Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Lancet Infect Dis ; 23(11): 1266-1279, 2023 11.
Article in English | MEDLINE | ID: mdl-37499679

ABSTRACT

BACKGROUND: Malaria transmission-blocking vaccines target mosquito-stage parasites and will support elimination programmes. Gamete vaccine Pfs230D1-EPA/Alhydrogel induced superior activity to zygote vaccine Pfs25-EPA/Alhydrogel in malaria-naive US adults. Here, we compared these vaccines in malaria-experienced Malians. METHODS: We did a pilot safety study then double-blind, block-randomised, comparator-controlled main-phase trial in malaria-intense Bancoumana, Mali. 18-50-year-old healthy non-pregnant, non-breastfeeding consenting adult residents were randomly assigned (1:1:1:1) to receive four doses at months 0, 1, 4·5, and 16·5 of either 47 µg Pfs25, 40 µg Pfs230D1 or comparator (Twinrix or Menactra)-all co-administered with normal saline for blinding-or 47 µg Pfs25 plus 40 µg Pfs230D1 co-administered. We documented safety and tolerability (primary endpoint in the as-treated populations) and immunogenicity (secondary endpoint in the as-treated populations: ELISA, standard-membrane-feeding assay, and mosquito direct skin feed assay). This trial is registered at ClinicalTrials.gov, NCT02334462. FINDINGS: Between March 19, and June 2, 2015, we screened 471 individuals. Of 225 enrolled for the pilot and main cohorts, we randomly assigned 25 participants to pilot safety cohort groups of five (20%) to receive a two-dose series of Pfs25-EPA/Alhydrogel (16 µg), Pfs230D1-EPA/Alhydrogel (15 µg) or comparator, followed by Pfs25-EPA/Alhydrogel (16 µg) plus Pfs230D1-EPA/Alhydrogel (15 µg) or comparator plus saline. For the main cohort, we enrolled 200 participants between May 11 and June 2, 2015, to receive a four-dose series of 47 µg Pfs25-EPA/Alhydrogel plus saline (n=50 [25%]; Pfs25), 40 µg Pfs230D1-EPA/Alhydrogel plus saline (n=49 [25%]; Pfs230D1), 47 µg Pfs25-EPA/Alhydrogel plus 40 µg Pfs230D1-EPA/Alhydrogel (n=50 [25%]; Pfs25 plus Pfs230D1), or comparator (Twinrix or Menactra) plus saline (n=51 [25%]). Vaccinations were well tolerated in the pilot safety and main phases. Most vaccinees became seropositive after two Pfs230D1 or three Pfs25 doses; peak titres increased with each dose thereafter (Pfs230D1 geometric mean: 77·8 [95% CI 56·9-106·3], 146·4 [108·3-198·0], and 410·2 [301·6-558·0]; Pfs25 geometric mean 177·7 [130·3-242·4] and 315·7 [209·9-474·6]). Functional activity (mean peak transmission-reducing activity) appeared for Pfs230D1 (74·5% [66·6-82·5]) and Pfs25 plus Pfs230D1 (68·6% [57·3-79·8]), after the third dose and after the fourth dose (88·9% [81·7-96·2] for Pfs230D1 and 85·0% [78·4-91·5] Pfs25 plus Pfs230D1) but not for Pfs25 (58·2% [49·1-67·3] after the third dose and 58·2% [48·5-67·9] after the fourth dose). Pfs230D1 transmission-reducing activity (73·7% [64·1-83·3]) persisted 10 weeks after the fourth dose. Transmission-reducing activity of 80% was estimated at 1659 ELISA units for Pfs25, 218 for Pfs230D1, and 223 for Pfs230D1 plus Pfs25. After 3850 direct skin feed assays, 35 participants (12 Pfs25, eight Pfs230D1, five Pfs25 plus Pfs230D1, and ten comparator) had transmitted parasites at least once. The proportion of positive assays in vaccine groups (Pfs25 33 [3%] of 982 [-0·013 to 0·014], Pfs230D1 22 [2%] of 954 [-0·005 to 0·027], and combination 11 [1%] of 940 [-0·024 to 0·002]) did not differ from that of the comparator (22 [2%] of 974), nor did Pfs230D1 and combination groups differ (-0·024 to 0·001). INTERPRETATION: Pfs230D1 but not Pfs25 vaccine induces durable serum functional activity in Malian adults. Direct skin feed assays detect parasite transmission to mosquitoes but increased event rates are needed to assess vaccine effectiveness. FUNDING: Intramural Research Program of the National Institute of Allergy and Infectious Diseases and US National Institutes of Health.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Meningococcal Vaccines , Animals , Adult , Humans , Adolescent , Young Adult , Middle Aged , Aluminum Hydroxide , Plasmodium falciparum , Malaria Vaccines/adverse effects , Double-Blind Method , Immunogenicity, Vaccine
2.
NPJ Vaccines ; 8(1): 20, 2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36808125

ABSTRACT

A malaria vaccine that blocks parasite transmission from human to mosquito would be a powerful method of disrupting the parasite lifecycle and reducing the incidence of disease in humans. Pfs48/45 is a promising antigen in development as a transmission blocking vaccine (TBV) against the deadliest malaria parasite Plasmodium falciparum. The third domain of Pfs48/45 (D3) is an established TBV candidate, but production challenges have hampered development. For example, to date, a non-native N-glycan is required to stabilize the domain when produced in eukaryotic systems. Here, we implement a SPEEDesign computational design and in vitro screening pipeline that retains the potent transmission blocking epitope in Pfs48/45 while creating a stabilized non-glycosylated Pfs48/45 D3 antigen with improved characteristics for vaccine manufacture. This antigen can be genetically fused to a self-assembling single-component nanoparticle, resulting in a vaccine that elicits potent transmission-reducing activity in rodents at low doses. The enhanced Pfs48/45 antigen enables many new and powerful approaches to TBV development, and this antigen design method can be broadly applied towards the design of other vaccine antigens and therapeutics without interfering glycans.

3.
Nature ; 595(7866): 289-294, 2021 07.
Article in English | MEDLINE | ID: mdl-34194041

ABSTRACT

The global decline in malaria has stalled1, emphasizing the need for vaccines that induce durable sterilizing immunity. Here we optimized regimens for chemoprophylaxis vaccination (CVac), for which aseptic, purified, cryopreserved, infectious Plasmodium falciparum sporozoites (PfSPZ) were inoculated under prophylactic cover with pyrimethamine (PYR) (Sanaria PfSPZ-CVac(PYR)) or chloroquine (CQ) (PfSPZ-CVac(CQ))-which kill liver-stage and blood-stage parasites, respectively-and we assessed vaccine efficacy against homologous (that is, the same strain as the vaccine) and heterologous (a different strain) controlled human malaria infection (CHMI) three months after immunization ( https://clinicaltrials.gov/ , NCT02511054 and NCT03083847). We report that a fourfold increase in the dose of PfSPZ-CVac(PYR) from 5.12 × 104 to 2 × 105 PfSPZs transformed a minimal vaccine efficacy (low dose, two out of nine (22.2%) participants protected against homologous CHMI), to a high-level vaccine efficacy with seven out of eight (87.5%) individuals protected against homologous and seven out of nine (77.8%) protected against heterologous CHMI. Increased protection was associated with Vδ2 γδ T cell and antibody responses. At the higher dose, PfSPZ-CVac(CQ) protected six out of six (100%) participants against heterologous CHMI three months after immunization. All homologous (four out of four) and heterologous (eight out of eight) infectivity control participants showed parasitaemia. PfSPZ-CVac(CQ) and PfSPZ-CVac(PYR) induced a durable, sterile vaccine efficacy against a heterologous South American strain of P. falciparum, which has a genome and predicted CD8 T cell immunome that differs more strongly from the African vaccine strain than other analysed African P. falciparum strains.


Subject(s)
Antibodies, Neutralizing/immunology , Liver/immunology , Liver/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/drug effects , Plasmodium falciparum/immunology , Vaccines, Attenuated/immunology , Adult , Animals , Antibody Formation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Life Cycle Stages/immunology , Malaria/blood , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria Vaccines/chemistry , Male , Middle Aged , Plasmodium falciparum/growth & development , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Vaccination/adverse effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/chemistry
4.
Malar J ; 19(1): 323, 2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32883286

ABSTRACT

BACKGROUND: The recent expansion of tools designed to accurately quantify malaria parasite-produced antigens has enabled us to evaluate the performance of rapid diagnostic tests (RDTs) as a function of the antigens they detect-typically histidine rich protein 2 (HRP2) or lactate dehydrogenase (LDH). METHODS: For this analysis, whole blood specimens from a longitudinal study in Bancoumana, Mali were used to evaluate the performance of the ultra-sensitive HRP2-based Alere™ Malaria Ag P.f RDT (uRDT). The samples were collected as part of a transmission-blocking vaccine trial in a high transmission region for Plasmodium falciparum malaria. Furthermore, antigen dynamics after successful anti-malarial drug treatment were evaluated in these samples using the Q-Plex Human Malaria Array (4-Plex) to quantify antigen concentrations. RESULTS: The uRDT had a 50% probability of a positive result at 207 pg/mL HRP2 [95% credible interval (CrI) 160-268]. Individuals with symptomatic infection remained positive by uRDT for a median of 33 days [95% confidence interval (CI) 28-47] post anti-malarial drug treatment. Biphasic exponential decay models accurately captured the population level post-treatment dynamics of both HRP2 and Plasmodium LDH (pLDH), with the latter decaying more rapidly. Motivated by these differences in rates of decay, a novel algorithm that used HRP2:pLDH ratios to predict if an individual had active versus recently cleared P. falciparum infection was developed. The algorithm had 77.5% accuracy in correctly classifying antigen-positive individuals as those with and without active infection. CONCLUSIONS: These results characterize the performance of the ultra-sensitive RDT and demonstrate the potential for emerging antigen-quantifying technologies in the field of malaria diagnostics to be helpful tools in distinguishing between active versus recently cleared malaria infections.


Subject(s)
Antigens, Protozoan/isolation & purification , Diagnostic Tests, Routine/statistics & numerical data , L-Lactate Dehydrogenase/isolation & purification , Malaria, Falciparum/diagnosis , Plasmodium falciparum/isolation & purification , Protozoan Proteins/isolation & purification , Adult , Humans , Mali , Middle Aged , Sensitivity and Specificity , Young Adult
5.
Clin Infect Dis ; 71(6): 1481-1490, 2020 09 12.
Article in English | MEDLINE | ID: mdl-31621832

ABSTRACT

BACKGROUND: Chemoprophylaxis vaccination with sporozoites (CVac) with chloroquine induces protection against a homologous Plasmodium falciparum sporozoite (PfSPZ) challenge, but whether blood-stage parasite exposure is required for protection remains unclear. Chloroquine suppresses and clears blood-stage parasitemia, while other antimalarial drugs, such as primaquine, act against liver-stage parasites. Here, we evaluated CVac regimens using primaquine and/or chloroquine as the partner drug to discern whether blood-stage parasite exposure impacts protection against homologous controlled human malaria infection. METHODS: In a Phase I, randomized, partial double-blind, placebo-controlled study of 36 malaria-naive adults, all CVac subjects received chloroquine prophylaxis and bites from 12-15 P. falciparum-infected mosquitoes (CVac-chloroquine arm) at 3 monthly iterations, and some received postexposure primaquine (CVac-primaquine/chloroquine arm). Drug control subjects received primaquine, chloroquine, and uninfected mosquito bites. After a chloroquine washout, subjects, including treatment-naive infectivity controls, underwent homologous, PfSPZ controlled human malaria infection and were monitored for parasitemia for 21 days. RESULTS: No serious adverse events occurred. During CVac, all but 1 subject in the study remained blood-smear negative, while only 1 subject (primaquine/chloroquine arm) remained polymerase chain reaction-negative. Upon challenge, compared to infectivity controls, 3/3 chloroquine arm subjects displayed delayed patent parasitemia (P = .01) but not sterile protection, while 3/11 primaquine/chloroquine subjects remained blood-smear negative. CONCLUSIONS: CVac-primaquine/chloroquine is safe and induces sterile immunity to P. falciparum in some recipients, but a single 45 mg dose of primaquine postexposure does not completely prevent blood-stage parasitemia. Unlike previous studies, CVac-chloroquine did not produce sterile immunity. CLINICAL TRIALS REGISTRATION: NCT01500980.


Subject(s)
Antimalarials , Malaria, Falciparum , Adult , Animals , Antimalarials/therapeutic use , Chemoprevention , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Plasmodium falciparum , Sporozoites , Vaccination
6.
Am J Trop Med Hyg ; 100(6): 1466-1476, 2019 06.
Article in English | MEDLINE | ID: mdl-31017084

ABSTRACT

18S rRNA is a biomarker that provides an alternative to thick blood smears in controlled human malaria infection (CHMI) trials. We reviewed data from CHMI trials at non-endemic sites that used blood smears and Plasmodium 18S rRNA/rDNA biomarker nucleic acid tests (NATs) for time to positivity. We validated a multiplex quantitative reverse transcription-polymerase chain reaction (qRT-PCR) for Plasmodium 18S rRNA, prospectively compared blood smears and qRT-PCR for three trials, and modeled treatment effects at different biomarker-defined parasite densities to assess the impact on infection detection, symptom reduction, and measured intervention efficacy. Literature review demonstrated accelerated NAT-based infection detection compared with blood smears (mean acceleration: 3.2-3.6 days). For prospectively tested trials, the validated Plasmodium 18S rRNA qRT-PCR positivity was earlier (7.6 days; 95% CI: 7.1-8.1 days) than blood smears (11.0 days; 95% CI: 10.3-11.8 days) and significantly preceded the onset of grade 2 malaria-related symptoms (12.2 days; 95% CI: 10.6-13.3 days). Discrepant analysis showed that the risk of a blood smear-positive, biomarker-negative result was negligible. Data modeling predicted that treatment triggered by specific biomarker-defined thresholds can differentiate complete, partial, and non-protective outcomes and eliminate many grade 2 and most grade 3 malaria-related symptoms post-CHMI. Plasmodium 18S rRNA is a sensitive and specific biomarker that can justifiably replace blood smears for infection detection in CHMI trials in non-endemic settings. This study led to biomarker qualification through the U.S. Food and Drug Administration for use in CHMI studies at non-endemic sites, which will facilitate biomarker use for the qualified context of use in drug and vaccine trials.


Subject(s)
Malaria/diagnosis , Plasmodium/genetics , RNA, Protozoan/genetics , RNA, Ribosomal, 18S/blood , Biomarkers/blood , Humans , Multiplex Polymerase Chain Reaction , Plasmodium/isolation & purification , RNA, Ribosomal, 18S/genetics , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Trop Med Hyg ; 97(3): 719-725, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28722588

ABSTRACT

Malaria transmission-blocking vaccines (TBV) have been evaluated in field trials in Mali since 2013. However, the assays currently used to measure serum antibody TB activity (TBA) after vaccination are highly variable, in part due to the lack of optimization and standardization for field assays in which mosquitoes feed on gametocytemic blood. Herein, we report a study conducted in Bancoumana village, Mali, where we identify and optimize the parameters that contribute to successful mosquito feeding outcomes in both direct skin feeds (DSFs) and direct membrane feeding assays (DMFA). These parameters include: 1) mosquito age, 2) duration of mosquito starvation prior to feeding, 3) membrane selection for DMFA, 4) anatomical location of DSF feeding (arm, calf, and ankle), and 5) time of day for DSF (dawn or dusk). We found that younger mosquitoes were significantly associated with higher feeding, survival, and infection rates. Longer starvation times were positively, but not significantly, associated with higher infection rates, but were negatively associated with feeding and survival. Membrane type and body location did not affect infection outcome significantly. Although dusk was found to be associated with higher infection rates, this may be confounded by the time from positive blood smear. Based on these findings, we make specific recommendations for optimal feeding parameters in the different assay types to maximize the chance of detecting parasite transmission in a standardized manner.


Subject(s)
Anopheles/physiology , Insect Vectors/physiology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Membranes, Artificial , Plasmodium falciparum/physiology , Adolescent , Adult , Aging , Animals , Anopheles/parasitology , Feeding Behavior , Humans , Insect Vectors/parasitology , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Mali/epidemiology , Parasitemia/transmission , Skin
8.
Vaccine ; 34(48): 5863-5870, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27789147

ABSTRACT

Shifting the malaria priorities from a paradigm of control and elimination to a goal of global eradication calls for renewed attention to the interruption of malaria transmission. Sustained progress toward eradication will require both improved understanding of infectious reservoirs and efficient development of novel transmission-blocking interventions, such as rapidly acting and highly efficacious therapeutics and vaccines. Here, we review the direct skin feeding assay (DSF), which has been proposed as a valuable tool for measuring the in natura transmission of malaria parasites from human hosts to mosquito vectors across heterogeneous populations. To capture the methodological breadth of this assay's use, we first systematically review and qualitatively synthesize previously published investigations using DSFs to study malaria transmission in humans. Then, using a recent Phase 1 trial in Mali of the Pfs25H-EPA/Alhydrogel® vaccine candidate (NCT01867463) designed to interrupt Plasmodium falciparum transmission as a case study, we describe the potential opportunities and current limitations of utilizing the endpoints measured by DSF in making early clinical decisions for individually randomized transmission-interrupting intervention candidates. Using simulations based on the data collected in the clinical trial, we demonstrate that the capacity of the DSF to serve as an evaluative tool is limited by the statistical power constraints of the "effective sample size" (i.e. the number of subjects that are capable of transmitting at the time of feeding). Altogether, our findings suggest DSFs have great potential utility for assessing the public health impacts of emerging antimalarial tools, but additional research is needed to address issues of scalability and to establish correlation with community-wide clinical endpoints as well as complementary in vitro measures, such as standard membrane feeding assays.


Subject(s)
Anopheles/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Mosquito Vectors/parasitology , Adult , Animals , Clinical Trials as Topic , Feeding Behavior , Female , Humans , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Male , Middle Aged , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Skin/immunology , Young Adult
9.
Mol Ther ; 23(5): 857-865, 2015 May.
Article in English | MEDLINE | ID: mdl-25648263

ABSTRACT

Eliminating malaria parasites during the asymptomatic but obligate liver stages (LSs) of infection would stop disease and subsequent transmission. Unfortunately, only a single licensed drug that targets all LSs, Primaquine, is available. Targeting host proteins might significantly expand the repertoire of prophylactic drugs against malaria. Here, we demonstrate that both Bcl-2 inhibitors and P53 agonists dramatically reduce LS burden in a mouse malaria model in vitro and in vivo by altering the activity of key hepatocyte factors on which the parasite relies. Bcl-2 inhibitors act primarily by inducing apoptosis in infected hepatocytes, whereas P53 agonists eliminate parasites in an apoptosis-independent fashion. In combination, Bcl-2 inhibitors and P53 agonists act synergistically to delay, and in some cases completely prevent, the onset of blood stage disease. Both families of drugs are highly effective at doses that do not cause substantial hepatocyte cell death in vitro or liver damage in vivo. P53 agonists and Bcl-2 inhibitors were also effective when administered to humanized mice infected with Plasmodium falciparum. Our data demonstrate that host-based prophylaxis could be developed into an effective intervention strategy that eliminates LS parasites before the onset of clinical disease and thus opens a new avenue to prevent malaria.


Subject(s)
Antimalarials/pharmacology , Life Cycle Stages/drug effects , Liver/parasitology , Malaria/parasitology , Plasmodium/drug effects , Plasmodium/physiology , Post-Exposure Prophylaxis , Animals , Antimalarials/administration & dosage , Cell Line , Disease Models, Animal , Female , Imidazoles/administration & dosage , Imidazoles/pharmacology , Indoles , Malaria/drug therapy , Malaria/metabolism , Malaria/prevention & control , Malaria, Falciparum/drug therapy , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Mice , Mice, Transgenic , Parasite Load , Piperazines/administration & dosage , Piperazines/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/administration & dosage , Pyrroles/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
10.
Eur J Med Chem ; 74: 562-73, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24531197

ABSTRACT

Malaria remains a major health concern for a large percentage of the world's population. While great strides have been made in reducing mortality due to malaria, new strategies and therapies are still needed. Therapies that are capable of blocking the transmission of Plasmodium parasites are particularly attractive, but only primaquine accomplishes this, and toxicity issues hamper its widespread use. In this study, we describe a series of pyrazolopyrimidine- and imidazopyrazine-based compounds that are potent inhibitors of PfCDPK4, which is a calcium-activated Plasmodium protein kinase that is essential for exflagellation of male gametocytes. Thus, PfCDPK4 is essential for the sexual development of Plasmodium parasites and their ability to infect mosquitoes. We demonstrate that two structural features in the ATP-binding site of PfCDPK4 can be exploited in order to obtain potent and selective inhibitors of this enzyme. Furthermore, we demonstrate that pyrazolopyrimidine-based inhibitors that are potent inhibitors of the in vitro activity of PfCDPK4 are also able to block Plasmodium falciparum exflagellation with no observable toxicity to human cells. This medicinal chemistry effort serves as a valuable starting point in the development of safe, transmission-blocking agents for the control of malaria.


Subject(s)
Antimalarials/pharmacology , Calcium/metabolism , Malaria, Falciparum/transmission , Plasmodium falciparum/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , Amino Acid Sequence , Animals , Molecular Sequence Data , Sequence Homology, Amino Acid
11.
Malar J ; 11: 421, 2012 Dec 17.
Article in English | MEDLINE | ID: mdl-23244590

ABSTRACT

BACKGROUND: Malaria remains a major human health problem, with no licensed vaccine currently available. Malaria infections initiate when infectious Plasmodium sporozoites are transmitted by Anopheline mosquitoes during their blood meal. Investigations of the malaria sporozoite are, therefore, of clear medical importance. However, sporozoites can only be produced in and isolated from mosquitoes, and their isolation results in large amounts of accompanying mosquito debris and contaminating microbes. METHODS: Here is described a discontinuous density gradient purification method for Plasmodium sporozoites that maintains parasite infectivity in vitro and in vivo and greatly reduces mosquito and microbial contaminants. RESULTS: This method provides clear advantages over previous approaches: it is rapid, requires no serum components, and can be scaled to purify >107 sporozoites with minimal operator involvement. Moreover, it can be effectively applied to both human (Plasmodium falciparum, Plasmodium vivax) and rodent (Plasmodium yoelii) infective species with excellent recovery rates. CONCLUSIONS: This novel method effectively purifies viable malaria sporozoites by greatly reducing contaminating mosquito debris and microbial burdens associated with parasite isolation. Large-scale preparations of purified sporozoites will allow for enhanced in vitro infections, proteomics, and biochemical characterizations. In conjunction with aseptic mosquito rearing techniques, this purification technique will also support production of live attenuated sporozoites for vaccination.


Subject(s)
Centrifugation, Density Gradient/methods , Parasitology/methods , Plasmodium/isolation & purification , Sporozoites/cytology , Animals , Anopheles/parasitology , Disease Models, Animal , Female , Humans , Liver/parasitology , Malaria/parasitology , Mice , Plasmodium/pathogenicity , Plasmodium falciparum/isolation & purification , Plasmodium vivax/isolation & purification , Plasmodium yoelii/isolation & purification , Virulence
12.
Mol Biochem Parasitol ; 186(2): 143-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23107927

ABSTRACT

Plasmodium falciparum is the pathogenic agent of the most lethal of human malarias. Transgenic P. falciparum parasites expressing luciferase have been created to study drug interventions of both asexual and sexual blood stages but luciferase-expressing mosquito stage and liver stage parasites have not been created which has prevented the easy quantification of mosquito stage development (e.g. for transmission blocking interventions) and liver stage development (for interventions that prevent infection). To overcome this obstacle, we have created a transgenic P. falciparum NF54 parasite that expresses a GFP-luciferase transgene throughout the life cycle. Luciferase expression is robust and measurable at all life cycle stages, including midgut oocyst, salivary gland sporozoites and liver stages, where in vivo development is easily measurable using humanized mouse infections in conjunction with an in vivo imaging system. This parasite reporter strain will accelerate testing of interventions against pre-erythrocytic life cycle stages.


Subject(s)
Gene Expression , Green Fluorescent Proteins/genetics , Life Cycle Stages , Luciferases/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Transgenes , Animals , Culicidae/parasitology , Disease Models, Animal , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Gene Order , Gene Targeting , Green Fluorescent Proteins/metabolism , Humans , Luciferases/metabolism , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Mice
13.
Arterioscler Thromb Vasc Biol ; 32(3): 786-98, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22116094

ABSTRACT

OBJECTIVE: The coagulation-inflammation cycle has been implicated as a critical component in malaria pathogenesis. Defibrotide (DF), a mixture of DNA aptamers, displays anticoagulant, anti-inflammatory, and endothelial cell (EC)-protective activities and has been successfully used to treat comatose children with veno-occlusive disease. DF was investigated here as a drug to treat cerebral malaria. METHODS AND RESULTS: DF blocks tissue factor expression by ECs incubated with parasitized red blood cells and attenuates prothrombinase activity, platelet aggregation, and complement activation. In contrast, it does not affect nitric oxide bioavailability. We also demonstrated that Plasmodium falciparum glycosylphosphatidylinositol (Pf-GPI) induces tissue factor expression in ECs and cytokine production by dendritic cells. Notably, dendritic cells, known to modulate coagulation and inflammation systemically, were identified as a novel target for DF. Accordingly, DF inhibits Toll-like receptor ligand-dependent dendritic cells activation by a mechanism that is blocked by adenosine receptor antagonist (8-p-sulfophenyltheophylline) but not reproduced by synthetic poly-A, -C, -T, and -G. These results imply that aptameric sequences and adenosine receptor mediate dendritic cells responses to the drug. DF also prevents rosetting formation, red blood cells invasion by P. falciparum and abolishes oocysts development in Anopheles gambiae. In a murine model of cerebral malaria, DF affected parasitemia, decreased IFN-γ levels, and ameliorated clinical score (day 5) with a trend for increased survival. CONCLUSION: Therapeutic use of DF in malaria is proposed.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anticoagulants/pharmacology , Antimalarials/pharmacology , Blood Coagulation/drug effects , Endothelial Cells/drug effects , Malaria, Cerebral/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Polydeoxyribonucleotides/pharmacology , Animals , Cells, Cultured , Complement Activation/drug effects , Cytokines/blood , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/parasitology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/parasitology , Female , Glycosylphosphatidylinositols/metabolism , Hemoglobins/metabolism , Humans , Inflammation Mediators/blood , Malaria, Cerebral/blood , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Plasmodium berghei/pathogenicity , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Platelet Aggregation/drug effects , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/metabolism , Severity of Illness Index , Thromboplastin/metabolism , Time Factors
14.
PLoS One ; 6(6): e20156, 2011.
Article in English | MEDLINE | ID: mdl-21694762

ABSTRACT

BACKGROUND: Vector susceptibility to Plasmodium infection is treated primarily as a vector trait, although it is a composite trait expressing the joint occurrence of the parasite and the vector with genetic contributions of both. A comprehensive approach to assess the specific contribution of genetic and environmental variation on "vector susceptibility" is lacking. Here we developed and implemented a simple scheme to assess the specific contributions of the vector, the parasite, and the environment to "vector susceptibility." To the best of our knowledge this is the first study that employs such an approach. METHODOLOGY/PRINCIPAL FINDINGS: We conducted selection experiments on the vector (while holding the parasite "constant") and on the parasite (while holding the vector "constant") to estimate the genetic contributions of the mosquito and the parasite to the susceptibility of Anopheles stephensi to Plasmodium gallinaceum. We separately estimated the realized heritability of (i) susceptibility to parasite infection by the mosquito vector and (ii) parasite compatibility (transmissibility) with the vector while controlling the other. The heritabilities of vector and the parasite were higher for the prevalence, i.e., fraction of infected mosquitoes, than the corresponding heritabilities of parasite load, i.e., the number of oocysts per mosquito. CONCLUSIONS: The vector's genetics (heritability) comprised 67% of "vector susceptibility" measured by the prevalence of mosquitoes infected with P. gallinaceum oocysts, whereas the specific contribution of parasite genetics (heritability) to this trait was only 5%. Our parasite source might possess minimal genetic diversity, which could explain its low heritability (and the high value of the vector). Notably, the environment contributed 28%. These estimates are relevant only to the particular system under study, but this experimental design could be useful for other parasite-host systems. The prospects and limitations of the genetic manipulation of vector populations to render the vector resistant to the parasite are better considered on the basis of this framework.


Subject(s)
Anopheles/parasitology , Disease Susceptibility/parasitology , Environment , Parasites/physiology , Plasmodium gallinaceum/physiology , Quantitative Trait, Heritable , Animals , Chickens/parasitology , Inheritance Patterns/genetics , Insect Vectors/parasitology , Malaria, Avian/epidemiology , Malaria, Avian/parasitology , Oocysts , Prevalence
15.
PLoS Negl Trop Dis ; 3(10): e532, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19841746

ABSTRACT

BACKGROUND: Triatomines are vectors of Trypanosoma cruzi, the etiological agent of Chagas disease in Latin America. The most effective vector, Triatoma infestans, has been controlled successfully in much of Latin America using insecticide spraying. Though rarely undertaken, surveillance programs are necessary in order to identify new infestations and estimate the intensity of triatomine bug infestations in domestic and peridomestic habitats. Since hosts exposed to triatomines develop immune responses to salivary antigens, these responses can be evaluated for their usefulness as epidemiological markers to detect infestations of T. infestans. METHODOLOGY/PRINCIPAL FINDINGS: T. infestans salivary proteins were separated by 2D-gel electrophoresis and tested for their immunogenicity by Western blotting using sera from chickens and guinea pigs experimentally exposed to T. infestans. From five highly immunogenic protein spots, eight salivary proteins were identified by nano liquid chromatography-electrospray ionization-tandem mass spectrometry (nanoLC-ESI-MS/MS) and comparison to the protein sequences of the National Center for Biotechnology Information (NCBI) database and expressed sequence tags of a unidirectionally cloned salivary gland cDNA library from T. infestans combined with the NCBI yeast protein sub-database. The 14.6 kDa salivary protein [gi|149689094] was produced as recombinant protein (rTiSP14.6) in a mammalian cell expression system and recognized by all animal sera. The specificity of rTiSP14.6 was confirmed by the lack of reactivity to anti-mosquito and anti-sand fly saliva antibodies. However, rTiSP14.6 was recognized by sera from chickens exposed to four other triatomine species, Triatoma brasiliensis, T. sordida, Rhodnius prolixus, and Panstrongylus megistus and by sera of chickens from an endemic area of T. infestans and Chagas disease in Bolivia. CONCLUSIONS/SIGNIFICANCE: The recombinant rTiSP14.6 is a suitable and promising epidemiological marker for detecting the presence of small numbers of different species of triatomines and could be developed for use as a new tool in surveillance programs, especially to corroborate vector elimination in Chagas disease vector control campaigns.


Subject(s)
Chagas Disease/immunology , Insect Proteins/immunology , Insect Vectors/immunology , Salivary Proteins and Peptides/immunology , Triatoma/immunology , Triatominae/immunology , Amino Acid Sequence , Animals , Antigens/chemistry , Antigens/genetics , Antigens/immunology , Chagas Disease/parasitology , Chickens , Enzyme-Linked Immunosorbent Assay , Guinea Pigs , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Vectors/chemistry , Insect Vectors/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/genetics , Sequence Alignment , Triatoma/chemistry , Triatoma/genetics , Triatominae/chemistry , Triatominae/genetics
16.
Int J Parasitol ; 39(9): 1021-9, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19248784

ABSTRACT

Hematophagous arthropods such as Triatoma infestans, the vector of Trypanosoma cruzi, elicit host-immune responses during feeding. Characterization of antibody responses to salivary antigens offers the potential to develop immunologically based monitoring techniques for exposure to re-emergent triatomine bug populations in peridomestic animals. IgG-antibody responses to the salivary antigens of T.infestans have been detected in chickens as soon as 2 days after the first exposure to five adult bugs. Chickens and guinea pigs regularly exposed to this number of triatomines showed a significantly lower anti-saliva antibody titre than animals exposed to 25 adults and fifth instars of four different T.infestans strains originating from Bolivia and from Northern Chile. Highly immunogenic salivary antigens of 14 and 21kDa were recognised by all chicken sera and of 79kDa by all guinea pig sera. Cross-reactivity studies using saliva or salivary gland extracts from different hematophagous species, e.g. different triatomines, bed bugs, mosquitoes, sand flies and ticks, as well as chicken sera exposed to triatomines and mosquitoes, demonstrated that the 14 and 21kDa salivary antigens were only found in triatomines. Sera from peridomestic chickens and guinea pigs in sites of known T.infestans challenge in Bolivia also recognised the 14 and 21kDa antigens. These represent promising epidemiological markers for the detection of small numbers of feeding bugs and hence may be a new tool for vector surveillance in Chagas disease control programs.


Subject(s)
Animals, Domestic , Antibody Formation/immunology , Chagas Disease/immunology , Salivary Proteins and Peptides/immunology , Triatoma/immunology , Animals , Biomarkers , Bolivia , Chagas Disease/transmission , Chagas Disease/veterinary , Chickens , Chile , Guinea Pigs , Humans , Molecular Sequence Data , Psychodidae , Salivary Proteins and Peptides/genetics , Triatoma/genetics , Triatoma/pathogenicity
17.
PLoS One ; 4(2): e4549, 2009.
Article in English | MEDLINE | ID: mdl-19234606

ABSTRACT

BACKGROUND: As pathogens that circumvent the host immune response are favoured by selection, so are host alleles that reduce parasite load. Such evolutionary processes leave their signature on the genes involved. Deciphering modes of selection operating on immune genes might reveal the nature of host-pathogen interactions and factors that govern susceptibility in host populations. Such understanding would have important public health implications. METHODOLOGY/FINDINGS: We analyzed polymorphisms in four mosquito immune genes (SP14D1, GNBP, defensin, and gambicin) to decipher selection effects, presumably mediated by pathogens. Using samples of Anopheles arabiensis, An. quadriannulatus and four An. gambiae populations, as well as published sequences from other Culicidae, we contrasted patterns of polymorphisms between different functional units of the same gene within and between populations. Our results revealed selection signatures operating on different time scales. At the most recent time scale, within-population diversity revealed purifying selection. Between populations and between species variation revealed reduced differentiation (GNBP and gambicin) at coding vs. noncoding- regions, consistent with balancing selection. McDonald-Kreitman tests between An. quadriannulatus and both sibling species revealed higher fixation rate of synonymous than nonsynonymous substitutions (GNBP) in accordance with frequency dependent balancing selection. At the longest time scale (>100 my), PAML analysis using distant Culicid taxa revealed positive selection at one codon in gambicin. Patterns of genetic variation were independent of exposure to human pathogens. SIGNIFICANCE AND CONCLUSIONS: Purifying selection is the most common form of selection operating on immune genes as it was detected on a contemporary time scale on all genes. Selection for "hypervariability" was not detected, but negative balancing selection, detected at a recent evolutionary time scale between sibling species may be rather common. Detection of positive selection at the deepest evolutionary time scale suggests that it occurs infrequently, possibly in association with speciation events. Our results provided no evidence to support the hypothesis that selection was mediated by pathogens that are transmitted to humans.


Subject(s)
Anopheles/genetics , Evolution, Molecular , Genes, Insect , Immunity/genetics , Animals , Humans , Polymorphism, Genetic , Selection, Genetic
18.
Malar J ; 7: 33, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18282270

ABSTRACT

BACKGROUND: It is estimated that over 70% of patients with suspected malaria in sub-Saharan Africa, diagnose and manage their illness at home without referral to a formal health clinic. Of those patients who do attend a formal health clinic, malaria overdiagnosis rates are estimated to range between 30-70%. METHODS: This paper details an observational cohort study documenting the number and cost of repeat consultations as a result of malaria overdiagnosis at two health care providers in a rural district of Mozambique. 535 adults and children with a clinical diagnosis of malaria were enrolled and followed over a 21 day period to assess treatment regimen, symptoms, number and cost of repeat visits to health providers in patients misdiagnosed with malaria compared to those with confirmed malaria (determined by positive bloodfilm reading). RESULTS: Diagnosis based solely on clinical symptoms overdiagnosed 23% of children (<16y) and 31% of adults with malaria. Symptoms persisted (p = 0.023) and new ones developed (p < 0.001) in more adults than children in the three weeks following initial presentation. Adults overdiagnosed with malaria had more repeat visits (67% v 46%, p = 0.01-0.06) compared to those with true malaria. There was no difference in costs between patients correctly or incorrectly diagnosed with malaria. Median costs over three weeks were $0.28 for those who had one visit and $0.76 for > or = 3 visits and were proportionally highest among the poorest (p < 0.001) CONCLUSION: Overdiagnosis of malaria results in a greater number of healthcare visits and associated cost for adult patients. Additionally, it is clear that the poorest individuals pay significantly more proportionally for their healthcare making it imperative that the treatment they receive is correct in order to prevent wastage of limited economic resources. Thus, investment in accurate malaria diagnosis and appropriate management at primary level is critical for improving health outcomes and reducing poverty.


Subject(s)
Cost of Illness , Malaria/diagnosis , Malaria/economics , Adolescent , Adult , Ambulatory Care/economics , Ambulatory Care/statistics & numerical data , Antimalarials/economics , Antimalarials/therapeutic use , Child , Child, Preschool , Cohort Studies , Family Characteristics , Female , Humans , Infant , Malaria/drug therapy , Male , Middle Aged , Mozambique , Rural Population
19.
Vector Borne Zoonotic Dis ; 7(4): 659-66, 2007.
Article in English | MEDLINE | ID: mdl-18021022

ABSTRACT

Tick-borne relapsing fevers (TBRF) are caused by infection with Borrelia spirochetes and transmitted to humans by ticks. All except East African TBRF, caused by Borrelia duttonii, are known zoonoses. This widespread, endemic and pathogenic infection has only been found in humans and the Ornithodoros sp. soft tick vectors. We investigated the role of domestic animals as possible reservoirs of infection in a TBRF endemic region. Tick infestations in households and pigpens were investigated in the villages near Mvumi hospital in central Tanzania. Blood from chickens and pigs was examined by PCR and flagellin gene sequencing was performed on any Borrelia sp. infections detected. A mark-recapture experiment investigated tick movement between pigpens and houses. The acceptability of chickens as tick hosts was also investigated. Tick infestation of the 122 houses investigated was high (47%). Pigpens also were tick infested (16%) and were more likely to be so if they were located close to tick infested households (p<0.001). PCR screening of peripheral blood found Borrelia infections in both chickens and pigs (11% and 8.9% respectively). Sequencing of a subset of positive samples revealed that the amplified Borrelia sp. flagellin gene fragments shared greatest homology with B. duttonii. In a mark-recapture experiment, ticks released in pigpens were recaptured inside human bedrooms. When offered chickens as hosts, over 20% of ticks fed. For the first time in East Africa, we record natural infections of Borrelia in domestic animals and show that tick populations may act as bridging vectors between animals and humans. These results, from villages where B. duttonii is already known to be prevalent and a major cause of illness in humans, and where it has been found at high levels in ticks, strongly support the case that it is a zoonosis. This increases understanding of the epidemiology and control of this important but neglected human disease.


Subject(s)
Arthropod Vectors/microbiology , Borrelia/physiology , Disease Reservoirs/microbiology , Ornithodoros/microbiology , Relapsing Fever/microbiology , Animals , Arthropod Vectors/physiology , Borrelia/classification , Borrelia/genetics , Borrelia/isolation & purification , Chickens/microbiology , Flagellin/genetics , Housing , Humans , Ornithodoros/physiology , Phylogeny , Population Dynamics , Relapsing Fever/epidemiology , Relapsing Fever/transmission , Swine/microbiology , Tanzania/epidemiology , Tick Infestations
SELECTION OF CITATIONS
SEARCH DETAIL
...