Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 1629, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712609

ABSTRACT

The structural integrity of the host red blood cell (RBC) is crucial for propagation of Plasmodium spp. during the disease-causing blood stage of malaria infection. To assess the stability of Plasmodium vivax-infected reticulocytes, we developed a flow cytometry-based assay to measure osmotic stability within characteristically heterogeneous reticulocyte and P. vivax-infected samples. We find that erythroid osmotic stability decreases during erythropoiesis and reticulocyte maturation. Of enucleated RBCs, young reticulocytes which are preferentially infected by P. vivax, are the most osmotically stable. P. vivax infection however decreases reticulocyte stability to levels close to those of RBC disorders that cause hemolytic anemia, and to a significantly greater degree than P. falciparum destabilizes normocytes. Finally, we find that P. vivax new permeability pathways contribute to the decreased osmotic stability of infected-reticulocytes. These results reveal a vulnerability of P. vivax-infected reticulocytes that could be manipulated to allow in vitro culture and develop novel therapeutics.


Subject(s)
Malaria, Vivax , Plasmodium vivax , Reticulocytes/metabolism , Reticulocytes/parasitology , Anemia, Hemolytic , Bone Marrow , Cell Differentiation , Erythrocytes , Hemolysis , Humans , Malaria
2.
J Infect Dis ; 223(10): 1817-1821, 2021 05 28.
Article in English | MEDLINE | ID: mdl-32941614

ABSTRACT

Plasmodium vivax has 2 invasion ligand/host receptor pathways (P. vivax Duffy-binding protein/Duffy antigen receptor for chemokines [DARC] and P. vivax reticulocyte binding protein 2b/transferrin receptor [TfR1]) that are promising targets for therapeutic intervention. We optimized invasion assays with isogenic cultured reticulocytes. Using a receptor blockade approach with multiple P. vivax isolates, we found that all strains utilized both DARC and TfR1, but with significant variation in receptor usage. This suggests that P. vivax, like Plasmodium falciparum, uses alternative invasion pathways, with implications for pathogenesis and vaccine development.


Subject(s)
Antigens, CD , Duffy Blood-Group System , Malaria, Vivax , Plasmodium vivax , Receptors, Cell Surface , Receptors, Transferrin , Cells, Cultured , Humans , Plasmodium vivax/pathogenicity , Reticulocytes/parasitology
3.
PLoS Negl Trop Dis ; 14(3): e0008104, 2020 03.
Article in English | MEDLINE | ID: mdl-32119669

ABSTRACT

Approximately one-third of the global population is at risk of Plasmodium vivax infection, and an estimated 7.51 million cases were reported in 2017. Although, P. vivax research is currently limited by the lack of a robust continuous in vitro culture system for this parasite, recent work optimizing short-term ex vivo culture of P. vivax from cryopreserved isolates has facilitated quantitative assays on synchronous parasites. Pairing this improved culture system with low-input Smart-seq2 RNAseq library preparation, we sought to determine whether transcriptional profiling of P. vivax would provide insight into the differential survival of parasites in different culture media. To this end we probed the transcriptional signature of three different ex vivo P. vivax samples in four different culture media using only 1000 cells for each time point taken during the course of the intraerythrocytic development cycle (IDC). Using this strategy, we achieved similar quality transcriptional data to previously reported P. vivax transcriptomes. We found little effect with varying culture media on parasite transcriptional signatures, identified many novel gametocyte-specific genes from transcriptomes of FACS-isolated gametocytes, and determined invasion ligand expression in schizonts in biological isolates and across the IDC. In total, these data demonstrate the feasibility and utility of P. vivax RNAseq-based transcriptomic studies using minimal biomass input to maximize experimental capacity.


Subject(s)
Erythrocytes/parasitology , Gene Expression Profiling , Host-Pathogen Interactions , Malaria, Vivax/parasitology , Plasmodium vivax/growth & development , Adolescent , Child , Child, Preschool , Culture Media/chemistry , Female , Humans , Infant , Infant, Newborn , Male , Parasitology/methods , Plasmodium vivax/genetics , Sequence Analysis, RNA
4.
Am J Hematol ; 94(9): 963-974, 2019 09.
Article in English | MEDLINE | ID: mdl-31148215

ABSTRACT

Malaria pathogenesis is caused by the replication of Plasmodium parasites within the red blood cells (RBCs) of the vertebrate host. This selective pressure has favored the evolution of protective polymorphisms in erythrocyte proteins, a subset of which serve as cognate receptors for parasite invasion ligands. Recently, the generation of RBCs from immortalized hematopoietic stem cells (HSCs) has offered a more tractable system for genetic manipulation and long-term in vitro culture, enabling elucidation of the functional determinants of host susceptibility in vitro. Here we report the generation of an immortalized erythroid progenitor cell line (EJ cells) from as few as 100 000 peripheral blood mononuclear cells. It offers a robust method for the creation of customized model systems from small volumes of peripheral blood. The EJ cell differentiation mirrored erythropoiesis of primary HSCs, yielding orthochromatic erythroblasts and enucleated RBCs after eight days (ejRBCs). The ejRBCs supported invasion by both P. vivax and P. falciparum. To demonstrate the genetic tractability of this system, we used CRISPR/Cas9 to disrupt the Duffy Antigen/Receptor for Chemokines (DARC) gene, which encodes the canonical receptor of P. vivax in humans. Invasion of P. vivax into this DARC-knockout cell line was strongly inhibited providing direct genetic evidence that P. vivax requires DARC for RBC invasion. Further, genetic complementation of DARC restored P. vivax invasion. Taken together, the peripheral blood immortalization method presented here offers the capacity to generate biologically representative model systems for studies of blood-stage malaria invasion from the peripheral blood of donors harboring unique genetic backgrounds, or rare polymorphisms.


Subject(s)
Erythroid Precursor Cells , Malaria, Falciparum , Malaria, Vivax , Models, Biological , Peripheral Blood Stem Cells , Plasmodium falciparum/metabolism , Plasmodium vivax/metabolism , Cell Line, Transformed , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/physiology , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Malaria, Vivax/metabolism , Malaria, Vivax/pathology , Peripheral Blood Stem Cells/metabolism , Peripheral Blood Stem Cells/parasitology , Peripheral Blood Stem Cells/pathology
5.
Curr Opin Microbiol ; 46: 109-115, 2018 12.
Article in English | MEDLINE | ID: mdl-30366310

ABSTRACT

Plasmodium vivax is uniquely restricted to invading reticulocytes, the youngest of red blood cells. Parasite invasion relies on the sequential deployment of multiple parasite invasion ligands. Correct targeting of the host reticulocyte is mediated by two families of invasion ligands: the reticulocyte binding proteins (RBPs) and erythrocyte binding proteins (EBPs). The Duffy receptor has long been established as a key determinant for P. vivax invasion. However, recently, the RBP protein PvRBP2b has been shown to bind to transferrin receptor, which is expressed on reticulocytes but lost on normocytes, implicating the ligand-receptor in the reticulocyte tropism of P. vivax. Furthermore there is increasing evidence for P. vivax growth and sexual development in reticulocyte-enriched tissues such as the bone marrow.


Subject(s)
Malaria, Vivax/parasitology , Plasmodium vivax/physiology , Reticulocytes/parasitology , Animals , Host-Parasite Interactions , Humans , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Tropism
6.
Proc Natl Acad Sci U S A ; 115(28): 7350-7355, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29946035

ABSTRACT

Sickle cell trait (AS) confers partial protection against lethal Plasmodium falciparum malaria. Multiple mechanisms for this have been proposed, with a recent focus on aberrant cytoadherence of parasite-infected red blood cells (RBCs). Here we investigate the mechanistic basis of AS protection through detailed temporal mapping. We find that parasites in AS RBCs maintained at low oxygen concentrations stall at a specific stage in the middle of intracellular growth before DNA replication. We demonstrate that polymerization of sickle hemoglobin (HbS) is responsible for this growth arrest of intraerythrocytic P. falciparum parasites, with normal hemoglobin digestion and growth restored in the presence of carbon monoxide, a gaseous antisickling agent. Modeling of growth inhibition and sequestration revealed that HbS polymerization-induced growth inhibition following cytoadherence is the critical driver of the reduced parasite densities observed in malaria infections of individuals with AS. We conclude that the protective effect of AS derives largely from effective sequestration of infected RBCs into the hypoxic microcirculation.


Subject(s)
DNA Replication , DNA, Protozoan/biosynthesis , Erythrocytes, Abnormal/metabolism , Oxygen/metabolism , Plasmodium falciparum/metabolism , Sickle Cell Trait/metabolism , Antisickling Agents/pharmacology , Carbon Monoxide/pharmacology , Erythrocytes, Abnormal/parasitology , Humans , Malaria, Falciparum/metabolism , Sickle Cell Trait/parasitology
7.
mBio ; 9(3)2018 05 08.
Article in English | MEDLINE | ID: mdl-29739900

ABSTRACT

Plasmodium vivax causes heavy burdens of disease across malarious regions worldwide. Mature P. vivax asexual and transmissive gametocyte stages occur in the blood circulation, and it is often assumed that accumulation/sequestration in tissues is not an important phase in their development. Here, we present a systematic study of P. vivax stage distributions in infected tissues of nonhuman primate (NHP) malaria models as well as in blood from human infections. In a comparative analysis of the transcriptomes of P. vivax and Plasmodium falciparum blood-stage parasites, we found a conserved cascade of stage-specific gene expression despite the greatly different gametocyte maturity times of these two species. Using this knowledge, we validated a set of conserved asexual- and gametocyte-stage markers both by quantitative real-time PCR and by antibody assays of peripheral blood samples from infected patients and NHP (Aotus sp.). Histological analyses of P. vivax parasites in organs of 13 infected NHP (Aotus and Saimiri species) demonstrated a major fraction of immature gametocytes in the parenchyma of the bone marrow, while asexual schizont forms were enriched to a somewhat lesser extent in this region of the bone marrow as well as in sinusoids of the liver. These findings suggest that the bone marrow is an important reservoir for gametocyte development and proliferation of malaria parasites.IMPORTANCEPlasmodium vivax malaria continues to cause major public health burdens worldwide. Yet, significant knowledge gaps in the basic biology and epidemiology of P. vivax malaria remain, largely due to limited available tools for research and diagnostics. Here, we present a systematic examination of tissue sequestration during P. vivax infection. Studies of nonhuman primates and malaria patients revealed enrichment of developing sexual stages (gametocytes) and mature replicative stages (schizonts) in the bone marrow and liver, relative to those present in peripheral blood. Identification of the bone marrow as a major P. vivax tissue reservoir has important implications for parasite diagnosis and treatment.


Subject(s)
Bone Marrow/parasitology , Malaria, Falciparum/parasitology , Malaria, Vivax/parasitology , Plasmodium falciparum/growth & development , Plasmodium vivax/growth & development , Animals , Aotidae , Female , Humans , Male , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Plasmodium vivax/genetics , Plasmodium vivax/isolation & purification , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Saimiri
8.
Article in English | MEDLINE | ID: mdl-29378713

ABSTRACT

Plasmodium vivax chloroquine resistance has been documented in nearly every region where this malaria-causing parasite is endemic. Unfortunately, P. vivax resistance surveillance and drug discovery are challenging due to the low parasitemias of patient isolates and poor parasite survival through ex vivo maturation that reduce the sensitivity and scalability of current P. vivax antimalarial assays. Using cryopreserved patient isolates from Brazil and fresh patient isolates from India, we established a robust enrichment method for P. vivax parasites. We next performed a medium screen for formulations that enhance ex vivo survival. Finally, we optimized an isotopic metabolic labeling assay for measuring P. vivax maturation and its sensitivity to antimalarials. A KCl Percoll density gradient enrichment method increased parasitemias from small-volume ex vivo isolates by an average of >40-fold. The use of Iscove's modified Dulbecco's medium for P. vivax ex vivo culture approximately doubled the parasite survival through maturation. Coupling these with [3H]hypoxanthine metabolic labeling permitted sensitive and robust measurements of parasite maturation, which was used to measure the sensitivities of Brazilian P. vivax isolates to chloroquine and several novel antimalarials. These techniques can be applied to rapidly and robustly assess the P. vivax isolate sensitivities to antimalarials for resistance surveillance and drug discovery.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Parasitic Sensitivity Tests/methods , Plasmodium vivax/drug effects , Brazil , Humans , India
9.
Cell ; 171(7): 1532-1544.e15, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29129376

ABSTRACT

Transmission represents a population bottleneck in the Plasmodium life cycle and a key intervention target of ongoing efforts to eradicate malaria. Sexual differentiation is essential for this process, as only sexual parasites, called gametocytes, are infective to the mosquito vector. Gametocyte production rates vary depending on environmental conditions, but external stimuli remain obscure. Here, we show that the host-derived lipid lysophosphatidylcholine (LysoPC) controls P. falciparum cell fate by repressing parasite sexual differentiation. We demonstrate that exogenous LysoPC drives biosynthesis of the essential membrane component phosphatidylcholine. LysoPC restriction induces a compensatory response, linking parasite metabolism to the activation of sexual-stage-specific transcription and gametocyte formation. Our results reveal that malaria parasites can sense and process host-derived physiological signals to regulate differentiation. These data close a critical knowledge gap in parasite biology and introduce a major component of the sexual differentiation pathway in Plasmodium that may provide new approaches for blocking malaria transmission.


Subject(s)
Lysophosphatidylcholines/metabolism , Malaria/parasitology , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Animals , Female , Humans , Malaria/immunology , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Plasmodium berghei/physiology , Reproduction
11.
Blood ; 126(16): 1940-8, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26324704

ABSTRACT

Factor XIII(a) [FXIII(a)] stabilizes clots and increases resistance to fibrinolysis and mechanical disruption. FXIIIa also mediates red blood cell (RBC) retention in contracting clots and determines venous thrombus size, suggesting FXIII(a) is a potential target for reducing thrombosis. However, the mechanism by which FXIIIa retains RBCs in clots is unknown. We determined the effect of FXIII(a) on human and murine clot weight and composition. Real-time microscopy revealed extensive RBC loss from clots formed in the absence of FXIIIa activity, and RBCs exhibited transient deformation as they exited the clots. Fibrin band-shift assays and flow cytometry did not reveal crosslinking of fibrin or FXIIIa substrates to RBCs, suggesting FXIIIa does not crosslink RBCs directly to the clot. RBCs were retained in clots from mice deficient in α2-antiplasmin, thrombin-activatable fibrinolysis inhibitor, or fibronectin, indicating RBC retention does not depend on these FXIIIa substrates. RBC retention in clots was positively correlated with fibrin network density; however, FXIIIa inhibition reduced RBC retention at all network densities. FXIIIa inhibition reduced RBC retention in clots formed with fibrinogen that lacks γ-chain crosslinking sites, but not in clots that lack α-chain crosslinking sites. Moreover, FXIIIa inhibitor concentrations that primarily block α-, but not γ-, chain crosslinking decreased RBC retention in clots. These data indicate FXIIIa-dependent retention of RBCs in clots is mediated by fibrin α-chain crosslinking. These findings expose a newly recognized, essential role for fibrin crosslinking during whole blood clot formation and consolidation and establish FXIIIa activity as a key determinant of thrombus composition and size.


Subject(s)
Blood Coagulation Factors/metabolism , Blood Coagulation/physiology , Erythrocytes/metabolism , gamma-Glutamyltransferase/metabolism , Animals , Blood Coagulation Factors/genetics , Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Hemorrhagic Disorders/genetics , Hemorrhagic Disorders/metabolism , Humans , Mice , Mice, Knockout , alpha-2-Antiplasmin/deficiency , alpha-2-Antiplasmin/genetics , alpha-2-Antiplasmin/metabolism , gamma-Glutamyltransferase/genetics
12.
Am J Trop Med Hyg ; 91(4): 824-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25184829

ABSTRACT

Because of emerging resistance to existing drugs, new chemical classes of antimalarial drugs are urgently needed. We have rationally designed a library of compounds that were predicted to accumulate in the digestive vacuole and then decrystallize hemozoin by breaking the iron carboxylate bond in hemozoin. We report the synthesis of 16 naphthothiazolium salts with amine-bearing side chains and their activities against the erythrocytic stage of Plasmodium falciparum in vitro. KSWI-855, the compound with the highest efficacy against the asexual stages of P. falciparum in vitro, also had in vitro activity against P. falciparum gametocytes and in vivo activity against P. berghei in a murine malaria model.


Subject(s)
Antimalarials/chemical synthesis , Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Surface-Active Agents/pharmacology , Amines/chemistry , Animals , Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacology , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Malaria, Falciparum/parasitology , Mice , Mice, Inbred C57BL , Naphthoquinones/chemistry , Parasitic Sensitivity Tests , Plasmodium berghei/drug effects , Surface-Active Agents/chemical synthesis , Thiourea/chemistry
13.
Nat Commun ; 5: 4446, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-25059846

ABSTRACT

Iron deficiency and malaria have similar global distributions, and frequently co-exist in pregnant women and young children. Where both conditions are prevalent, iron supplementation is complicated by observations that iron deficiency anaemia protects against falciparum malaria, and that iron supplements increase susceptibility to clinically significant malaria, but the mechanisms remain obscure. Here, using an in vitro parasite culture system with erythrocytes from iron-deficient and replete human donors, we demonstrate that Plasmodium falciparum infects iron-deficient erythrocytes less efficiently. In addition, owing to merozoite preference for young erythrocytes, iron supplementation of iron-deficient individuals reverses the protective effects of iron deficiency. Our results provide experimental validation of field observations reporting protective effects of iron deficiency and harmful effects of iron administration on human malaria susceptibility. Because recovery from anaemia requires transient reticulocytosis, our findings imply that in malarious regions iron supplementation should be accompanied by effective measures to prevent falciparum malaria.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Erythrocytes/parasitology , Iron/blood , Iron/pharmacology , Plasmodium falciparum/pathogenicity , Adult , Anemia, Iron-Deficiency/parasitology , Dietary Supplements , Disease Susceptibility , Humans , Malaria, Falciparum/prevention & control , Middle Aged , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Young Adult
14.
PLoS One ; 9(7): e101041, 2014.
Article in English | MEDLINE | ID: mdl-24984000

ABSTRACT

Plasmodium falciparum invasion of host erythrocytes is essential for the propagation of the blood stage of malaria infection. Additionally, the brief extracellular merozoite stage of P. falciparum represents one of the rare windows during which the parasite is directly exposed to the host immune response. Therefore, efficient invasion of the host erythrocyte is necessary not only for productive host erythrocyte infection, but also for evasion of the immune response. Host traits, such as hemoglobinopathies and differential expression of erythrocyte invasion ligands, can protect individuals from malaria by impeding parasite erythrocyte invasion. Here we combine RBC barcoding with flow cytometry to study P. falciparum invasion. This novel high-throughput method allows for the (i) direct comparison of P. falciparum invasion into different erythrocyte populations and (ii) assessment of the impact of changing erythrocyte population dynamics on P. falciparum invasion.


Subject(s)
Erythrocytes/parasitology , Plasmodium falciparum/physiology , Animals , Flow Cytometry , Humans
15.
Front Pharmacol ; 5: 84, 2014.
Article in English | MEDLINE | ID: mdl-24834053

ABSTRACT

Iron deficiency affects one quarter of the world's population and causes significant morbidity, including detrimental effects on immune function and cognitive development. Accordingly, the World Health Organization (WHO) recommends routine iron supplementation in children and adults in areas with a high prevalence of iron deficiency. However, a large body of clinical and epidemiological evidence has accumulated which clearly demonstrates that host iron deficiency is protective against falciparum malaria and that host iron supplementation may increase the risk of malaria. Although many effective antimalarial treatments and preventive measures are available, malaria remains a significant public health problem, in part because the mechanisms of malaria pathogenesis remain obscured by the complexity of the relationships that exist between parasite virulence factors, host susceptibility traits, and the immune responses that modulate disease. Here we review (i) the clinical and epidemiological data that describes the relationship between host iron status and malaria infection and (ii) the current understanding of the biological basis for these clinical and epidemiological observations.

16.
J Clin Microbiol ; 52(6): 2144-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24740073

ABSTRACT

Malaria parasite infections that are only detectable by molecular methods are highly prevalent and represent a potential transmission reservoir. The methods used to detect these infections are not standardized, and their operating characteristics are often unknown. We designed a proficiency panel of Plasmodium spp. in order to compare the accuracy of parasite detection of molecular protocols used by labs in a clinical trial consortium. Ten dried blood spots (DBSs) were assembled that contained P. falciparum, P. vivax, P. malariae, and P. ovale; DBSs contained either a single species or a species mixed with P. falciparum. DBS panels were tested in 9 participating laboratories in a masked fashion. Of 90 tests, 68 (75.6%) were correct; there were 20 false-negative results and 2 false positives. The detection rate was 77.8% (49/63) for P. falciparum, 91.7% (11/12) for P. vivax, 83.3% (10/12) for P. malariae, and 70% (7/10) for P. ovale. Most false-negative P. falciparum results were from samples with an estimated ≤ 5 parasites per µl of blood. Between labs, accuracy ranged from 100% to 50%. In one lab, the inability to detect species in mixed-species infections prompted a redesign and improvement of the assay. Most PCR-based protocols were able to detect P. falciparum and P. vivax at higher densities, but these assays may not reliably detect parasites in samples with low P. falciparum densities. Accordingly, formal quality assurance for PCR should be employed whenever this method is used for diagnosis or surveillance. Such efforts will be important if PCR is to be widely employed to assist malaria elimination efforts.


Subject(s)
Biomedical Research/standards , Laboratory Proficiency Testing , Malaria/diagnosis , Molecular Diagnostic Techniques/standards , Plasmodium/isolation & purification , Polymerase Chain Reaction/standards , Biomedical Research/methods , False Negative Reactions , Humans , Malaria/parasitology , Molecular Diagnostic Techniques/methods , Plasmodium/classification , Plasmodium/genetics , Polymerase Chain Reaction/methods , Quality Control , Sensitivity and Specificity
17.
PLoS One ; 7(5): e37171, 2012.
Article in English | MEDLINE | ID: mdl-22629364

ABSTRACT

Iron chelators for the treatment of malaria have proven therapeutic activity in vitro and in vivo in both humans and mice, but their clinical use is limited by the unsuitable absorption and pharmacokinetic properties of the few available iron chelators. FBS0701, (S)3"-(HO)-desazadesferrithiocin-polyether [DADFT-PE], is an oral iron chelator currently in Phase 2 human studies for the treatment of transfusional iron overload. The drug has very favorable absorption and pharmacokinetic properties allowing for once-daily use to deplete circulating free iron with human plasma concentrations in the high µM range. Here we show that FBS0701 has inhibition concentration 50% (IC(50)) of 6 µM for Plasmodium falciparum in contrast to the IC(50) for deferiprone and deferoxamine at 15 and 30 µM respectively. In combination, FBS0701 interfered with artemisinin parasite inhibition and was additive with chloroquine or quinine parasite inhibition. FBS0701 killed early stage P. falciparum gametocytes. In the P. berghei Thompson suppression test, a single dose of 100 mg/kg reduced day three parasitemia and prolonged survival, but did not cure mice. Treatment with a single oral dose of 100 mg/kg one day after infection with 10 million lethal P. yoelii 17XL cured all the mice. Pretreatment of mice with a single oral dose of FBS0701 seven days or one day before resulted in the cure of some mice. Plasma exposures and other pharmacokinetics parameters in mice of the 100 mg/kg dose are similar to a 3 mg/kg dose in humans. In conclusion, FBS0701 demonstrates a single oral dose cure of the lethal P. yoelii model. Significantly, this effect persists after the chelator has cleared from plasma. FBS0701 was demonstrated to remove labile iron from erythrocytes as well as enter erythrocytes to chelate iron. FBS0701 may find clinically utility as monotherapy, a malarial prophylactic or, more likely, in combination with other antimalarials.


Subject(s)
Antimalarials/therapeutic use , Ethyl Ethers/therapeutic use , Iron Chelating Agents/therapeutic use , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Thiazoles/therapeutic use , Animals , Antimalarials/pharmacology , Disease Models, Animal , Ethyl Ethers/pharmacology , Iron Chelating Agents/pharmacology , Mice , Parasitemia/drug therapy , Thiazoles/pharmacology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...