Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(11)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687615

ABSTRACT

A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.


Subject(s)
Immunity, Innate , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Sporozoites , Vaccines, Attenuated , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Immunity, Innate/immunology , Humans , Animals , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Mice , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Sporozoites/immunology , Sporozoites/radiation effects , CD8-Positive T-Lymphocytes/immunology , Infant , Protozoan Proteins/immunology , Antibodies, Protozoan/immunology , Female , Parasitemia/immunology , Parasitemia/prevention & control , Immunoglobulin G/immunology , Immunoglobulin G/blood , Vaccine Efficacy
2.
Cell Rep ; 35(2): 108996, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852850

ABSTRACT

Antibodies targeting the NANP/NVDP repeat domain of the Plasmodium falciparum circumsporozoite protein (CSPRepeat) can protect against malaria. However, it has also been suggested that the CSPRepeat is a decoy that prevents the immune system from mounting responses against other domains of CSP. Here, we show that, following parasite immunization, B cell responses to the CSPRepeat are immunodominant over responses to other CSP domains despite the presence of similar numbers of naive B cells able to bind these regions. We find that this immunodominance is driven by avid binding of the CSPRepeat to cognate B cells that are able to expand at the expense of B cells with other specificities. We further show that mice immunized with repeat-truncated CSP molecules develop responses to subdominant epitopes and are protected against malaria. These data demonstrate that the CSPRepeat functions as a decoy, but truncated CSP molecules may be an approach for malaria vaccination.


Subject(s)
Antibodies, Protozoan/biosynthesis , Immunization/methods , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Peptides/administration & dosage , Plasmodium berghei/drug effects , Protozoan Proteins/genetics , Animals , Anopheles/parasitology , Antibodies, Neutralizing/biosynthesis , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Female , Gene Expression , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/biosynthesis , Malaria Vaccines/genetics , Mice , Mice, Inbred C57BL , Peptides/genetics , Peptides/immunology , Plasmodium berghei/immunology , Plasmodium berghei/pathogenicity , Plasmodium falciparum/drug effects , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protein Binding , Protozoan Proteins/immunology , Sporozoites/immunology , Sporozoites/radiation effects , Transgenes , Vaccines, Attenuated
3.
PLoS One ; 15(6): e0233840, 2020.
Article in English | MEDLINE | ID: mdl-32555601

ABSTRACT

BACKGROUND: Immunization with radiation-attenuated sporozoites (RAS) by mosquito bite provides >90% sterile protection against Plasmodium falciparum (Pf) malaria in humans. RAS invade hepatocytes but do not replicate. CD8+ T cells recognizing parasite-derived peptides on the surface of infected hepatocytes are likely the primary protective mechanism. We conducted a randomized clinical trial of RAS immunization to assess safety, to achieve 50% vaccine efficacy (VE) against controlled human malaria infection (CHMI), and to generate reagents from protected and non-protected subjects for future identification of protective immune mechanisms and antigens. METHODS: Two cohorts (Cohort 1 and Cohort 2) of healthy, malaria-naïve, non-pregnant adults age 18-50 received five monthly immunizations with infected (true-immunized, n = 21) or non-infected (mock-immunized, n = 5) mosquito bites and underwent homologous CHMI at 3 weeks. Immunization parameters were selected for 50% protection based on prior clinical data. Leukapheresis was done to collect plasma and peripheral blood mononuclear cells. RESULTS: Adverse event rates were similar in true- and mock-immunized subjects. Two true- and two mock-immunized subjects developed large local reactions likely caused by mosquito salivary gland antigens. In Cohort 1, 11 subjects received 810-1235 infected bites; 6/11 (55%) were protected against CHMI vs. 0/3 mock-immunized and 0/6 infectivity controls (VE 55%). In Cohort 2, 10 subjects received 839-1131 infected bites with a higher first dose and a reduced fifth dose; 9/10 (90%) were protected vs. 0/2 mock-immunized and 0/6 controls (VE 90%). Three/3 (100%) protected subjects administered three booster immunizations were protected against repeat CHMI vs. 0/6 controls (VE 100%). Cohort 2 uniquely showed a significant rise in IFN-γ responses after the third and fifth immunizations and higher antibody responses to CSP. CONCLUSIONS: PfRAS were generally safe and well tolerated. Cohort 2 had a higher first dose, reduced final dose, higher antibody responses to CSP and significant rise of IFN-γ responses after the third and fifth immunizations. Whether any of these factors contributed to increased protection in Cohort 2 requires further investigation. A cryobank of sera and cells from protected and non-protected individuals was generated for future immunological studies and antigen discovery. TRIAL REGISTRATION: ClinicalTrials.gov NCT01994525.


Subject(s)
Insect Bites and Stings/immunology , Malaria/prevention & control , Sporozoites/immunology , Vaccination/methods , Vaccines, Attenuated/adverse effects , Adult , Animals , Anopheles/parasitology , Anopheles/physiology , Female , Gamma Rays , Humans , Malaria/immunology , Male , Middle Aged , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/immunology , Sporozoites/pathogenicity , Sporozoites/radiation effects , Vaccination/adverse effects
4.
Front Immunol ; 11: 190, 2020.
Article in English | MEDLINE | ID: mdl-32153565

ABSTRACT

Plasmodium spp.-infected mosquitos inject sporozoites into the skin of a mammalian host during a blood meal. These enter the host's circulatory system and establish an infection in the liver. After a silent metamorphosis, merozoites invade the blood leading to the symptomatic and transmissible stages of malaria. The silent pre-erythrocytic malaria stage represents a bottleneck in the disease which is ideal to block progression to clinical malaria, through chemotherapeutic and immunoprophylactic interventions. RTS,S/AS01, the only malaria vaccine close to licensure, although with poor efficacy, blocks the sporozoite invasion mainly through the action of antibodies against the CSP protein, a major component of the pellicle of the sporozoite. Strikingly, sterile protection against malaria can be obtained through immunization with radiation-attenuated sporozoites, genetically attenuated sporozoites or through chemoprophylaxis with infectious sporozoites in animals and humans, but the deployability of sporozoite-based live vaccines pose tremendous challenges. The protection induced by sporozoites occurs in the pre-erythrocytic stages and is mediated mainly by antibodies against the sporozoite and CD8+ T cells against peptides presented by MHC class I molecules in infected hepatocytes. Thus, the identification of malaria antigens expressed in the sporozoite and liver-stage may provide new vaccine candidates to be included, alone or in combination, as recombinant protein-based, virus-like particles or sub-unit virally-vectored vaccines. Here I review the efforts being made to identify Plasmodium falciparum antigens expressed during liver-stage with focus on the development of parasite, hepatocyte, mouse models, and resulting rate of infection in order to identify new vaccine candidates and to improve the efficacy of the current vaccines. Finally, I propose new approaches for the identification of liver-stage antigens based on immunopeptidomics.


Subject(s)
Antigens, Protozoan/immunology , Erythrocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Erythrocytes/parasitology , Hepatocytes/immunology , Hepatocytes/parasitology , Humans , Immunization/methods , Liver/immunology , Liver/parasitology , Malaria, Falciparum/parasitology , Mice , Sporozoites/immunology , Sporozoites/radiation effects , Transcriptome , Vaccines, Attenuated
5.
Malar J ; 19(1): 56, 2020 Feb 03.
Article in English | MEDLINE | ID: mdl-32013956

ABSTRACT

Worldwide strategies between 2010 and 2017 aimed at controlling malarial parasites (mainly Plasmodium falciparum) led to a reduction of just 18% regarding disease incidence rates. Many biologically-derived anti-malarial vaccine candidates have been developed to date; this has involved using many experimental animals, an immense amount of work and the investment of millions of dollars. This review provides an overview of the current state and the main results of clinical trials for sporozoite-targeting vaccines (i.e. the parasite stage infecting the liver) carried out by research groups in areas having variable malaria transmission rates. However, none has led to promising results regarding the effective control of the disease, thereby making it necessary to complement such efforts at finding/introducing new vaccine candidates by adopting a multi-epitope, multi-stage approach, based on minimal subunits of the main sporozoite proteins involved in the invasion of the liver.


Subject(s)
Malaria Vaccines , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Animals , Anopheles/parasitology , Erythrocytes/parasitology , Humans , Liver/parasitology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/transmission , Mosquito Vectors/parasitology , Plasmodium falciparum/growth & development , Sporozoites/immunology , Sporozoites/radiation effects , Vaccines, Attenuated , Vaccines, Subunit , Vaccines, Synthetic
6.
Sci Rep ; 9(1): 13436, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530862

ABSTRACT

Given the number of global malaria cases and deaths, the need for a vaccine against Plasmodium falciparum (Pf) remains pressing. Administration of live, radiation-attenuated Pf sporozoites can fully protect malaria-naïve individuals. Despite the fact that motility of these attenuated parasites is key to their infectivity and ultimately protective efficacy, sporozoite motility in human tissue (e.g. skin) remains wholly uncharacterized to date. We show that the ability to quantitatively address the complexity of sporozoite motility in human tissue provides an additional tool in the development of attenuated sporozoite vaccines. We imaged Pf movement in the skin of its natural host and compared wild-type and radiation-attenuated GFP-expressing Pf sporozoites. Using custom image analysis software and human skin explants we were able to quantitatively study their key motility features. This head-to-head comparison revealed that radiation attenuation impaired the capacity of sporozoites to vary their movement angle, velocity and direction, promoting less refined movement patterns. Understanding and overcoming these changes in motility will contribute to the development of an efficacious attenuated parasite malaria vaccine.


Subject(s)
Plasmodium falciparum/radiation effects , Skin/parasitology , Sporozoites/pathogenicity , Sporozoites/radiation effects , Animals , Anopheles/parasitology , Green Fluorescent Proteins/genetics , Host-Parasite Interactions , Humans , Image Processing, Computer-Assisted , Organisms, Genetically Modified , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Software
7.
Malar J ; 18(1): 186, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31142328

ABSTRACT

BACKGROUND: Whole parasite vaccination is an efficacious strategy to induce sterile immunity and to prevent malaria transmission. Understanding the mechanism and response of immune cells to vaccines plays a critical role in deciphering correlates of protection against infection and disease. Immunoassays, such as ELISpot, are commonly used to assess the immunogenicity of vaccines towards T cells and B cells. To date, these assays only analyse responses to specific antigens since they are based on recombinant parasite-derived proteins or peptides. There is the need for an agnostic approach that allows the evaluation of all sporozoite-associated antigens. METHODS: ELISpot plates coated with a defined amount of lysed Plasmodium falciparum sporozoites were used to assess the frequency of sporozoite-specific B cells in peripheral blood mononuclear cells from donors immunized with either a recombinant malaria vaccine or irradiated sporozoites. RESULTS: This report describes the assay conditions for a specific and sensitive sporozoite-based B cell ELISpot assay. The assay development considers the quality of sporozoite preparation as well as the detection threshold of the frequency of antigen-specific B cells. The assay enables the detection of sporozoite-specific IgM and IgG-producing B cells. Moreover, the assay can detect sporozoite-reactive B cells from subjects that were either vaccinated with the radiation attenuated sporozoite vaccine or a recombinant pre-erythrocytic vaccine. CONCLUSION: The newly developed sporozoite-based B cell ELISpot enables the monitoring of changes in the frequency of sporozoite-specific B cells. Applying this assay to assess the potency of vaccination regimens or seasonal changes in B cell populations from subjects residing in malaria-endemic areas will provide an opportunity to gain insight into immune mechanisms involved in protection and/or disease.


Subject(s)
B-Lymphocytes/immunology , Enzyme-Linked Immunospot Assay , Malaria Vaccines/immunology , Sporozoites/immunology , Sporozoites/radiation effects , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Clinical Trials as Topic , Humans , Leukocytes, Mononuclear/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Sensitivity and Specificity , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
8.
Parasite Immunol ; 41(7): e12622, 2019 07.
Article in English | MEDLINE | ID: mdl-30854655

ABSTRACT

Immunization with radiation-attenuated Plasmodium sporozoites (RAS) induces sterile and long-lasting protective immunity. Although intravenous (IV) route of RAS immunization is reported to induce superior immunity compared to intradermal (ID) injection, its role in the maintenance of sterile immunity is yet to be understood. We investigated whether the route of homologous sporozoite challenge of Plasmodium berghei (Pb) RAS-immunized mice would influence the longevity of protection. C57BL/6 mice immunized with Pb-RAS by IV were 100% protected upon primary IV/ID sporozoite challenge. In contrast, ID immunization resulted in 80% protection, regardless of primary challenge route. Interestingly, the route of primary challenge was found to bring difference in the maintenance of sterile protection. While IV Pb RAS-immunized mice remained protected at all challenges regardless of the route of primary challenge, ID Pb-RAS-immunized mice receiving ID primary challenge became parasitaemic upon secondary IV challenge. Significantly, primary IV challenge of Pb RAS ID-immunized mice resulted in 80% and 50% survival at secondary and tertiary challenges, respectively. According to phenotypically diverse liver CD8+ T cells, the percentages and the numbers of both CD8+ T effector memory and resident memory cells were significantly higher in IV than in ID Pb RAS-immunized mice. IFN-γ-producing CD8+ T cells specific to Pb TRAP130 and MIP-4-Kb-17 were also found significantly higher in IV mice than in ID mice. The enhanced T-cell generation and the longevity of protection appear to be dependent on the parasite load during challenge when infection is tolerated under suboptimal CD8+ T-cell response.


Subject(s)
Immunologic Memory , Liver/immunology , Malaria/immunology , Plasmodium berghei/immunology , Sporozoites/immunology , Administration, Intravenous , Animals , Antigens, Protozoan/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Female , Immunization , Injections, Intradermal , Liver/parasitology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Parasite Load , Sporozoites/radiation effects
9.
J Immunol ; 201(7): 1984-1993, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30127085

ABSTRACT

Tissue-resident memory CD8+ T (Trm) cells in the liver are critical for long-term protection against pre-erythrocytic Plasmodium infection. Such protection can usually be induced with three to five doses of i.v. administered radiation-attenuated sporozoites (RAS). To simplify and accelerate vaccination, we tested a DNA vaccine designed to induce potent T cell responses against the SYVPSAEQI epitope of Plasmodium yoelii circumsporozoite protein. In a heterologous "prime-and-trap" regimen, priming using gene gun-administered DNA and boosting with one dose of RAS attracted expanding Ag-specific CD8+ T cell populations to the liver, where they became Trm cells. Vaccinated in this manner, BALB/c mice were completely protected against challenge, an outcome not reliably achieved following one dose of RAS or following DNA-only vaccination. This study demonstrates that the combination of CD8+ T cell priming by DNA and boosting with liver-homing RAS enhances formation of a completely protective liver Trm cell response and suggests novel approaches for enhancing T cell-based pre-erythrocytic malaria vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Liver/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium yoelii/physiology , Plasmodium/immunology , Plasmodium/physiology , Protozoan Proteins/immunology , Sporozoites/immunology , T-Lymphocytes, Regulatory/immunology , Vaccination/methods , Animals , Cell Proliferation , Humans , Immunologic Memory , Liver/parasitology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Radiation , Sporozoites/radiation effects , Vaccines, Attenuated , Vaccines, DNA
10.
Am J Trop Med Hyg ; 99(4): 827-832, 2018 10.
Article in English | MEDLINE | ID: mdl-30141395

ABSTRACT

In August 2017, the National Institute of Allergy and Infectious Diseases convened a meeting, entitled "Understanding the Liver-Stage Biology of Malaria Parasites to Enable and Accelerate the Development of a Highly Efficacious Vaccine," to discuss the needs and strategies to develop a highly efficacious, whole organism-based vaccine targeting the liver stage of malaria parasites. It was concluded that attenuated sporozoite platforms have proven to be promising approaches, and that late-arresting sporozoites could potentially offer greater vaccine performance than early-arresting sporozoites against malaria. New knowledge and emerging technologies have made the development of late-arresting sporozoites feasible. Highly integrated approaches involving liver-stage research, "omics" studies, and cutting-edge genetic editing technologies, combined with in vitro culture systems or unique animal models, are needed to accelerate the discovery of candidates for a late-arresting, genetically attenuated parasite vaccine.


Subject(s)
Liver/immunology , Malaria Vaccines/genetics , Malaria, Falciparum/prevention & control , Malaria, Vivax/prevention & control , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Sporozoites/immunology , Animals , Disease Models, Animal , Gamma Rays , Genetic Engineering/methods , Humans , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Mice , Plasmodium berghei/chemistry , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/radiation effects , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/radiation effects , Plasmodium vivax/chemistry , Plasmodium vivax/genetics , Plasmodium vivax/radiation effects , Plasmodium yoelii/chemistry , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Plasmodium yoelii/radiation effects , Sporozoites/chemistry , Sporozoites/genetics , Sporozoites/radiation effects , Vaccines, Attenuated
11.
Front Immunol ; 9: 1137, 2018.
Article in English | MEDLINE | ID: mdl-29892289

ABSTRACT

Whole sporozoite vaccines represent one of the most promising strategies to induce protection against malaria. However, the development of efficient vaccination protocols still remains a major challenge. To understand how the generation of immunity is affected by variations in vaccination dosage and frequency, we systematically analyzed intrasplenic and intrahepatic CD8+ T cell responses following varied immunizations of mice with radiation-attenuated sporozoites. By combining experimental data and mathematical modeling, our analysis indicates a reversing role of spleen and liver in the generation of protective liver-resident CD8+ T cells during priming and booster injections: While the spleen acts as a critical source compartment during priming, the increase in vaccine-induced hepatic T cell levels is likely due to local reactivation in the liver in response to subsequent booster injections. Higher dosing accelerates the efficient generation of liver-resident CD8+ T cells by especially affecting their local reactivation. In addition, we determine the differentiation and migration pathway from splenic precursors toward hepatic memory cells thereby presenting a mechanistic framework for the impact of various vaccination protocols on these dynamics. Thus, our work provides important insights into organ-specific CD8+ T cell dynamics and their role and interplay in the formation of protective immunity against malaria.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Plasmodium/radiation effects , Sporozoites/immunology , Sporozoites/radiation effects , Algorithms , Animals , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Host-Pathogen Interactions/immunology , Immunization , Immunologic Memory , Immunophenotyping , Liver/immunology , Liver/parasitology , Lymphocyte Count , Malaria/prevention & control , Mice , Models, Biological , Models, Theoretical , Organ Specificity/immunology , Plasmodium berghei/immunology , Spleen/immunology , Spleen/parasitology , Vaccination
12.
Front Immunol ; 9: 192, 2018.
Article in English | MEDLINE | ID: mdl-29472929

ABSTRACT

Immunization with radiation-attenuated sporozoites (RAS) shown to confer complete sterile protection against Plasmodia liver-stage (LS) infection that lasts about 6 to 9 months in mice. We have found that the intermittent infectious sporozoite challenge to immune mice following RAS vaccination extends the longevity of sterile protection by maintaining CD8+ T cell memory responses to LS infection. It is reported that CD8α+ dendritic cells (DCs) are involved in the induction of LS-specific CD8+ T cells following RAS or genetically attenuated parasite (GAP) vaccination. In this study, we demonstrate that CD8α+ DCs respond differently to infectious sporozoite or RAS inoculation. The higher accumulation and activation of CD8α+ DCs was seen in the liver in response to infectious sporozoite 72 h postinoculation and found to be associated with higher expression of chemokines (CCL-20 and CCL-21) and type I interferon response via toll-like receptor signaling in liver. Moreover, the infectious sporozoites were found to induce qualitative changes in terms of the increased MHCII expression as well as costimulatory molecules including CD40 on the CD8α+ DCs compared to RAS inoculation. We have also found that infectious sporozoite challenge increased CD40L-expressing CD4+ T cells, which could help CD8+ T cells in the liver through "licensing" of the antigen-presenting cells. Our results suggest that infectious sporozoite challenge to prior RAS immunized mice modulates the CD8α+ DCs, which might be shaping the fate of memory CD8+ T cells against Plasmodium LS infection.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , Immunologic Memory , Liver/immunology , Malaria/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/genetics , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Chemokines/immunology , Dendritic Cells/parasitology , Female , Interferon Type I/immunology , Liver/cytology , Liver/parasitology , Mice , Mice, Inbred BALB C , Plasmodium berghei , Sporozoites/immunology , Sporozoites/radiation effects
13.
PLoS Negl Trop Dis ; 10(10): e0005070, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27760143

ABSTRACT

BACKGROUND: Immunizing human volunteers by mosquito bite with radiation-attenuated Plasmodium falciparum sporozoites (RAS) results in high-level protection against infection. Only two volunteers have been similarly immunized with P. vivax (Pv) RAS, and both were protected. A phase 2 controlled clinical trial was conducted to assess the safety and protective efficacy of PvRAS immunization. METHODOLOGY/PRINCIPAL FINDINGS: A randomized, single-blinded trial was conducted. Duffy positive (Fy+; Pv susceptible) individuals were enrolled: 14 received bites from irradiated (150 ± 10 cGy) Pv-infected Anopheles mosquitoes (RAS) and 7 from non-irradiated non-infected mosquitoes (Ctl). An additional group of seven Fy- (Pv refractory) volunteers was immunized with bites from non-irradiated Pv-infected mosquitoes. A total of seven immunizations were carried out at mean intervals of nine weeks. Eight weeks after last immunization, a controlled human malaria infection (CHMI) with non-irradiated Pv-infected mosquitoes was performed. Nineteen volunteers completed seven immunizations (12 RAS, 2 Ctl, and 5 Fy-) and received a CHMI. Five of 12 (42%) RAS volunteers were protected (receiving a median of 434 infective bites) compared with 0/2 Ctl. None of the Fy- volunteers developed infection by the seventh immunization or after CHMI. All non-protected volunteers developed symptoms 8-13 days after CHMI with a mean pre-patent period of 12.8 days. No serious adverse events related to the immunizations were observed. Specific IgG1 anti-PvCS response was associated with protection. CONCLUSION: Immunization with PvRAS was safe, immunogenic, and induced sterile immunity in 42% of the Fy+ volunteers. Moreover, Fy- volunteers were refractory to Pv malaria. TRIAL REGISTRATION: Identifier: NCT01082341.


Subject(s)
Anopheles/parasitology , Immunization/methods , Insect Bites and Stings , Malaria Vaccines/immunology , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Plasmodium vivax/immunology , Adolescent , Adult , Animals , Antibodies, Protozoan/blood , Colombia , Duffy Blood-Group System , Female , Humans , Immunization/adverse effects , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Malaria, Vivax/ethnology , Malaria, Vivax/parasitology , Male , Middle Aged , Plasmodium vivax/physiology , Plasmodium vivax/radiation effects , Single-Blind Method , Sporozoites/radiation effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Volunteers , Young Adult
14.
Malar J ; 15(1): 377, 2016 Jul 22.
Article in English | MEDLINE | ID: mdl-27448805

ABSTRACT

BACKGROUND: In this phase 1 clinical trial, healthy adult, malaria-naïve subjects were immunized with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) by mosquito bite and then underwent controlled human malaria infection (CHMI). The PfRAS model for immunization against malaria had previously induced >90 % sterile protection against homologous CHMI. This study was to further explore the safety, tolerability and protective efficacy of the PfRAS model and to provide biological specimens to characterize protective immune responses and identify protective antigens in support of malaria vaccine development. METHODS: Fifty-seven subjects were screened, 41 enrolled and 30 received at least one immunization. The true-immunized subjects received PfRAS via mosquito bite and the mock-immunized subjects received mosquito bites from irradiated uninfected mosquitoes. Sera and peripheral blood mononuclear cells (PBMCs) were collected before and after PfRAS immunizations. RESULTS: Immunization with PfRAS was generally safe and well tolerated, and repeated immunization via mosquito bite did not appear to increase the risk or severity of AEs. Local adverse events (AEs) of true-immunized and mock-immunized groups consisted of erythaema, papules, swelling, and induration and were consistent with reactions from mosquito bites seen in nature. Two subjects, one true- and one mock-immunized, developed large local reactions that completely resolved, were likely a result of mosquito salivary antigens, and were withdrawn from further participation as a safety precaution. Systemic AEs were generally rare and mild, consisting of headache, myalgia, nausea, and low-grade fevers. Two true-immunized subjects experienced fever, malaise, myalgia, nausea, and rigours approximately 16 h after immunization. These symptoms likely resulted from pre-formed antibodies interacting with mosquito salivary antigens. Ten subjects immunized with PfRAS underwent CHMI and five subjects (50 %) were sterilely protected and there was a significant delay to parasitaemia in the other five subjects. All ten subjects developed humoral immune responses to whole sporozoites and to the circumsporozoite protein prior to CHMI, although the differences between protected and non-protected subjects were not statistically significant for this small sample size. CONCLUSIONS: The protective efficacy of this clinical trial (50 %) was notably less than previously reported (>90 %). This may be related to differences in host genetics or the inherent variability in mosquito biting behavior and numbers of sporozoites injected. Differences in trial procedures, such as the use of leukapheresis prior to CHMI and of a longer interval between the final immunization and CHMI in these subjects compared to earlier trials, may also have reduced protective efficacy. This trial has been retrospectively registered at ISRCTN ID 17372582, May 31, 2016.


Subject(s)
Antibodies, Protozoan/blood , Culicidae/physiology , Insect Bites and Stings , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Adolescent , Adult , Animals , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Malaria Vaccines/administration & dosage , Male , Middle Aged , Plasmodium falciparum/radiation effects , Sporozoites/immunology , Sporozoites/radiation effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Young Adult
15.
Parasitol Res ; 115(1): 329-37, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26435485

ABSTRACT

Cryptosporidium is an important waterborne pathogen for which no treatment or vaccination is available. This study set out to quantify DNA replication of Cryptosporidium parvum in vitro. Cryptosporidium DNA could be detected at up to 60 % of input level in both host-cell-free and host cell containing cultures 6 days after infection with living sporozoites, but was lost within 2 days in cultures inoculated with UV-inactivated sporozoites. Total DNA increased between days 2 and 6, evidence of successful DNA replication in both cell-free and host-cell-containing cultures. Overall however, only a small fraction (up to 5 %) of parasite DNA could be found associated with host cells or bound to plastic of the cell-free cultures, and the majority of parasite DNA was present in the cell culture medium, separable by simple decantation. After 2 days, in host-cell-containing cultures, the parasite DNA could be concentrated by slow centrifugation, suggesting that it was associated with intact parasite cells, but at 6 days, the majority could not be centrifuged and is therefore thought to have represented copies associated with dead and degraded parasites. In cell-free cultures and in larger plates, the majority of DNA was in this form. Performance of the parasite was best in small culture plates, and least in the largest plate sizes. We interpret these results as suggesting that Cryptosporidium sporozoites first bind to the host cell monolayer or to the plasticware, but then by 2 days, there has been a substantial release of parasites back into the medium. Host-cell-free cultures also supported modest replication and may have represented DNA synthesis in cells beginning merogony. The role of the host cells is unclear, as so much of the parasite DNA is released into the medium. Host cells may provide a feeder role, conditioning the medium for Cryptosporidium development.


Subject(s)
Cryptosporidium parvum/growth & development , DNA Replication , DNA, Protozoan/analysis , Cryptosporidiosis/parasitology , Cryptosporidium parvum/genetics , Cryptosporidium parvum/radiation effects , Culture Media , DNA, Protozoan/isolation & purification , Humans , Real-Time Polymerase Chain Reaction , Sporozoites/growth & development , Sporozoites/radiation effects , Tumor Cells, Cultured , Ultraviolet Rays
16.
Eur J Immunol ; 46(4): 885-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26703789

ABSTRACT

MHC class I dependent CD8(+) T cells are essential for protection induced by radiation-attenuated Plasmodium sporozoites (RAS) in murine malaria models. Apart from the mechanism of activation of CD8(+) T cells specific for the circumsporozoite protein, the major sporozoite antigen (Ag), CD8(+) T cells specific for other exoerythrocytic Ags that have been shown to mediate protection have not been thoroughly investigated. Specifically, mechanisms of processing and presentation of exoerythrocytic Ags, which includes liver stage (LS) Ags, remain poorly understood. We hypothesize that as exogenous proteins, LS Ags are processed by mechanisms involving either the TAP-dependent phagosomal-to-cytosol or TAP-independent vacuolar pathway of cross-presentation. We used TAP-deficient mice to investigate whether LS Ag mediated induction of naïve CD8(+) T cells and their recall during sporozoite challenge occur by the TAP-dependent or TAP-independent pathways. On the basis of functional attributes, CD8(+) T cells were activated via the TAP-independent pathway during immunizations with Plasmodium berghei RAS; however, IFN-γ(+) CD8(+) T cells previously induced by P. berghei RAS in TAP-deficient mice failed to be recalled against sporozoite challenge and the mice became parasitemic. On the basis of these observations, we propose that TAP-associated Ag processing is indispensable for sterile protection induced with P. berghei RAS.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antigen Presentation/immunology , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Plasmodium berghei/immunology , Sporozoites/immunology , ATP-Binding Cassette Transporters/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Female , Histocompatibility Antigens Class I/immunology , Immunization , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Liver/cytology , Liver/immunology , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/radiation effects , Protozoan Proteins/immunology , Sporozoites/radiation effects
17.
Vaccine ; 33(52): 7462-8, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26469716

ABSTRACT

The parasitic disease malaria threatens more than 3 billion people worldwide, resulting in more than 200 million clinical cases and almost 600,000 deaths annually. Vaccines remain crucial for prevention and ultimately eradication of infectious diseases and, for malaria, whole sporozoite based immunization has been shown to be the most effective in experimental settings. In addition to immunization with radiation-attenuated sporozoites, chemoprophylaxis and sporozoites (CPS) is a highly efficient strategy to induce sterile protection in humans. Genetically attenuated parasites (GAP) have demonstrated significant protection in rodent studies, and are now being advanced into clinical testing. This review describes the existing pre-clinical and clinical data on CPS and GAP, discusses recent developments and examines how to transform these immunization approaches into vaccine candidates for clinical development.


Subject(s)
Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium/immunology , Sporozoites/immunology , Vaccination/methods , Animals , Antimalarials/therapeutic use , Chloroquine/therapeutic use , Drug Evaluation, Preclinical , Humans , Malaria/immunology , Malaria Vaccines/genetics , Mice , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Pre-Exposure Prophylaxis , Sporozoites/radiation effects , Vaccines, Attenuated/immunology
18.
Vaccine ; 33(25): 2851-7, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-25917675

ABSTRACT

Great progress has been made in the development of whole sporozoite vaccines including the manufacturing of cryopreserved Plasmodium falciparum sporozoites (PfSPZ) suitable for clinical application. Such whole sporozoites are being used for clinical studies of controlled human malaria infection (CHMI) as well as for evaluation of candidate vaccine approaches (both attenuated sporozoites and infectious sporozoites administered with chemoprophylaxis) and as reagents for immunology and cell biology assays. CHMI studies with whole sporozoites provide a great opportunity to better understand the intrinsic mechanisms of resistance to P. falciparum (e.g. due to sickle cell trait and other hemoglobinopathies) as well as host responses to an initial P. falciparum infection. High-level protective efficacy has been demonstrated in a small number of volunteers after intravenous (IV) inoculation of radiation-attenuated PfSPZ or in those who were exposed to live PfSPZ while on malaria chemoprophylaxis. These advances and data warrant further investigations of the immunological mechanism(s) whereby whole sporozoite inoculation elicits protective immunity in order to facilitate whole sporozoite vaccine development. The National Institute of Allergy and Infectious Diseases (NIAID) convened a workshop on Sept. 2-3, 2014 involving participation of international experts in the field of malaria vaccine development, and in basic and clinical immunology research. The workshop discussed the current understanding of host immune responses to whole malaria sporozoite inoculation, identified gaps in knowledge, resources to facilitate progress, and applicable new technologies and approaches to accelerate immunologic and vaccinologic studies and biomarker identification. This report summarizes the discussions and major conclusions from the workshop participants.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Sporozoites/immunology , Adaptive Immunity , Animals , Humans , Sporozoites/radiation effects , Vaccination
19.
J Control Release ; 204: 30-7, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25725360

ABSTRACT

Immunization with radiation-attenuated sporozoites (RAS) via mosquito bites has been shown to induce sterile immunity against malaria in humans, but this route of vaccination is neither practical nor ethical. The importance of delivering RAS to the liver through circulation in eliciting immunity against this parasite has been recently verified by human studies showing that high-level protection was achieved only by intravenous (IV) administration of RAS, not by intradermal (ID) or subcutaneous (SC) vaccination. Here, we report in a murine model that ID inoculation of RAS into laser-illuminated skin confers immune protection against malarial infection almost as effectively as IV immunization. Brief illumination of the inoculation site with a low power 532 nm Nd:YAG laser enhanced the permeability of the capillary beneath the skin, owing to hemoglobin-specific absorbance of the light. The increased blood vessel permeability appeared to facilitate an association of RAS with blood vessel walls by an as-yet-unknown mechanism, ultimately promoting a 7-fold increase in RAS entering circulation and reaching the liver over ID administration. Accordingly, ID immunization of RAS at a laser-treated site stimulated much stronger sporozoite-specific antibody and CD8(+)IFN-γ(+) T cell responses than ID vaccination and provided nearly full protection against malarial infection, whereas ID immunization alone was ineffective. This novel, safe, and convenient strategy to augment efficacy of ID sporozoite-based vaccines warrants further investigation in large animals and in humans.


Subject(s)
Insect Bites and Stings , Lasers , Malaria Vaccines/administration & dosage , Skin/immunology , Sporozoites/immunology , Vaccination , Animals , Capillary Permeability/radiation effects , Female , Injections, Intradermal , Malaria Vaccines/immunology , Mice, Inbred BALB C , Microscopy, Confocal , Plasmodium yoelii/immunology , Plasmodium yoelii/pathogenicity , Skin/blood supply , Skin/parasitology , Skin/radiation effects , Sporozoites/radiation effects , Vaccination/instrumentation , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
20.
J Immunol ; 193(3): 1246-57, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24981449

ABSTRACT

Malarial infection is initiated when the sporozoite form of the Plasmodium parasite is inoculated into the skin by a mosquito. Sporozoites invade hepatocytes in the liver and develop into the erythrocyte-infecting form of the parasite, the cause of clinical blood infection. Protection against parasite development in the liver can be induced by injection of live attenuated parasites that do not develop in the liver and thus do not cause blood infection. Radiation-attenuated sporozoites (RAS) and genetically attenuated parasites are now considered as lead candidates for vaccination of humans against malaria. Although the skin appears as the preferable administration route, most studies in rodents, which have served as model systems, have been performed after i.v. injection of attenuated sporozoites. In this study, we analyzed the early response to Plasmodium berghei RAS or wild-type sporozoites (WTS) injected intradermally into C57BL/6 mice. We show that RAS have a similar in vivo distribution to WTS and that both induce a similar inflammatory response consisting of a biphasic recruitment of polymorphonuclear neutrophils and inflammatory monocytes in the skin injection site and proximal draining lymph node (dLN). Both WTS and RAS associate with neutrophils and resident myeloid cells in the skin and the dLN, transform inside CD11b(+) cells, and induce a Th1 cytokine profile in the dLN. WTS and RAS are also similarly capable of priming parasite-specific CD8(+) T cells. These studies delineate the early and local response to sporozoite injection into the skin, and suggest that WTS and RAS prime the host immune system in a similar fashion.


Subject(s)
Malaria/immunology , Plasmodium berghei/immunology , Skin/immunology , Skin/parasitology , Sporozoites/immunology , Sporozoites/transplantation , Animals , Anopheles/immunology , Anopheles/parasitology , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Cell Line , Ear , Female , Hep G2 Cells , Host-Parasite Interactions/immunology , Humans , Inflammation/immunology , Inflammation/parasitology , Inflammation/pathology , Injections, Intradermal , Lymph Nodes/immunology , Lymph Nodes/parasitology , Malaria/parasitology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmodium berghei/parasitology , Skin/cytology , Sporozoites/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...