study groups (12 each in the 30 µg and 100 µg experimental vaccine groups and eight in the control group). Both study vaccines were well tolerated in both dose groups. The most common adverse events were mild-to-moderate injection-site pain, headache, myalgia, and nausea. No severe or serious adverse events related to the vaccines were recorded. 52 unsolicited vaccine-related adverse events occurred during the study, but there was no difference in frequency between vaccine groups. IgG antibodies were induced to each of the vaccine antigens, with mean IgG levels increasing after each vaccination. Vaccination with 100 µg of each vaccine antigen consistently induced IgG seroconversion (IgG levels above the reactivity threshold). Peak IgG responses were observed 2 weeks after the third vaccine dose for both antigens, with all participants who received the 100 µg doses seroconverting at that timepoint. IgG levels steadily declined until the final study visit 6 months after the third vaccination, although they remained significantly higher than baseline in the 100 µg dose group. INTERPRETATION: Vaccination with recombinant Na-GST-1 and Na-APR-1 (M74) in healthy adults living in N americanus-endemic areas of Gabon was safe and induced IgG to each antigen. To our knowledge, this study is the first to report results of Na-APR-1 (M74) co-administered with Alhydrogel in participants from an N americanus-endemic area. Further clinical development of these vaccines should involve efficacy studies. FUNDING: European Union Seventh Framework Programme.


Subject(s)
Hookworm Infections/prevention & control , Necator americanus/immunology , Vaccines/immunology , Adult , Animals , Antibodies, Helminth/blood , Dose-Response Relationship, Immunologic , Double-Blind Method , Female , Gabon/epidemiology , Hookworm Infections/epidemiology , Humans , Immunoglobulin G/blood , Male , Vaccines/administration & dosage , Young Adult
5.
Gut Microbes ; 12(1): 1-15, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33222610

ABSTRACT

Hookworms are soil-transmitted helminths that use immune-evasive strategies to persist in the human duodenum where they are responsible for anemia and protein loss. Given their location and immune regulatory effects, hookworms likely impact the bacterial microbiota. However, microbiota studies struggle to deconvolute the effect of hookworms from confounders such as coinfections and malnutrition. We thus used an experimental human hookworm infection model to explore temporal changes in the gut microbiota before and during hookworm infection. Volunteers were dermally exposed to cumulative dosages of 50, 100 or 150 L3 Necator americanus larvae. Fecal samples were collected for microbiota profiling through 16S rRNA gene amplicon sequencing at weeks zero, four, eight, fourteen and twenty. During the acute infection phase (trial week zero to eight) no changes in bacterial diversity were detected. During the established infection phase (trial week eight to twenty), bacterial richness (Chao1, p = .0174) increased significantly over all volunteers. No relation was found between larval dosage and diversity, stability or relative abundance of individual bacterial taxa. GI symptoms were associated with an unstable microbiota during the first eight weeks and rapid recovery at week twenty. Barnesiella, amongst other taxa, was more abundant in volunteers with more GI symptoms throughout the study. In conclusion, this study showed that clinical GI symptoms following N. americanus infection are associated with temporary microbiota instability and relative abundance of specific bacterial taxa. These results suggest a possible role of hookworm-induced enteritis on microbiota stability.


Subject(s)
Bacteria/classification , Bacteria/isolation & purification , Gastrointestinal Microbiome/physiology , Necator americanus/immunology , Necatoriasis/immunology , Adult , Animals , Bacteria/genetics , Enteritis/microbiology , Enteritis/parasitology , Female , Humans , Male , Middle Aged , Necator americanus/embryology , Necator americanus/genetics , RNA, Ribosomal, 16S/genetics , Young Adult
6.
Parasitology ; 145(9): 1183-1190, 2018 08.
Article in English | MEDLINE | ID: mdl-29455681

ABSTRACT

Despite the profound health implications of Necator americanus infection in humans, many aspects of its interaction with the host immune system are poorly understood. Here we investigated the early events at the interface of N. americanus larvae (L3) and human dendritic cells (DCs). Our data show that co-culturing DCs and the larvae trigger ex-sheathing of hookworms rapidly where a majority of DCs are sequestered onto the larval sheath allowing the ex-sheathed larvae to migrate away unchallenged. Intriguingly, DCs show negligible interaction with the ex-sheathed larvae, alluding to differences between the surface chemistry of the larva and its sheath. Furthermore, blocking of two key C-type lectin receptors on DC surface (i.e. DC-SIGN and mannose receptor) resulted in inhibition of ex-sheathing process and DC sequestration, highlighting the importance of C-type lectins on DCs in the induction of the ex-sheathing. Analyses of DC phenotype and cytokine profile after co-culture with the N. americanus larvae showed an immature phenotype as evidenced by the low expression of the maturation markers and cytokines. These data provide new insights into early events at the interface of human DCs and N. americanus larvae and could explain how L3 evade immune recognition upon initial interaction with DCs.


Subject(s)
Dendritic Cells/immunology , Host-Parasite Interactions/immunology , Immune Evasion , Larva/physiology , Necator americanus/physiology , Animals , Antigens, Helminth/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cells, Cultured , Cytokines/immunology , Dendritic Cells/parasitology , Humans , Larva/immunology , Lectins, C-Type/antagonists & inhibitors , Mannose Receptor , Mannose-Binding Lectins/antagonists & inhibitors , Necator americanus/immunology , Necatoriasis/immunology , Receptors, Cell Surface/antagonists & inhibitors
7.
Parasite Immunol ; 39(5)2017 05.
Article in English | MEDLINE | ID: mdl-27977856

ABSTRACT

Helminth infections likely provide a protective influence against some immune-mediated and metabolic diseases because helminth infection dramatically decreased in developed countries shortly before the explosive rise in the prevalence of these diseases. The capacity of helminths to activate immune-regulatory circuits in their hosts and to modulate the composition of intestinal flora appears to be the mechanisms of protective action. Animal models of disease show that various helminth species prevent and/or block inflammation in various organs in a diverse range of diseases. Clinical trials have demonstrated that medicinal exposure to Trichuris suis or small numbers of Necator americanus is safe with minor, if any, reported adverse effects. This includes exposure of inflamed intestine to T. suis, asthmathic lung to N. americanus and in patients with atopy. Efficacy has been suggested in some small studies, but is absent in others. Factors that may have led to inconclusive results in some trials are discussed. To date, there have been no registered clinical trials using helminths to treat metabolic syndrome or its component conditions. However, the excellent safety profile of T. suis or N. americanus suggests that such studies should be possible.


Subject(s)
Helminthiasis/immunology , Inflammation/therapy , Therapy with Helminths , Animals , Humans , Necator americanus/immunology , Trichuris/immunology
8.
Nat Rev Dis Primers ; 2: 16088, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929101

ABSTRACT

Hookworms are soil-transmitted nematode parasites that can reside for many years in the small intestine of their human hosts; Necator americanus is the predominant infecting species. Adult worms feed on the blood of a host and can cause iron deficiency anaemia, especially in high-risk populations (children and women of childbearing age). Almost 500 million people in developing tropical countries are infected, and simulation models estimate that hookworm infection is responsible for >4 million disability-adjusted life years lost annually. Humans mount an immune response to hookworms, but it is mostly unsuccessful at removing adult worms from the bowel. Accordingly, the host switches to an immune-tolerant state that enables hookworms to reside in the gut for many years. Although anthelmintic drugs are available and widely used, their efficacy varies and the drugs do not prevent reinfection. Thus, other control strategies aimed at improving water quality, sanitation and hygiene are needed. In addition, efforts are underway to develop a human hookworm vaccine through public-private partnerships. However, hookworms could also be a resource; as hookworms have the capability to regulate the host's inflammation, researchers are experimentally infecting patients to treat some inflammatory diseases as an approach to discover new anti-inflammatory molecules. This area of endeavour might well yield new biotherapeutics for autoimmune and allergic diseases.


Subject(s)
Hookworm Infections/complications , Hookworm Infections/physiopathology , Albendazole/pharmacology , Albendazole/therapeutic use , Ancylostomatoidea/immunology , Ancylostomatoidea/pathogenicity , Anemia/complications , Anemia/etiology , Animals , Anthelmintics/pharmacology , Anthelmintics/therapeutic use , Factor VIIa/adverse effects , Factor XIa/adverse effects , Factor Xa/adverse effects , Feces/parasitology , Hemorrhage/etiology , Hemorrhage/parasitology , Hookworm Infections/epidemiology , Humans , Intestine, Small/parasitology , Intestine, Small/physiopathology , Larva Migrans/etiology , Mebendazole/pharmacology , Mebendazole/therapeutic use , Necator americanus/immunology , Necator americanus/pathogenicity , Prevalence , Risk Factors , Soil/parasitology
9.
Methods Mol Biol ; 1403: 639-53, 2016.
Article in English | MEDLINE | ID: mdl-27076158

ABSTRACT

A protein-based vaccine approach against hookworm infection has failed to deliver the expected outcome, due to a problem with an allergic response in the patient or difficulties in the proteins' production. This implication could be overcome by using a chemically synthesized peptide-based vaccine approach. This approach utilizes minimal pathogenic components that are necessary for the stimulation of the immune response without triggering adverse side effects. To boost the peptide's immunogenicity, a lipid core peptide (LCP) system can be utilized as a carrier molecule/immunostimulant. This chapter describes in detail the synthesizing of protected lipoamino acid, the self-adjuvanting moiety (LCP core), the peptide epitope, and the final vaccine candidate. The subunit peptide and the LCP core were synthesized using microwave-assisted solid-phase peptide synthesis (SPPS). Then the final hookworm vaccine construct was assembled using the copper-catalyzed azide-alkyne cycloaddition, or "click," reaction.


Subject(s)
Click Chemistry/methods , Hookworm Infections/prevention & control , Microwaves , Solid-Phase Synthesis Techniques/methods , Vaccines/chemical synthesis , Animals , Epitopes/immunology , Necator americanus/immunology , Palmitic Acid/chemistry , Vaccines/chemistry , Vaccines/immunology , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
10.
Immunology ; 148(2): 115-24, 2016 06.
Article in English | MEDLINE | ID: mdl-26928141

ABSTRACT

Cellular and molecular investigation of parasitic helminth infections has greatly accelerated the understanding of type 2 immune responses. However, there remains considerable debate regarding the specific leucocytes that kill parasites and whether these mechanisms are distinct from those responsible for tissue repair. Herein, we chronicle discoveries over the past decade highlighting current paradigms in type 2 immunity with a particular emphasis upon how CD4(+) T helper type 2 cells, type 2 innate lymphoid cells and alternatively activated macrophages coordinately control helminth-induced parasitism. Primarily, this review will draw from studies of the murine nematode parasite Nippostrongylus brasiliensis, which bears important similarities to the human hookworms Ancylostoma duodenale and Necator americanus. Given that one or more hookworm species currently infect millions of individuals across the globe, we propose that vaccine and/or pharmaceutical-based cure strategies targeting these affected human populations should incorporate the conceptual advances outlined herein.


Subject(s)
Ancylostoma/immunology , Ancylostomiasis/immunology , Macrophages/immunology , Necator americanus/immunology , Necatoriasis/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Antigens, Helminth/immunology , Cell Differentiation , Complement Pathway, Alternative , Humans , Immunity, Innate , Macrophage Activation , Macrophages/parasitology , Th2 Cells/parasitology
11.
ChemMedChem ; 10(10): 1647-54, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269385

ABSTRACT

Necator americanus (hookworm) infects over half a billion people worldwide. Anthelminthic drugs are commonly used to treat the infection; however, vaccination is a more favorable strategy to combat this parasite. We designed new B-cell peptide epitopes based on the aspartic protease of N. americanus (Na-APR-1). The peptides were conjugated to self-adjuvanting lipid core peptide (LCP) systems via stepwise solid-phase peptide synthesis (SPPS) and copper catalyst azide-alkyne cycloaddition (CuAAC) reactions. The LCP vaccine candidates were able to self-assemble into nanoparticles, were administered to mice without the use of additional adjuvant, and generated antibodies that recognized the parent epitope. However, only one LCP derivative was able to produce a high titer of antibodies specific to Na-APR-1; circular dichroism analyses of this compound showed a ß-sheet conformation for the incorporated epitope. This study provides important insight in epitope and delivery system design for the development of a vaccine against hookworm infections.


Subject(s)
Aspartic Acid Proteases/immunology , Hookworm Infections/parasitology , Lipopeptides/immunology , Nanoparticles/chemistry , Necator americanus/immunology , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , Animals , Antibodies, Helminth/immunology , Aspartic Acid Proteases/chemistry , Female , Hookworm Infections/immunology , Lipopeptides/chemistry , Mice , Mice, Inbred BALB C , Molecular Conformation , Necator americanus/enzymology , Particle Size , Structure-Activity Relationship
12.
J Allergy Clin Immunol ; 135(2): 508-16, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25248819

ABSTRACT

BACKGROUND: Celiac disease (CeD) is a common gluten-sensitive autoimmune enteropathy. A gluten-free diet is an effective treatment, but compliance is demanding; hence, new treatment strategies for CeD are required. OBJECTIVE: Parasitic helminths hold promise for treating inflammatory disorders, so we examined the influence of experimental hookworm infection on the predicted outcomes of escalating gluten challenges in CeD subjects. METHODS: A 52-week study was conducted involving 12 adults with diet-managed CeD. Subjects were inoculated with 20 Necator americanus larvae, and escalating gluten challenges consumed as pasta were subsequently administered: (1) 10 to 50 mg for 12 weeks (microchallenge); (2) 25 mg daily + 1 g twice weekly for 12 weeks (GC-1g); and (3) 3 g daily (60-75 straws of spaghetti) for 2 weeks (GC-3g). Symptomatic, serologic, and histological outcomes evaluated gluten toxicity. Regulatory and inflammatory T cell populations in blood and mucosa were examined. RESULTS: Two gluten-intolerant subjects were withdrawn after microchallenge. Ten completed GC-1g, 8 of whom enrolled in and completed GC-3g. PRIMARY OUTCOMES: median villous height-to-crypt depth ratios (2.60-2.63; P = .98) did not decrease as predicted after GC-1g, and the mean IgA-tissue transglutaminase titers declined, contrary to the predicted rise after GC-3g. SECONDARY OUTCOMES: quality of life scores improved (46.3-40.6; P = .05); celiac symptom indices (24.3-24.3; P = .53), intra-epithelial lymphocyte percentages (32.5-35.0; P = .47), and Marsh scores were unchanged by gluten challenge. Intestinal T cells expressing IFNγ were reduced following hookworm infection (23.9%-11.5%; P = .04), with corresponding increases in CD4(+) Foxp3(+) regulatory T cells (0.19%-1.12%; P = .001). CONCLUSIONS: Necator americanus and gluten microchallenge promoted tolerance and stabilized or improved all tested indices of gluten toxicity in CeD subjects.


Subject(s)
Ancylostomatoidea/immunology , Celiac Disease/immunology , Glutens/immunology , Hookworm Infections/immunology , Immune Tolerance , Adult , Aged , Animals , Celiac Disease/complications , Celiac Disease/diagnosis , Celiac Disease/therapy , Duodenum/immunology , Duodenum/parasitology , Duodenum/pathology , Female , Glutens/administration & dosage , Hookworm Infections/complications , Humans , Immunophenotyping , Male , Middle Aged , Necator americanus/immunology , Patient Outcome Assessment , Surveys and Questionnaires , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
13.
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
14.
Am J Trop Med Hyg ; 90(4): 661-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24493672

ABSTRACT

We explored the practicality of integrating surveillance for soil-transmitted helminthiasis (STH, assessed by Kato-Katz) with transmission assessment surveys for lymphatic filariasis (LF) in two evaluation units (EUs) in Gampaha district, Sri Lanka (population 2.3 million). The surveys were performed 6 years after five annual rounds of mass drug administration with diethylcarbamazine and albendazole. Each transmission assessment survey tested children (N = 1,462 inland EU; 1,642 coastal EU) sampled from 30 primary schools. Low filarial antigenemia rates (0% and 0.1% for the inland and coastal EUs) suggest that LF transmission is very low in this district. The STH rates and stool sample participation rates were 0.8% and 61% (inland) and 2.8% and 58% (coastal). Most STH detected were low or moderate intensity Trichuris trichiura infections. The added cost of including STH testing was ∼$5,000 per EU. These results suggest that it is feasible to integrate school-based surveillance for STH and LF.


Subject(s)
Antigens, Helminth/immunology , Elephantiasis, Filarial/epidemiology , Epidemiological Monitoring , Helminthiasis/epidemiology , School Health Services , Animals , Ascariasis/epidemiology , Ascariasis/immunology , Ascariasis/transmission , Ascaris lumbricoides/immunology , Child , Elephantiasis, Filarial/immunology , Elephantiasis, Filarial/transmission , Feasibility Studies , Feces/parasitology , Helminthiasis/immunology , Helminthiasis/transmission , Humans , Necator americanus/immunology , Necatoriasis/epidemiology , Necatoriasis/immunology , Necatoriasis/transmission , Parasite Egg Count , School Health Services/economics , Soil/parasitology , Sri Lanka/epidemiology , Trichuriasis/epidemiology , Trichuriasis/immunology , Trichuriasis/transmission , Trichuris/immunology , Wuchereria bancrofti/immunology
15.
Nat Genet ; 46(3): 261-269, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24441737

ABSTRACT

The hookworm Necator americanus is the predominant soil-transmitted human parasite. Adult worms feed on blood in the small intestine, causing iron-deficiency anemia, malnutrition, growth and development stunting in children, and severe morbidity and mortality during pregnancy in women. We report sequencing and assembly of the N. americanus genome (244 Mb, 19,151 genes). Characterization of this first hookworm genome sequence identified genes orchestrating the hookworm's invasion of the human host, genes involved in blood feeding and development, and genes encoding proteins that represent new potential drug targets against hookworms. N. americanus has undergone a considerable and unique expansion of immunomodulator proteins, some of which we highlight as potential treatments against inflammatory diseases. We also used a protein microarray to demonstrate a postgenomic application of the hookworm genome sequence. This genome provides an invaluable resource to boost ongoing efforts toward fundamental and applied postgenomic research, including the development of new methods to control hookworm and human immunological diseases.


Subject(s)
Genome, Helminth , Necator americanus/genetics , Animals , Caenorhabditis elegans/genetics , Female , Gene Expression Regulation, Developmental , Host-Parasite Interactions/immunology , Humans , Male , Molecular Sequence Data , Necator americanus/growth & development , Necator americanus/immunology , Necatoriasis/immunology , Necatoriasis/parasitology , Necatoriasis/prevention & control , Pregnancy , Species Specificity
16.
Int J Parasitol ; 43(3-4): 275-82, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291460

ABSTRACT

We recently completed clinical trials in people with diet-treated celiac disease who were purposefully infected with the ubiquitous human hookworm, Necator americanus. Hookworm infection elicited not only parasite-specific immunity but also modified the host's immune response to gluten. After infection, mucosal IL-1ß and IL-22 responses were enhanced, but IFNγ and IL-17A levels and circulating regulatory T cells following gluten challenge were suppressed, and the adaptive response to gluten acquired a helper T cell type-2 profile. In this review, we briefly, (i) highlight the utility celiac disease offers autoimmune research, (ii) discuss safety and personal experience with N. americanus, (iii) summarise the direct and bystander impact that hookworm infection has on mucosal immunity to the parasite and gluten, respectively, and (iv) speculate why this hookworm's success depends on healing its host and how this might impact on a propensity to autoimmunity.


Subject(s)
Autoimmunity , Celiac Disease/immunology , Glutens/immunology , Necator americanus/immunology , Animals , Celiac Disease/parasitology , Celiac Disease/therapy , Humans , Necator americanus/physiology , Therapy with Helminths
17.
Immunobiology ; 218(5): 706-11, 2013 May.
Article in English | MEDLINE | ID: mdl-22999162

ABSTRACT

According to the hygiene hypothesis, reduced exposure to infections could explain the rise of atopic diseases in high-income countries. Helminths are hypothesised to alter the host's immune response in order to avoid elimination and, as a consequence, also reduce the host responsiveness to potential allergens. To elucidate the effect of current helminth infections on immune responsiveness in humans, we measured cytokine production in a rural Ghanaian population in an area with multiple endemic parasites including malaria, intestinal helminths and protozoa. Multiplex real-time PCR in stool samples was used for the detection of four gastrointestinal helminths, of which only Necator americanus was commonly present. A similar assay was used to test for Giardia lamblia in stool samples and malaria infection in venous blood samples. Levels of the cytokines interleukin (IL)-10, tumour necrosis factor (TNF)-α, IL-17, IL-6, IL-13, and interferon (IFN)-γ were determined in whole-blood samples ex vivo-stimulated either with lipopolysaccharide (LPS) and zymosan (for innate cytokine production) or the T-cell mitogen phytohaemagglutinin (PHA). There were no significant differences in either innate or PHA-stimulated cytokine production dependent on current N. americanus infection. Plasmodium falciparum malarial infection was associated with a pro-inflammatory response indicated by increased innate production of TNF-α, IL-17 and IL-6. There was no clear pattern in cytokine responses dependent on G. lamblia-infection. In conclusion, in this rural Ghanaian population current N. americanus infections are not associated with altered immune function, while infection with P. falciparum is associated with pro-inflammatory innate immune responses.


Subject(s)
Endemic Diseases , Giardiasis/immunology , Helminthiasis/immunology , Immunity, Innate , Malaria, Falciparum/immunology , Adult , Aged , Animals , Coinfection , Cytokines/blood , Cytokines/immunology , Feces/microbiology , Feces/parasitology , Female , Ghana/epidemiology , Giardia lamblia/immunology , Giardiasis/blood , Giardiasis/epidemiology , Giardiasis/parasitology , Helminthiasis/blood , Helminthiasis/epidemiology , Helminthiasis/parasitology , Humans , Malaria, Falciparum/blood , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Male , Middle Aged , Necator americanus/immunology , Plasmodium falciparum/immunology , Rural Population
18.
PLoS One ; 7(10): e46870, 2012.
Article in English | MEDLINE | ID: mdl-23056500

ABSTRACT

Hookworms infect more people than HIV and malaria combined, predominantly in third world countries. Treatment of infection with chemotherapy can have limited efficacy and re-infections after treatment are common. Heavy infection often leads to debilitating diseases. All these factors suggest an urgent need for development of vaccine. In an attempt to develop a vaccine targeting the major human hookworm, Necator americanus, a B-cell peptide epitope was chosen from the apical enzyme in the hemoglobin digestion cascade, the aspartic protease Na-APR-1. The A(291)Y alpha helical epitope is known to induce neutralizing antibodies that inhibit the enzymatic activity of Na-APR-1, thus reducing the capacity for hookworms to digest hemoglobin and obtain nutrients. A(291)Y was engineered such that it was flanked on both termini by a coil-promoting sequence to maintain native conformation, and subsequently incorporated into a Lipid Core Peptide (LCP) self-adjuvanting system. While A(291)Y alone or the chimeric epitope with or without Freund's adjuvants induced negligible IgG responses, the LCP construct incorporating the chimeric peptide induced a strong IgG response in mice. Antibodies produced were able to bind to and completely inhibit the enzymatic activity of Na-APR-1. The results presented show that the new chimeric LCP construct can induce effective enzyme-neutralising antibodies in mice, without the help of any additional toxic adjuvants. This approach offers promise for the development of vaccines against helminth parasites of humans and their livestock and companion animals.


Subject(s)
Hookworm Infections/prevention & control , Vaccines, Subunit/immunology , Amino Acid Sequence , Animals , Antibodies, Helminth/immunology , Aspartic Acid Proteases/immunology , Drug Design , Epitopes/immunology , Female , Humans , Mice , Necator americanus/immunology , Protein Structure, Secondary , Vaccines, Subunit/chemical synthesis , Vaccines, Subunit/chemistry
19.
Parasite Immunol ; 34(8-9): 404-11, 2012.
Article in English | MEDLINE | ID: mdl-22651670

ABSTRACT

Na-ASP-2 is a major protein secreted by infective third-stage larvae (L3) of the human hookworm Necator americanus upon host entry. It was chosen as a lead vaccine candidate for its ability to elicit protective immune responses. However, clinical development of this antigen as a recombinant vaccine was halted because it caused allergic reactions among some of human volunteers previously infected with N. americanus. To prevent IgE-mediated allergic reactions induced by Na-ASP-2 but keep its immunogenicity as a vaccine antigen, we designed and tested a genetically engineered fusion protein, Fcγ/Na-ASP-2, composed of full-length Na-ASP-2 and truncated human IgG Fcγ1 that targets the negative signalling receptor FcγRIIb expressed on pro-allergic cells. The chimeric recombinant Fcγ/Na-ASP-2 protein was expressed in Pichia pastoris and shared the similar antigenicity as native Na-ASP-2. Compared to Na-ASP-2, the chimeric fusion protein efficiently reduced the release of histamine in human basophils sensitized with anti-Na-ASP-2 IgE obtained from individuals living in a hookworm-endemic area. In dogs infected with canine hookworm, Fcγ/Na-ASP-2 resulted in significantly reduced immediate-type skin reactivity when injected intradermally compared with Na-ASP-2. Hamsters vaccinated with Fcγ/Na-ASP-2 formulated with Alhydrogel(®) produced specific IgG that recognized Na-ASP-2 and elicited similar protection level against N. americanus L3 challenge as native Na-ASP-2.


Subject(s)
Basophils/immunology , Histamine Release , Immunization , Immunoglobulin E/immunology , Immunoglobulin Fc Fragments/immunology , Necator americanus/immunology , Vaccination/methods , Animals , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Cricetinae , Dogs , Gene Expression , Humans , Hypersensitivity/prevention & control , Immunoglobulin Fc Fragments/genetics , Immunoglobulins , Pichia/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Skin/pathology , Vaccination/adverse effects
20.
J Allergy Clin Immunol ; 130(1): 169-76.e6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22633322

ABSTRACT

BACKGROUND: Necator americanus Ancylostoma-secreted protein 2 (Na-ASP-2) is secreted by infective hookworm larvae on entry into human hosts. Vaccination of laboratory animals with recombinant Na-ASP-2 provides significant protection against challenge infections. In endemic areas antibodies to Na-ASP-2 are associated with reduced risk of heavy N americanus infections. OBJECTIVE: To assess the safety and immunogenicity of recombinant Na-ASP-2 adjuvanted with Alhydrogel in healthy Brazilian adults previously infected with N americanus. METHODS: Participants were randomized to receive Na-ASP-2 or hepatitis B vaccine. Major IgG and IgE epitopes of the Na-ASP-2 molecule were mapped by using sera from these same subjects. Seroepidemiologic studies in adults and children residing in hookworm-endemic areas were conducted to assess the prevalence of IgE responses to Na-ASP-2. RESULTS: Vaccination with a single dose of Na-ASP-2 resulted in generalized urticarial reactions in several volunteers. These reactions were associated with pre-existing Na-ASP-2-specific IgE likely induced by previous hookworm infection. Surveys revealed that a significant proportion of the population in hookworm-endemic areas had increased levels of IgE to Na-ASP-2. Epitope mapping demonstrated sites on the Na-ASP-2 molecule that are uniquely or jointly recognized by IgG and IgE antibodies. CONCLUSION: Infection with N americanus induces increased levels of total and specific IgE to Na-ASP-2 that result in generalized urticaria on vaccination with recombinant Na-ASP-2. These data advance knowledge of vaccine development for helminths given their propensity to induce strong T(H)2 responses. Study data highlight the important differences between the immune responses to natural helminth infection and to vaccination with a recombinant helminth antigen.


Subject(s)
Antigens, Helminth/adverse effects , Helminth Proteins/adverse effects , Necator americanus/immunology , Necatoriasis/prevention & control , Urticaria/epidemiology , Vaccines, Synthetic/adverse effects , Adolescent , Adult , Animals , Antigens, Helminth/administration & dosage , Antigens, Helminth/immunology , Brazil/epidemiology , Epitope Mapping , Female , Helminth Proteins/administration & dosage , Helminth Proteins/immunology , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Male , Middle Aged , Necatoriasis/epidemiology , Necatoriasis/immunology , Seroepidemiologic Studies , Treatment Outcome , Urticaria/etiology , Vaccination/adverse effects , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...