1
|
Manh ND, Thanh NV, Quang HH, Van NTT, San NN, Phong NC, Birrell GW, Edgel KA, Martin NJ, Edstein MD, Chavchich M. Therapeutic efficacy of pyronaridine-artesunate (Pyramax) in treating Plasmodium vivax malaria in the central highlands of Vietnam. Antimicrob Agents Chemother 2024; 68:e0004424. [PMID: 39046237 PMCID: PMC11373200 DOI: 10.1128/aac.00044-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/19/2024] [Indexed: 07/25/2024] Open
Abstract
The emergence and spread of chloroquine-resistant Plasmodium vivax have necessitated the assessment of alternative blood schizonticidal drugs. In Vietnam, chloroquine-resistant P. vivax malaria has been reported. In an open-label, single-arm trial, the safety, tolerability, and efficacy of pyronaridine-artesunate (Pyramax, PA) was evaluated in Dak Nong province, Vietnam. A 3-day course of PA was administered to adults and children (≥20 kg) infected with P. vivax. Patients also received primaquine (0.25 mg/kg daily for 14 days). PA was well tolerated with transient asymptomatic increases in liver transaminases. The per-protocol proportion of patients with day 42 PCR-unadjusted adequate clinical and parasitological response was 96.0% (95% CI, 84.9%-99.0%, n = 48/50). The median parasite clearance time was 12 h (range, 12-36 h), with a median fever clearance time of 24 h (range, 12-60 h). Single nucleotide polymorphisms (SNPs) as potential genetic markers of reduced drug susceptibility were analyzed in three putative drug resistance markers, Pvcrt-o, Pvmdr1, and PvK12. Insertion at position K10 of the Pvcrt-o gene was found in 74.6% (44/59) of isolates. Pvmdr1 SNPs at Y976F and F1076L were present in 61% (36/59) and 78% (46/59), respectively. Amplification of Pvmdr1 gene (two copies) was found in 5.1% (3/59) of parasite samples. Only 5.1% (3/59) of isolates had mutation 552I of the PvK12 gene. Overall, PA rapidly cleared P. vivax blood asexual stages and was highly efficacious in treating vivax malaria, with no evidence of artemisinin resistance found. PA provides an alternative to chloroquine treatment for vivax malaria in Vietnam. CLINICAL TRIALS This study is registered with the Australian New Zealand Clinical Trials Registry as ACTRN12618001429246.
Collapse
Affiliation(s)
- Nguyen Duc Manh
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Nguyen Van Thanh
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Huynh Hong Quang
- Vietnam Ministry of Health Institute of Malariology, Parasitology and Entomology, Qui Nhon, Vietnam
| | | | - Nguyen Ngoc San
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Nguen Chinh Phong
- Vietnam People's Army Military Institute of Preventive Medicine, Hanoi, Vietnam
| | - Geoffrey W Birrell
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | | | | | - Michael D Edstein
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Marina Chavchich
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| |
Collapse
|
2
|
Hazzard B, Sá JM, Bogale HN, Pascini TV, Ellis AC, Amin S, Armistead JS, Adams JH, Wellems TE, Serre D. Single-cell analyses of polyclonal Plasmodium vivax infections and their consequences on parasite transmission. Nat Commun 2024; 15:7625. [PMID: 39223117 PMCID: PMC11369214 DOI: 10.1038/s41467-024-51949-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Most Plasmodium vivax infections contain genetically distinct parasites, but the consequences of this polyclonality on the development of asexual parasites, their sexual differentiation, and their transmission remain unknown. We describe infections of Saimiri monkeys with two strains of P. vivax and the analyses of 80,024 parasites characterized by single cell RNA sequencing and individually genotyped. In our model, consecutive inoculations fail to establish polyclonal infections. By contrast, simultaneous inoculations of two strains lead to sustained polyclonal infections, although without detectable differences in parasite regulation or sexual commitment. Analyses of sporozoites dissected from mosquitoes fed on coinfected monkeys show that all genotypes are successfully transmitted to mosquitoes. However, after sporozoite inoculation, not all genotypes contribute to the subsequent blood infections, highlighting an important bottleneck during pre-erythrocytic development. Overall, these studies provide new insights on the mechanisms regulating the establishment of polyclonal P. vivax infections and their consequences for disease transmission.
Collapse
Affiliation(s)
- Brittany Hazzard
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Haikel N Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tales V Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angela C Ellis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shuchi Amin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer S Armistead
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - John H Adams
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious, Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Pinheiro AAS, Caruso-Neves C, Rocco PRM. Extracellular vesicles in malaria: Pathogenesis, diagnosis and therapy. CURRENT TOPICS IN MEMBRANES 2024; 94:107-132. [PMID: 39370204 DOI: 10.1016/bs.ctm.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Malaria is a life-threatening disease caused by parasites from the genus Plasmodium. Five species can cause malaria in humans, with Plasmodium vivax being the most common in many countries and Plasmodium falciparum having the highest lethality, which can lead to cerebral malaria. Extracellular vesicles (EVs) are in focus in malaria research to better understand pathogenesis, diagnosis, therapy, and prognosis. Malaria-causing parasites use EVs to transfer their molecules to host cells, a mechanism that significantly contributes to parasite survival and successful infection. EVs have thus emerged as an essential component of the immunopathological cascade of malaria, playing a pivotal role in disease progression and severity. This chapter discusses the epidemiology and pathogenesis of malaria and the role of EVs as new diagnostic and therapeutic tools, emphasizing their potential clinical significance.
Collapse
Affiliation(s)
- Ana Acacia S Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
4
|
Cao Y, Hayashi CTH, Araujo MDS, Tripathi AK, Andrade AO, Medeiros JF, Vinetz J, Kumar N. Evaluation of combination vaccines targeting transmission of Plasmodium falciparum and P. vivax. Vaccine 2024; 42:126140. [PMID: 39033079 PMCID: PMC11338703 DOI: 10.1016/j.vaccine.2024.07.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Transmission-blocking vaccines interrupting malaria transmission within mosquitoes represent an ideal public health tool to eliminate malaria at the population level. Plasmodium falciparum and P. vivax account for more than 90% of the global malaria burden, co-endemic in many regions of the world. P25 and P48/45 are two leading candidates for both species and have shown promising transmission-blocking activity in preclinical and clinical studies. However, neither of these target antigens as individual vaccines has induced complete transmission inhibition in mosquitoes. In this study, we assessed immunogenicity of combination vaccines based on P25 and P48/45 using a DNA vaccine platform to broaden vaccine specificity against P. falciparum and P. vivax. Individual DNA vaccines encoding Pvs25, Pfs25, Pvs48/45 and Pfs48/45, as well as various combinations including (Pvs25 + Pvs48/45), (Pfs25 + Pfs48/45), (Pvs25 + Pfs25), and (Pvs48/45 + Pfs48/45), were evaluated in mice using in vivo electroporation. Potent antibody responses were induced in mice immunized with individual and combination DNA vaccines, and specific antibody responses were not compromised when combinations of DNA vaccines were evaluated against individual DNA vaccines. The anti-Pvs25 IgG from individual and combination groups revealed concentration-dependent transmission-reducing activity (TRA) in direct membrane feeding assays (DMFA) using blood from P. vivax-infected donors in Brazil and independently in ex vivo MFA using Pvs25-transgenic P. berghei. Similarly, anti-Pfs25 and anti-Pfs48/45 IgGs from mice immunized with Pfs25 and Pfs48/45 DNA vaccines individually and in various combinations revealed antibody dose-dependent TRA in standard membrane feeding assays (SMFA) using culture-derived P. falciparum gametocytes. However, antibodies induced by immunization with Pvs48/45 DNA vaccines were ineffective in DMFA and require further vaccine construct optimization, considering the possibility of induction of both transmission-blocking and transmission-enhancing antibodies revealed by competition ELISA. These studies provide a rationale for combining multiple antigens to simultaneously target transmission of malaria caused by P. falciparum and P. vivax.
Collapse
MESH Headings
- Malaria Vaccines/immunology
- Malaria Vaccines/administration & dosage
- Animals
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/transmission
- Malaria, Falciparum/immunology
- Plasmodium falciparum/immunology
- Plasmodium falciparum/genetics
- Plasmodium vivax/immunology
- Plasmodium vivax/genetics
- Malaria, Vivax/prevention & control
- Malaria, Vivax/transmission
- Malaria, Vivax/immunology
- Mice
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Antibodies, Protozoan/immunology
- Antibodies, Protozoan/blood
- Female
- Vaccines, Combined/immunology
- Vaccines, Combined/administration & dosage
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Mice, Inbred BALB C
- Humans
Collapse
Affiliation(s)
- Yi Cao
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA
| | - Clifford T H Hayashi
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA
| | - Maisa da Silva Araujo
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Saúde Pública, Faculdade de Saúde Pública, Universidade Federal de São Paulo, São Paulo 01246-904, SP, Brazil
| | - Abhai K Tripathi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Alice Oliveira Andrade
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Saúde Pública, Faculdade de Saúde Pública, Universidade Federal de São Paulo, São Paulo 01246-904, SP, Brazil
| | - Jansen Fernandes Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Fiocruz Rondônia 76812-245, Brazil
| | - Joseph Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Sciences, Faculty of Sciences, and Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nirbhay Kumar
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA.
| |
Collapse
|
5
|
Omondi P, Musyoka B, Okai T, Kongere J, Kagaya W, Chan CW, Ngara M, Kanoi BN, Kido Y, Gitaka J, Kaneko A. Non-random distribution of Plasmodium Species infections and associated clinical features in children in the lake Victoria region, Kenya, 2012-2018. Trop Med Health 2024; 52:52. [PMID: 39103954 PMCID: PMC11299388 DOI: 10.1186/s41182-024-00622-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND While Plasmodium falciparum (Pf) stands out as the most lethal malaria parasite species in humans, the impact of other species should not be dismissed. Moreover, there is a notable lack of understanding of mixed-species infections and their clinical implications. METHODS We conducted eight school-based cross-sectional malariometric surveys in the Lake Victoria region of western Kenya between January-February 2012 and September-October 2018. In each survey, a minimum of 100 children aged 3 to 15 years were randomly chosen from a school in Ungoye village on the mainland and as well as from each school selected in every catchment area on Mfangano island. Plasmodium infection was determined by microscopy and nested polymerase chain reaction (PCR). The multiple-kind lottery (MKL) model calculated the expected distribution of Plasmodium infections in the population and compared it to observed values using a chi-squared test (χ2). RESULTS The Plasmodium prevalence was 25.9% (2521/9724) by microscopy and 51.1% (4969/9724) by PCR. Among all infections detected by PCR, Pf, P. malariae (Pm), and P. ovale (Po) mono-infections were 58.6%, 3.1%, and 1.8%, respectively. Pf/Pm, Pf/Po, Pm/Po, and Pf/Pm/Po co-infections were 23.5%, 4.3%, 0.1%, and 8.6%, respectively. MKL modelling revealed non-random distributions, with frequencies of Pf/Pm and Pf/Pm/Po co-infections being significantly higher than expected (χ2 = 3385.60, p < 0.001). Pf co-infections with Pm and Po were associated with a decreased risk of fever (aOR 0.64, 95% CI 0.46-0.83; p = 0.01) and increased risks of splenomegaly (aOR 12.79, 95% CI 9.69-16.9; p < 0.001) and anaemia (aOR 2.57, 95% CI 2.09-3.15; p < 0.001), compared to single-species infections. CONCLUSION This study sheds light on the potential interaction between Pf and Pm and/or Po. Given the clinical significance of mixed-species infections, improved diagnostics, and case management of Pm and Po are urgently needed.
Collapse
Affiliation(s)
- Protus Omondi
- Department of Virology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Brian Musyoka
- Department of Virology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Takatsugu Okai
- Department of Virology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - James Kongere
- Department of Parasitology/ Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Wataru Kagaya
- Department of Eco-Epidemiology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Chim W Chan
- Department of Parasitology/ Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Mtakai Ngara
- Department of Clinical Medicine, Mount Kenya University, Thika, Kenya
- Island Malaria Group, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Bernard N Kanoi
- Department of Clinical Medicine, Mount Kenya University, Thika, Kenya
| | - Yasutoshi Kido
- Department of Virology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Jesse Gitaka
- Department of Clinical Medicine, Mount Kenya University, Thika, Kenya
| | - Akira Kaneko
- Department of Parasitology/ Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan.
- Island Malaria Group, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
6
|
Dickey TH, McAleese H, Salinas ND, Lambert LE, Tolia NH. Structure-based design of a Plasmodium vivax Duffy-binding protein immunogen focuses the antibody response to functional epitopes. Protein Sci 2024; 33:e5095. [PMID: 38988315 PMCID: PMC11237555 DOI: 10.1002/pro.5095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 07/12/2024]
Abstract
The Duffy-binding protein (DBP) is a promising antigen for a malaria vaccine that would protect against clinical symptoms caused by Plasmodium vivax infection. Region II of DBP (DBP-II) contains the receptor-binding domain that engages host red blood cells, but DBP-II vaccines elicit many non-neutralizing antibodies that bind distal to the receptor-binding surface. Here, we engineered a truncated DBP-II immunogen that focuses the immune response to the receptor-binding surface. This immunogen contains the receptor-binding subdomain S1S2 and lacks the immunodominant subdomain S3. Structure-based computational design of S1S2 identified combinatorial amino acid changes that stabilized the isolated S1S2 without perturbing neutralizing epitopes. This immunogen elicited DBP-II-specific antibodies in immunized mice that were significantly enriched for blocking activity compared to the native DBP-II antigen. This generalizable design process successfully stabilized an integral core fragment of a protein and focused the immune response to desired epitopes to create a promising new antigen for malaria vaccine development.
Collapse
MESH Headings
- Protozoan Proteins/immunology
- Protozoan Proteins/chemistry
- Protozoan Proteins/genetics
- Antigens, Protozoan/immunology
- Antigens, Protozoan/chemistry
- Antigens, Protozoan/genetics
- Plasmodium vivax/immunology
- Animals
- Malaria Vaccines/immunology
- Malaria Vaccines/chemistry
- Epitopes/immunology
- Epitopes/chemistry
- Mice
- Antibodies, Protozoan/immunology
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Models, Molecular
- Malaria, Vivax/immunology
- Malaria, Vivax/prevention & control
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Thayne H. Dickey
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Holly McAleese
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Nichole D. Salinas
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Lynn E. Lambert
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| | - Niraj H. Tolia
- Host‐Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious DiseasesNational Institutes of Health (NIH)BethesdaMarylandUSA
| |
Collapse
|
7
|
Nakayama K, Haraguchi A, Hakozaki J, Nakamura S, Kusakisako K, Ikadai H. The C-terminal region of the Plasmodium berghei gamete surface 184-kDa protein Pb184 contributes to fertilization and male gamete binding to the residual body. Parasit Vectors 2024; 17:304. [PMID: 39003498 PMCID: PMC11246575 DOI: 10.1186/s13071-024-06374-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Malaria, a global health concern, is caused by parasites of the Plasmodium genus, which undergo gametogenesis in the midgut of mosquitoes after ingestion of an infected blood meal. The resulting male and female gametes fuse to form a zygote, which differentiates into a motile ookinete. After traversing the midgut epithelium, the ookinete differentiates into an oocyst on the basal side of the epithelium. METHODS Membrane proteins with increased gene expression levels from the gamete to oocyst stages in P. berghei were investigated utilizing PlasmoDB, the functional genomic database for Plasmodium spp. Based on this analysis, we selected the 184-kDa membrane protein, Pb184, for further study. The expression of Pb184 was further confirmed through immunofluorescence staining, following which we examined whether Pb184 is involved in fertilization using antibodies targeting the C-terminal region of Pb184 and biotin-labeled C-terminal region peptides of Pb184. RESULTS Pb184 is expressed on the surface of male and female gametes. The antibody inhibited zygote and ookinete formation in vitro. When mosquitoes were fed on parasite-infected blood containing the antibody, oocyst formation decreased on the second day after feeding. Synthesized biotin-labeled peptides matching the C-terminal region of Pb184 bound to the female gamete and the residual body of male gametes, and inhibited differentiation into ookinetes in the in vitro culture system. CONCLUSIONS These results may be useful for the further studying the fertilization mechanism of Plasmodium protozoa. There is also the potential for their application as future tools to prevent malaria transmission.
Collapse
Affiliation(s)
- Kazuhiko Nakayama
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Asako Haraguchi
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Jun Hakozaki
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Sakure Nakamura
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Kodai Kusakisako
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Hiromi Ikadai
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan.
| |
Collapse
|
8
|
Hazzard B, Sá JM, Bogale HN, Pascini TV, Ellis AC, Amin S, Armistead JS, Adams JH, Wellems TE, Serre D. Single-cell analyses of polyclonal Plasmodium vivax infections and their consequences on parasite transmission. RESEARCH SQUARE 2024:rs.3.rs-3888175. [PMID: 38410426 PMCID: PMC10896380 DOI: 10.21203/rs.3.rs-3888175/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Most Plasmodium vivax infections contain genetically distinct parasites, but the consequences of this polyclonality on the development of asexual parasites, their sexual differentiation, and their transmission remain unknown. We describe infections of Saimiri monkeys with two strains of P. vivax and the analyses of 117,350 parasites characterized by single cell RNA sequencing and individually genotyped. In our model, consecutive inoculations fail to establish polyclonal infections. By contrast, simultaneous inoculations of two strains lead to sustained polyclonal infections, although without detectable differences in parasite regulation or sexual commitment. Analyses of sporozoites dissected from mosquitoes fed on coinfected monkeys show that all genotypes are successfully transmitted to mosquitoes. However, after sporozoite inoculation, not all genotypes contribute to the subsequent blood infections, highlighting an important bottleneck during pre-erythrocytic development. Overall, these studies provide new insights on the mechanisms regulating the establishment of polyclonal P. vivax infections and their consequences for disease transmission.
Collapse
Affiliation(s)
- Brittany Hazzard
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Juliana M. Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Haikel N. Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tales V. Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Angela C. Ellis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Shuchi Amin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jennifer S. Armistead
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - John H. Adams
- Center for Global Health and Inter-Disciplinary Research, College of Public Health, University of South Florida, Tampa, USA
| | - Thomas E. Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Lead contact
| |
Collapse
|
9
|
Liu H, Xu JW, Deng DW, Yaw B, Nbwi HS, Wei C, Zhou XW, Li JX. Artemisinin-naphthoquine plus lower-dose primaquine to treat and prevent recurrence of Plasmodium vivax malaria: an open-label randomized and non-inferiority trial. Parasit Vectors 2024; 17:28. [PMID: 38254128 PMCID: PMC10804781 DOI: 10.1186/s13071-023-06058-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/15/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Plasmodium vivax malaria, with the widest geographic distribution, can cause severe disease and death. Primaquine is the main licensed antimalarial drug that can kill hypnozoites. The dose-dependent acute haemolysis in individuals with glucose-6-phospate dehydrogenase (G6PD) deficiency is the main safety concern when using primaquine. The recommended treatment regimen for P. vivax malaria is chloroquine plus primaquine for 14 days (CQPQ14) in Myanmar. The study aimed to evaluate the therapeutic efficacy, safety and adherence for the regimen of artemisinin-naphthoquine plus primaquine for 3 days (ANPQ3) in patients with P. vivax infections compared to those with CQPQ14. METHODS The patients in the ANPQ3 group were given fixed-dose artemisinin-naphthoquine (a total 24.5 mg/kg bodyweight) plus a lower total primaquine dose (0.9 mg/kg bodyweight) for 3 days. The patients in the CQPQ14 group were given a total chloroquine dose of 30 mg/kg body weight for 3 days plus a total primaquine dose of 4.2 mg/kg bodyweight for 14 days. All patients were followed up for 365 days. RESULTS A total of 288 patients completed follow-up, 172 in the ANPQ3 group and 116 in the CQPQ14 group. The first recurrence patients were detected by day 58 in both groups. By day 182, 16 recurrences had been recorded: 12 (7.0%) patients in the ANPQ3 group and 4 (3.4%) in the CQPQ14 group. The difference in recurrence-free patients was 3.5 (-8.6 to 1.5) percentage points between ANPQ3 and CQPQ14 group (P = 0.2946). By day 365, the percentage of recurrence-free patients was not significant between the two groups (P = 0.2257). Mean fever and parasite clearance time of ANPQ3 group were shorter than those in CQPQ14 group (P ≤ 0.001). No severe adverse effect was observed in ANPQ3 group, but five (3.9%) patients had acute haemolysis in CQPQ14 group (P = 0.013). Medication percentage of ANPQ3 group was significantly higher than that of CQPQ14 group (P < 0.0001). CONCLUSIONS Both ANPQ3 and CQPQ14 promised clinical cure efficacy, and the radical cure efficacy was similar between the ANPQ3 and CQPQ14 group. ANPQ3 clears fever and parasites faster than CQPQ14. ANPQ3 is safer and shows better patient adherence to the regimen for treatment of P. vivax malaria along the China-Myanmar border. TRIAL REGISTRATION ChiCTR-INR-17012523. Registered 31 August 2017, https://www.chictr.org.cn/showproj.html?proj=21352.
Collapse
Affiliation(s)
- Hui Liu
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Key Laboratory of Vector-Borne Disease Control and Research, Yunnan International Joint Laboratory of Tropical Infectious Diseases, Pu'er, China, 665000.
| | - Jian-Wei Xu
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Key Laboratory of Vector-Borne Disease Control and Research, Yunnan International Joint Laboratory of Tropical Infectious Diseases, Pu'er, China, 665000
| | - Dao-Wei Deng
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Key Laboratory of Vector-Borne Disease Control and Research, Yunnan International Joint Laboratory of Tropical Infectious Diseases, Pu'er, China, 665000
| | - Bi Yaw
- Laiza City Hospital, Laiza Town, Kachin Special Region II, Myanmar
| | | | - Chun Wei
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Key Laboratory of Vector-Borne Disease Control and Research, Yunnan International Joint Laboratory of Tropical Infectious Diseases, Pu'er, China, 665000
| | - Xing-Wu Zhou
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Key Laboratory of Vector-Borne Disease Control and Research, Yunnan International Joint Laboratory of Tropical Infectious Diseases, Pu'er, China, 665000
| | - Jian-Xiong Li
- Yunnan Institute of Parasitic Diseases, Yunnan Provincial Key Laboratory of Vector-Borne Disease Control and Research, Yunnan International Joint Laboratory of Tropical Infectious Diseases, Pu'er, China, 665000
| |
Collapse
|
10
|
Kumar A, Singh PP, Tyagi S, Hari Kishan Raju K, Sahu SS, Rahi M. Vivax malaria: a possible stumbling block for malaria elimination in India. Front Public Health 2024; 11:1228217. [PMID: 38259757 PMCID: PMC10801037 DOI: 10.3389/fpubh.2023.1228217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Plasmodium vivax is geographically the most widely dispersed human malaria parasite species. It has shown resilience and a great deal of adaptability. Genomic studies suggest that P. vivax originated from Asia or Africa and moved to the rest of the world. Although P. vivax is evolutionarily an older species than Plasmodium falciparum, its biology, transmission, pathology, and control still require better elucidation. P. vivax poses problems for malaria elimination because of the ability of a single primary infection to produce multiple relapses over months and years. P. vivax malaria elimination program needs early diagnosis, and prompt and complete radical treatment, which is challenging, to simultaneously exterminate the circulating parasites and dormant hypnozoites lodged in the hepatocytes of the host liver. As prompt surveillance and effective treatments are rolled out, preventing primaquine toxicity in the patients having glucose-6-phosphate dehydrogenase (G6PD) deficiency should be a priority for the vivax elimination program. This review sheds light on the burden of P. vivax, changing epidemiological patterns, the hurdles in elimination efforts, and the essential tools needed not just in India but globally. These tools encompass innovative treatments for eliminating dormant parasites, coping with evolving drug resistance, and the development of potential vaccines against the parasite.
Collapse
Affiliation(s)
- Ashwani Kumar
- ICMR - Vector Control Research Centre, Puducherry, India
| | | | - Suchi Tyagi
- ICMR - Vector Control Research Centre, Puducherry, India
| | | | | | - Manju Rahi
- ICMR - Vector Control Research Centre, Puducherry, India
- Indian Council of Medical Research, Hqrs New Delhi, India
| |
Collapse
|
11
|
Thriemer K, Degaga TS, Christian M, Alam MS, Rajasekhar M, Ley B, Hossain MS, Kibria MG, Tego TT, Abate DT, Weston S, Mnjala H, Rumaseb A, Satyagraha AW, Sadhewa A, Panggalo LV, Ekawati LL, Lee G, Anose RT, Kiros FG, Simpson JA, Karahalios A, Woyessa A, Baird JK, Sutanto I, Hailu A, Price RN. Primaquine radical cure in patients with Plasmodium falciparum malaria in areas co-endemic for P falciparum and Plasmodium vivax (PRIMA): a multicentre, open-label, superiority randomised controlled trial. Lancet 2023; 402:2101-2110. [PMID: 37979594 PMCID: PMC10714037 DOI: 10.1016/s0140-6736(23)01553-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 11/20/2023]
Abstract
BACKGROUND In areas co-endemic for Plasmodium vivax and Plasmodium falciparum there is an increased risk of P vivax parasitaemia following P falciparum malaria. Radical cure is currently only recommended for patients presenting with P vivax malaria. Expanding the indication for radical cure to patients presenting with P falciparum malaria could reduce their risk of subsequent P vivax parasitaemia. METHODS We did a multicentre, open-label, superiority randomised controlled trial in five health clinics in Bangladesh, Indonesia, and Ethiopia. In Bangladesh and Indonesia, patients were excluded if they were younger than 1 year, whereas in Ethiopia patients were excluded if they were younger than 18 years. Patients with uncomplicated P falciparum monoinfection who had fever or a history of fever in the 48 h preceding clinic visit were eligible for enrolment and were required to have a glucose-6-dehydrogenase (G6PD) activity of 70% or greater. Patients received blood schizontocidal treatment (artemether-lumefantrine in Ethiopia and Bangladesh and dihydroartemisinin-piperaquine in Indonesia) and were randomly assigned (1:1) to receive either high-dose short-course oral primaquine (intervention arm; total dose 7 mg/kg over 7 days) or standard care (standard care arm; single dose oral primaquine of 0·25 mg/kg). Random assignment was done by an independent statistician in blocks of eight by use of sealed envelopes. All randomly assigned and eligible patients were included in the primary and safety analyses. The per-protocol analysis excluded those who did not complete treatment or had substantial protocol violations. The primary endpoint was the incidence risk of P vivax parasitaemia on day 63. This trial is registered at ClinicalTrials.gov, NCT03916003. FINDINGS Between Aug 18, 2019, and March 14, 2022, a total of 500 patients were enrolled and randomly assigned, and 495 eligible patients were included in the intention-to-treat analysis (246 intervention and 249 control). The incidence risk of P vivax parasitaemia at day 63 was 11·0% (95% CI 7·5-15·9) in the standard care arm compared with 2·5% (1·0-5·9) in the intervention arm (hazard ratio 0·20, 95% CI 0·08-0·51; p=0·0009). The effect size differed with blood schizontocidal treatment and site. Routine symptom reporting on day 2 and day 7 were similar between groups. In the first 42 days, there were a total of four primaquine-related adverse events reported in the standard care arm and 26 in the intervention arm; 132 (92%) of all 143 adverse events were mild. There were two serious adverse events in the intervention arm, which were considered unrelated to the study drug. None of the patients developed severe anaemia (defined as haemoglobin <5 g/dL). INTERPRETATION In patients with a G6PD activity of 70% or greater, high-dose short-course primaquine was safe and relatively well tolerated and reduced the risk of subsequent P vivax parasitaemia within 63 days by five fold. Universal radical cure therefore potentially offers substantial clinical, public health, and operational benefits, but these benefits will vary with endemic setting. FUNDING Australian Academy of Science Regional Collaborations Program, Bill & Melinda Gates Foundation, and National Health and Medical Research Council.
Collapse
Affiliation(s)
- Kamala Thriemer
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia.
| | - Tamiru Shibiru Degaga
- College of Medicine and Health Sciences, Arba Minch University, Arba Minch, Ethiopia
| | - Michael Christian
- Oxford University Clinical Research Unit Indonesia, Jakarta, Indonesia
| | | | - Megha Rajasekhar
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Benedikt Ley
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | | | | | | | | | - Sophie Weston
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Hellen Mnjala
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Angela Rumaseb
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Ari Winasti Satyagraha
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Indonesia; Exeins Health Initiative, Jakarta, Indonesia
| | - Arkasha Sadhewa
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | | | - Lenny L Ekawati
- Oxford University Clinical Research Unit Indonesia, Jakarta, Indonesia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Grant Lee
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Rodas Temesgen Anose
- College of Medicine and Health Sciences, Arba Minch University, Arba Minch, Ethiopia
| | - Fitsum Getahun Kiros
- College of Medicine and Health Sciences, Arba Minch University, Arba Minch, Ethiopia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Amalia Karahalios
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Adugna Woyessa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - J Kevin Baird
- Oxford University Clinical Research Unit Indonesia, Jakarta, Indonesia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Inge Sutanto
- Department of Parasitology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Asrat Hailu
- College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Ric N Price
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
12
|
Dickey TH, Tolia NH. Designing an effective malaria vaccine targeting Plasmodium vivax Duffy-binding protein. Trends Parasitol 2023; 39:850-858. [PMID: 37481347 PMCID: PMC11099547 DOI: 10.1016/j.pt.2023.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/24/2023]
Abstract
Malaria caused by the Plasmodium vivax parasite is a major global health burden. Immunity against blood-stage infection reduces parasitemia and disease severity. Duffy-binding protein (DBP) is the primary parasite protein responsible for the invasion of red blood cells and it is a leading subunit vaccine candidate. An effective vaccine, however, is still lacking despite decades of interest in DBP as a vaccine candidate. This review discusses the reasons for targeting DBP, the challenges associated with developing a vaccine, and modern structural vaccinology methods that could be used to create an effective DBP vaccine. Next-generation DBP vaccines have the potential to elicit a broadly protective immune response and provide durable and potent protection from P. vivax malaria.
Collapse
Affiliation(s)
- Thayne H Dickey
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA
| | - Niraj H Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20894, USA.
| |
Collapse
|
13
|
Chu CS, Stolbrink M, Stolady D, Saito M, Beau C, Choun K, Wah TG, Mu N, Htoo K, Nu B, Keereevijit A, Wiladpaingern J, Carrara V, Phyo AP, Lwin KM, Luxemburger C, Proux S, Charunwatthana P, McGready R, White NJ, Nosten F. Severe Falciparum and Vivax Malaria on the Thailand-Myanmar Border: A Review of 1503 Cases. Clin Infect Dis 2023; 77:721-728. [PMID: 37144342 PMCID: PMC10495127 DOI: 10.1093/cid/ciad262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND The northwestern border of Thailand is an area of low seasonal malaria transmission. Until recent successful malaria elimination activities, malaria was a major cause of disease and death. Historically the incidences of symptomatic Plasmodium falciparum and Plasmodium vivax malaria were approximately similar. METHODS All malaria cases managed in the Shoklo Malaria Research Unit along the Thailand-Myanmar border between 2000 and 2016 were reviewed. RESULTS There were 80 841 consultations for symptomatic P. vivax and 94 467 for symptomatic P. falciparum malaria. Overall, 4844 (5.1%) patients with P. falciparum malaria were admitted to field hospitals, of whom 66 died, compared with 278 (0.34%) with P. vivax malaria, of whom 4 died (3 had diagnoses of sepsis, so the contribution of malaria to their fatal outcomes is uncertain). Applying the 2015 World Health Organization severe malaria criteria, 68 of 80 841 P. vivax admissions (0.08%) and 1482 of 94 467 P. falciparum admissions (1.6%) were classified as severe. Overall, patients with P. falciparum malaria were 15 (95% confidence interval, 13.2-16.8) times more likely than those with P. vivax malaria to require hospital admission, 19 (14.6-23.8) times more likely to develop severe malaria, and ≥14 (5.1-38.7) times more likely to die. CONCLUSIONS In this area, both P. falciparum and P. vivax infections were important causes of hospitalization, but life-threatening P. vivax illness was rare.
Collapse
Affiliation(s)
- Cindy S Chu
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Marie Stolbrink
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Daniel Stolady
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Makoto Saito
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Candy Beau
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Kan Choun
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Tha Gay Wah
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ne Mu
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Klay Htoo
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Be Nu
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Arunrot Keereevijit
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Jacher Wiladpaingern
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Verena Carrara
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Faculty of Medicine, Institute of Global Health, University of Geneva, Geneva, Switzerland
| | - Aung Pyae Phyo
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Khin Maung Lwin
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Christine Luxemburger
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Stephane Proux
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Prakaykaew Charunwatthana
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas J White
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol–Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
14
|
Taylor WRJ, Meagher N, Ley B, Thriemer K, Bancone G, Satyagraha A, Assefa A, Chand K, Chau NH, Dhorda M, Degaga TS, Ekawati LL, Hailu A, Hasanzai MA, Naddim MN, Pasaribu AP, Rahim AG, Sutanto I, Thanh NV, Tuyet-Trinh NT, Waithira N, Woyessa A, Dondorp A, von Seidlein L, Simpson JA, White NJ, Baird JK, Day NP, Price RN. Weekly primaquine for radical cure of patients with Plasmodium vivax malaria and glucose-6-phosphate dehydrogenase deficiency. PLoS Negl Trop Dis 2023; 17:e0011522. [PMID: 37672548 PMCID: PMC10482257 DOI: 10.1371/journal.pntd.0011522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 07/10/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND The World Health Organization recommends that primaquine should be given once weekly for 8-weeks to patients with Plasmodium vivax malaria and glucose-6-phosphate dehydrogenase (G6PD) deficiency, but data on its antirelapse efficacy and safety are limited. METHODS Within the context of a multicentre, randomised clinical trial of two primaquine regimens in P. vivax malaria, patients with G6PD deficiency were excluded and enrolled into a separate 12-month observational study. They were treated with a weekly dose of 0.75 mg/kg primaquine for 8 weeks (PQ8W) plus dihydroartemisinin piperaquine (Indonesia) or chloroquine (Afghanistan, Ethiopia, Vietnam). G6PD status was diagnosed using the fluorescent spot test and confirmed by genotyping for locally prevalent G6PD variants. The risk of P. vivax recurrence following PQ8W and the consequent haematological recovery were characterized in all patients and in patients with genotypically confirmed G6PD variants, and compared with the patients enrolled in the main randomised control trial. RESULTS Between July 2014 and November 2017, 42 male and 8 female patients were enrolled in Afghanistan (6), Ethiopia (5), Indonesia (19), and Vietnam (20). G6PD deficiency was confirmed by genotyping in 31 patients: Viangchan (14), Mediterranean (4), 357A-G (3), Canton (2), Kaiping (2), and one each for A-, Chatham, Gaohe, Ludhiana, Orissa, and Vanua Lava. Two patients had recurrent P. vivax parasitaemia (days 68 and 207). The overall 12-month cumulative risk of recurrent P. vivax malaria was 5.1% (95% CI: 1.3-18.9) and the incidence rate of recurrence was 46.8 per 1000 person-years (95% CI: 11.7-187.1). The risk of P. vivax recurrence was lower in G6PD deficient patients treated with PQ8W compared to G6PD normal patients in all treatment arms of the randomised controlled trial. Two of the 26 confirmed hemizygous males had a significant fall in haemoglobin (>5g/dl) after the first dose but were able to complete their 8 week regimen. CONCLUSIONS PQ8W was highly effective in preventing P. vivax recurrences. Whilst PQ8W was well tolerated in most patients across a range of different G6PD variants, significant falls in haemoglobin may occur after the first dose and require clinical monitoring. TRIAL REGISTRATION This trial is registered at ClinicalTrials.gov (NCT01814683).
Collapse
Affiliation(s)
- Walter R. J. Taylor
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Niamh Meagher
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Benedikt Ley
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | - Kamala Thriemer
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | - Germana Bancone
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Ari Satyagraha
- Eijkman Institute of Molecular Biology, Jakarta, Indonesia.8. Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | | | - Krisin Chand
- Oxford University Clinical Research Unit, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Nguyen Hoang Chau
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Mehul Dhorda
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tamiru S. Degaga
- College of Medicine & Health Sciences, Arbaminch University, Arbaminch, Ethiopia
| | - Lenny L. Ekawati
- Oxford University Clinical Research Unit, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Asrat Hailu
- College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | | | | | | | - Awab Ghulam Rahim
- Nangarhar Medical Faculty, Nangarhar University, Ministry of Higher Education, Jalalabad, Afghanistan
- Health and Social Development Organization, Kabul, Afghanistan
| | - Inge Sutanto
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ngo Viet Thanh
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Tuyet-Trinh
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Naomi Waithira
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Adugna Woyessa
- Ethiopian Public Health Institute, Addis Ababa, Ethiopia
| | - Arjen Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lorenz von Seidlein
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Julie A. Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Nicholas J. White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - J. Kevin Baird
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford University Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Nicholas P. Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ric N. Price
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| |
Collapse
|
15
|
Badugu V, Gaur BK, Maini B. Clinical profile and severity of Plasmodium vivax and falciparum malaria in hospitalized children from North India. J Vector Borne Dis 2023; 60:252-258. [PMID: 37843235 DOI: 10.4103/0972-9062.331406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND & OBJECTIVES Malaria is the most common parasitic infection in children and the most common cause of mortality by vector-borne disease in tropical countries. In these endemic countries there is limited published literature on the clinical profile and severity of Plasmodium vivax malaria in children. We highlight the clinical presentations and severity of malaria in children belonging to Uttar Pradesh, North India Methods: This observational study was conducted in a tertiary care hospital, in Moradabad, western Uttar Pradesh, India. Children (aged 6 months-18 years) hospitalized from June 2019 to May 2020 with a clinical picture consistent with malaria along with a positive rapid malaria antigen test (RMAT) and/or positive peripheral blood smear for malaria were enrolled. All data about the clinical profile and laboratory features were recorded. Results were analyzed for significance using appropriate statistical tests for continuous and categorical data. RESULTS One hundred children were enrolled in this study; 59 cases had Plasmodium vivax (PV) malaria, 33 cases had Plasmodium falciparum (PF) malaria, and 8 cases had mixed infections (both PV and PF). Among 59 children with P V malaria, 44 (74%) had severe malaria and 15 (26%) had uncomplicated malaria. Severe malarial anaemia (43.2%), followed by jaundice (36.4%), impaired renal function (21.6%), significant bleeding (18.9%), shock (18.9%), and prostration (16.2%), were the main severity parameters of malaria among these hospitalised children. Impaired renal function (P-value = 0.01) and unconsciousness (P-value = 0.02) were more frequent in Plasmodium falciparum cases. Other severity parameters were not significant between the vivax and falciparum species. INTERPRETATION & CONCLUSION We conclude that a significant proportion of severe malaria was caused by Plasmodium vivax in this region, where both species coexist. Plasmodium vivax malaria is no longer the benign entity it was around ten years ago in hospitalised children. Severe malarial anaemia was the most common severity parameter found in both Plasmodium vivax and P. falciparum species. The clinical presentation and a change in the severity parameters in vivax malaria indicate a recent shift in the disease severity from benign to fatal.
Collapse
Affiliation(s)
- Venkatesh Badugu
- Department of Pediatrics, Teerthanker Mahaveer Medical College and Research Center (TMMC&RC), Moradabad, Uttar Pradesh, India
| | - Bablu Kumar Gaur
- Department of Pediatrics, Teerthanker Mahaveer Medical College and Research Center (TMMC&RC), Moradabad, Uttar Pradesh, India
| | - Baljeet Maini
- Department of Pediatrics, Teerthanker Mahaveer Medical College and Research Center (TMMC&RC), Moradabad, Uttar Pradesh, India
| |
Collapse
|
16
|
Rai A, Pinto DS, Rai P, Teerthanath S, Karunasagar I, Adiga R. Misidentification of Plasmodium Species by Cross-Reacting Primers and Cerebral Malaria Caused by Plasmodium vivax. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2023. [DOI: 10.1055/s-0043-1761258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Abstract
Introduction The clinical presentation of a case as cerebral malaria with molecular identification confirming it as Plasmodium vivax underlines the importance of using molecular tools to identify the species and type of malaria. The possibility of the relationship between the complication observed during clinical diagnosis and the multifactorial molecular changes could likely be the reason for terming it cerebral malaria.
Methods We report four cases analyzed using the quantitative buffy coat technique followed by classical Giemsa stained thick-film microscopy, and nested polymerase chain reaction for the genus-specific region of Plasmodium targeting 18S rDNA followed by species-specific identification with a different set of primers and products confirmation with sequencing.
Results Primers targeting P. knowlesi generated the expected product size of 153 base pairs that, upon sequencing, matched with the P. vivax sequence reflecting the relatedness of the species. Likewise, primers targeting P. ovale generated a 456 product whose sequence matched the P. vivax sequence.
Conclusion Infection with P. vivax can potentially cause cerebral malaria, and P. vivax can cause severe malaria complications alone or mixed with other species and can show cerebral malaria signs, which are typically associated with P. falciparum infections. The sequence relatedness reflects the genome similarity between P. knowlesi and P. ovale with P. vivax. The need to reconfirm with an additional set of newly reported primers is mandatory.
Collapse
Affiliation(s)
- Anoopkrishna Rai
- Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, Karnataka, India
| | - Deepak Sebastian Pinto
- Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, Karnataka, India
| | - Praveen Rai
- Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, Karnataka, India
| | - Srinivas Teerthanath
- Department of Pathology, K S Hegde Medical Academy, Deralakatte, Mangaluru, Karnataka, India
| | - Indrani Karunasagar
- Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, Karnataka, India
| | - Rama Adiga
- Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, Karnataka, India
| |
Collapse
|
17
|
Drysdale M, Tan L, Martin A, Fuhrer IB, Duparc S, Sharma H. Plasmodium vivax in Children: Hidden Burden and Conspicuous Challenges, a Narrative Review. Infect Dis Ther 2023; 12:33-51. [PMID: 36378465 PMCID: PMC9868225 DOI: 10.1007/s40121-022-00713-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
There has been progress towards decreasing malaria prevalence globally; however, Plasmodium vivax has been less responsive to elimination efforts compared with Plasmodium falciparum. P. vivax malaria remains a serious public health concern in regions where it is the dominant species (South and South-East Asia, the Eastern Mediterranean region, and South America) and is increasingly recognized for its contribution to overall morbidity and mortality worldwide. The incidence of P. vivax decreases with increasing age owing to rapidly acquired clinical immunity and there is a disproportionate burden of P. vivax in infants and children, who remain highly vulnerable to severe disease, recurrence, and anemia with associated developmental impacts. Diagnosis is sometimes difficult owing to the sensitivity of diagnostic tests to detect low levels of parasitemia. Additionally, the propensity of P. vivax to relapse following reactivation of dormant hypnozoites in the liver contributes to disease recurrence in infants and children, and potentiates morbidity and transmission. The 8-aminoquinolines, primaquine and tafenoquine, provide radical cure (relapse prevention). However, the risk of hemolysis in patients with glucose-6-phosphate dehydrogenase (G6PD) deficiency necessitates testing prior to administration of 8-aminoquinolines, which has limited their uptake. Additional challenges include lack of availability of pediatric dose formulations and problems with adherence to primaquine owing to the length of treatment recommended. A paucity of data and studies specific to pediatric P. vivax malaria impacts the ability to deliver targeted interventions. It is imperative that P. vivax in infants and children be the focus of future research, control initiatives, and anti-malarial drug development.
Collapse
Affiliation(s)
| | - Lionel Tan
- GSK, 980 Great West Road, Brentford, TW8 9GS Middlesex UK
| | - Ana Martin
- GSK, 980 Great West Road, Brentford, TW8 9GS Middlesex UK
| | | | | | - Hema Sharma
- GSK, 980 Great West Road, Brentford, TW8 9GS Middlesex UK
| |
Collapse
|
18
|
Olvany JM, Williams SM, Zimmerman PA. Global perspectives on CYP2D6 associations with primaquine metabolism and Plasmodium vivax radical cure. Front Pharmacol 2022; 13:752314. [PMID: 36457706 PMCID: PMC9705595 DOI: 10.3389/fphar.2022.752314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Clinical trial and individual patient treatment outcomes have produced accumulating evidence that effective primaquine (PQ) treatment of Plasmodium vivax and P. ovale liver stage hypnozoites is associated with genetic variation in the human cytochrome P450 gene, CYP2D6. Successful PQ treatment of individual and population-wide infections by the Plasmodium species that generate these dormant liver stage forms is likely to be necessary to reach elimination of malaria caused by these parasites globally. Optimizing safe and effective PQ treatment will require coordination of efforts between the malaria and pharmacogenomics research communities.
Collapse
Affiliation(s)
- Jasmine M. Olvany
- The Center for Global Health and Diseases, Pathology Department, Case Western Reserve University, Cleveland, OH, United States
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Scott M. Williams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Peter A. Zimmerman
- The Center for Global Health and Diseases, Pathology Department, Case Western Reserve University, Cleveland, OH, United States
- Master of Public Health Program, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
19
|
Holzschuh A, Gruenberg M, Hofmann NE, Wampfler R, Kiniboro B, Robinson LJ, Mueller I, Felger I, White MT. Co-infection of the four major Plasmodium species: Effects on densities and gametocyte carriage. PLoS Negl Trop Dis 2022; 16:e0010760. [PMID: 36099312 PMCID: PMC9506632 DOI: 10.1371/journal.pntd.0010760] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/23/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
Background Co-infection of the four major species of human malaria parasite Plasmodium falciparum (Pf), P. vivax (Pv), P. malariae (Pm), and P. ovale sp. (Po) is regularly observed, but there is limited understanding of between-species interactions. In particular, little is known about the effects of multiple Plasmodium species co-infections on gametocyte production. Methods We developed molecular assays for detecting asexual and gametocyte stages of Pf, Pv, Pm, and Po. This is the first description of molecular diagnostics for Pm and Po gametocytes. These assays were implemented in a unique epidemiological setting in Papua New Guinea with sympatric transmission of all four Plasmodium species permitting a comprehensive investigation of species interactions. Findings The observed frequency of Pf-Pv co-infection for asexual parasites (14.7%) was higher than expected from individual prevalence rates (23.8%Pf x 47.4%Pv = 11.3%). The observed frequency of co-infection with Pf and Pv gametocytes (4.6%) was higher than expected from individual prevalence rates (13.1%Pf x 28.2%Pv = 3.7%). The excess risk of co-infection was 1.38 (95% confidence interval (CI): 1.09, 1.67) for all parasites and 1.37 (95% CI: 0.95, 1.79) for gametocytes. This excess co-infection risk was partially attributable to malaria infections clustering in some villages. Pf-Pv-Pm triple infections were four times more frequent than expected by chance alone, which could not be fully explained by infections clustering in highly exposed individuals. The effect of co-infection on parasite density was analyzed by systematic comparison of all pairwise interactions. This revealed a significant 6.57-fold increase of Pm density when co-infected with Pf. Pm gametocytemia also increased with Pf co-infection. Conclusions Heterogeneity in exposure to mosquitoes is a key epidemiological driver of Plasmodium co-infection. Among the four co-circulating parasites, Pm benefitted most from co-infection with other species. Beyond this, no general prevailing pattern of suppression or facilitation was identified in pairwise analysis of gametocytemia and parasitemia of the four species. Trial registration This trial is registered with ClinicalTrials.gov, Trial ID: NCT02143934. The majority of malaria research focuses on the Plasmodium falciparum and P. vivax parasite species, due to their large public health burden. The epidemiology of P. malariae and P. ovale parasites has been comparatively neglected, due to a lack of research tools, most notably diagnostics. We present new molecular diagnostic assays for detecting P. malariae and P. ovale gametocytes, the sexual stage of the malaria parasite transmitted to mosquitoes. These assays were applied to samples collected in Papua New Guinea, a rare region with high transmission of the four major malaria parasite species. Patterns of co-infections were characterized accounting for interactions between pairs and triples of parasites. Heterogeneity in exposure to mosquito bites was identified as a key driver of patterns of co-infection. The effect of co-infection on parasite density was analyzed by systematic comparison of all pairwise interactions. The most significant within-host interaction of parasites was the large increase in P. malariae parasite density due to co-infection with P. falciparum. This finding was replicated for P. malariae gametocytes (but did not attain statistical significance due to low sample numbers) suggesting that co-infection provides a key transmission advantage to P. malariae.
Collapse
Affiliation(s)
- Aurel Holzschuh
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Maria Gruenberg
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Natalie E. Hofmann
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Rahel Wampfler
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Benson Kiniboro
- Vector Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang and Maprik, Papua New Guinea
| | - Leanne J. Robinson
- Vector Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang and Maprik, Papua New Guinea
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
| | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
| | - Ingrid Felger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- * E-mail: (IF); (MTW)
| | - Michael T. White
- Institut Pasteur, Université de Paris Cité, G5 Épidémiologie et Analyse des Maladies Infectieuses, Département de Santé Globale, Paris, France
- * E-mail: (IF); (MTW)
| |
Collapse
|
20
|
Anwar MN, Hickson RI, Mehra S, McCaw JM, Flegg JA. A Multiscale Mathematical Model of Plasmodium Vivax Transmission. Bull Math Biol 2022; 84:81. [PMID: 35778540 PMCID: PMC9249727 DOI: 10.1007/s11538-022-01036-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/26/2022] [Indexed: 11/29/2022]
Abstract
Malaria is caused by Plasmodium parasites which are transmitted to humans by the bite of an infected Anopheles mosquito. Plasmodium vivax is distinct from other malaria species in its ability to remain dormant in the liver (as hypnozoites) and activate later to cause further infections (referred to as relapses). Mathematical models to describe the transmission dynamics of P. vivax have been developed, but most of them fail to capture realistic dynamics of hypnozoites. Models that do capture the complexity tend to involve many governing equations, making them difficult to extend to incorporate other important factors for P. vivax, such as treatment status, age and pregnancy. In this paper, we have developed a multiscale model (a system of integro-differential equations) that involves a minimal set of equations at the population scale, with an embedded within-host model that can capture the dynamics of the hypnozoite reservoir. In this way, we can gain key insights into dynamics of P. vivax transmission with a minimum number of equations at the population scale, making this framework readily scalable to incorporate more complexity. We performed a sensitivity analysis of our multiscale model over key parameters and found that prevalence of P. vivax blood-stage infection increases with both bite rate and number of mosquitoes but decreases with hypnozoite death rate. Since our mathematical model captures the complex dynamics of P. vivax and the hypnozoite reservoir, it has the potential to become a key tool to inform elimination strategies for P. vivax.
Collapse
Affiliation(s)
- Md Nurul Anwar
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia.,Department of Mathematics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Roslyn I Hickson
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia.,Australian Institute of Tropical Health and Medicine, and College of Public Health, Medical & Veterinary Sciences, James Cook University, Townsville, Australia.,Health and Biosecurity, CSIRO, Townsville, Australia
| | - Somya Mehra
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
| | - James M McCaw
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia.,Peter Doherty Institute for Infection and Immunity, The Royal Melbourne Hospital and The University of Melbourne, Parkville, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
21
|
Lima BAS, Fernandes GM, Torres LM, Pires CV, Alves JRS, Moreira-Nascimento SL, Nascimento MFA, Afonso SL, Costa HL, Cerávolo IP, Sousa TN, Soares IS, Ntumngia FB, Adams JH, Carvalho LH, Kano FS. Antibody response to a new member of the DBL family (EBP2) after a brief Plasmodium vivax exposure. PLoS Negl Trop Dis 2022; 16:e0010493. [PMID: 35714097 PMCID: PMC9205486 DOI: 10.1371/journal.pntd.0010493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/12/2022] [Indexed: 12/03/2022] Open
Abstract
Plasmodium vivax blood-stage invasion into reticulocyte is critical for parasite development. Thus, validation of novel parasite invasion ligands is essential for malaria vaccine development. Recently, we demonstrated that EBP2, a Duffy binding protein (DBP) paralog, is antigenically distinct from DBP and could not be functionally inhibited by anti-DBP antibodies. Here, we took advantage of a small outbreak of P.vivax malaria, located in a non-malarious area of Brazil, to investigate for the first time IgM/IgG antibodies against EBP2 and DEKnull-2 (an engineering DBPII vaccine) among individuals who had their first and brief exposure to P.vivax (16 cases and 22 non-cases). Our experimental approach included 4 cross sectional surveys at 3-month interval (12-month follow-up). The results demonstrated that while a brief initial P.vivax infection was not efficient to induce IgM/ IgG antibodies to either EBP2 or DEKnull-2, IgG antibodies against DEKnull-2 (but not EBP2) were boosted by recurrent blood-stage infections following treatment. Of interest, in most recurrent P. vivax infections (4 out of 6 patients) DEKnull-2 IgG antibodies were sustained for 6 to 12 months. Polymorphisms in the ebp2 gene does not seem to explain EBP2 low immunogenicity as the ebp2 allele associated with the P.vivax outbreak presented high identity to the original EBP2 isolate used as recombinant protein. Although EBP2 antibodies were barely detectable after a primary episode of P.vivax infection, EBP2 was highly recognized by serum IgG from long-term malaria-exposed Amazonians (range from 35 to 92% according to previous malaria episodes). Taken together, the results showed that individuals with a single and brief exposure to P.vivax infection develop very low anti-EBP2 antibodies, which tend to increase after long-term malaria exposure. Finally, the findings highlighted the potential of DEKnull-2 as a vaccine candidate, as in non-immune individuals anti-DEKnull-2 IgG antibodies were boosted even after a brief exposure to P.vivax blood stages.
Collapse
Affiliation(s)
- Bárbara A. S. Lima
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela M. Fernandes
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Letícia M. Torres
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Camilla V. Pires
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Jéssica R. S. Alves
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Sâmick L. Moreira-Nascimento
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Fernanda A. Nascimento
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Sofia L. Afonso
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Helena L. Costa
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Isabela P. Cerávolo
- Laboratório de Imunopatologia, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Tais N. Sousa
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Irene S. Soares
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Francis B. Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Luzia H. Carvalho
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Flora S. Kano
- Biologia Molecular e Imunologia da Malária, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
22
|
Kucharska I, Hossain L, Ivanochko D, Yang Q, Rubinstein JL, Pomès R, Julien JP. Structural basis of Plasmodium vivax inhibition by antibodies binding to the circumsporozoite protein repeats. eLife 2022; 11:e72908. [PMID: 35023832 PMCID: PMC8809896 DOI: 10.7554/elife.72908] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022] Open
Abstract
Malaria is a global health burden, with Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) responsible for the majority of infections worldwide. Circumsporozoite protein (CSP) is the most abundant protein on the surface of Plasmodium sporozoites, and antibodies targeting the central repeat region of CSP can prevent parasite infection. Although much has been uncovered about the molecular basis of antibody recognition of the PfCSP repeats, data remains scarce for PvCSP. Here, we performed molecular dynamics simulations for peptides comprising the PvCSP repeats from strains VK210 and VK247 to reveal how the PvCSP central repeats are highly disordered, with minor propensities to adopt turn conformations. Next, we solved eight crystal structures to unveil the interactions of two inhibitory monoclonal antibodies (mAbs), 2F2 and 2E10.E9, with PvCSP repeats. Both antibodies can accommodate subtle sequence variances in the repeat motifs and recognize largely coiled peptide conformations that also contain isolated turns. Our structural studies uncover various degrees of Fab-Fab homotypic interactions upon recognition of the PvCSP central repeats by these two inhibitory mAbs, similar to potent mAbs against PfCSP. These findings augment our understanding of host-Plasmodium interactions and contribute molecular details of Pv inhibition by mAbs to unlock structure-based engineering of PvCSP-based vaccines.
Collapse
Affiliation(s)
- Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Lamia Hossain
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
| | - Qiren Yang
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
- Department of Medical Biophysics, University of TorontoTorontoCanada
| | - Régis Pomès
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research InstituteTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
- Department of Immunology, University of TorontoTorontoCanada
| |
Collapse
|
23
|
Meier L, Casagrande G, Abdulla S, Masanja H. A brief history of selected malaria vaccine and medical interventions pursued by the Swiss Tropical and Public Health Institute and partners, 1943-2021. Acta Trop 2022; 225:106115. [PMID: 34464588 DOI: 10.1016/j.actatropica.2021.106115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 11/01/2022]
Abstract
In order to be successful in global health today, all the long-established European tropical research institutes had to undergo a transition which can be described as "hunter-gatherer" and descriptive approaches during colonial and postcolonial times to a deeper understanding of infection biology and finally to public health interventions from which populations at large can benefit. During the 1980s and 1990s, the Swiss Tropical Institute (today: Swiss Tropical and Public Health Institute, Swiss TPH) based in Basel too has changed its focus from individual medicine to a public health context. This article does not present new scientific data but takes a historical perspective. Its aim is to highlight the above-mentioned transformation by focusing on selected malaria research-cum-action interventions during the crucial period of the 1990s, which were tailored to the social-ecological settings where the disease was endemic. In order for this transformation to be successful, we intend to emphasise the importance of (i) having a fundamental understanding of local transmission; (ii) building and nurturing relationships with partner institutions; and (iii) developing a coherent research portfolio as key elements for researching and applying evidence in malaria control and elimination as part of national malaria control programmes.
Collapse
|
24
|
Clark IA. How diseases caused by parasites allowed a wider understanding of disease in general: my encounters with parasitology in Australia and elsewhere over the last 50 years. Int J Parasitol 2021; 51:1265-1276. [PMID: 34757090 DOI: 10.1016/j.ijpara.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/18/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
This is an account of how it can prove possible to carve a reasonable scientific career by following what brought most scientific thrill rather than pursue a safe, institution-directed, path. The fascination began when I noticed, quite unexpectedly, that the normal mouse immune response causes Babesia microti to die, en masse, inside circulating red cells. It eventuated that prior Bacillus Calmette Guerin infection caused the same outcome, even before the protozoal infection became patent. It also rendered mice quite immune, long term. I acquired an obsession about this telling us how little we know. Surrounded by basic immunologists, parasitologists and virologists in London, I had been given, in the days that funding was ample, the opportunity to follow any promising lead with a free hand. Through Bacillus Calmette Guerin, this meant stumbling through a set of phenomena that were in their infancies, and could be explained only through nebulous novel soluble mediators such as TNF, described the following year as causing the in vivo necrosis of tumours in mice. Beginning with malarial disease pathogenesis, I followed TNF wherever it led, into innate immunity, acute and chronic infections, neurophysiology and neurodegenerative diseases, in all of which states awareness of the role of this cytokine is still growing fast. Many of these steps can be illustrated and expanded upon in parasitic diseases. Covering the importance of TNF in the pathogenesis of neurodegenerative disease has proved to be highly illuminating, scientifically and otherwise. But the insights it has given me into understanding the temptations to which patent-owners can succumb when faced with opportunities to put money before people is not for the faint hearted. Clearly, parasitologists inhabit a much more common-good yet science-orientated, civilised, world.
Collapse
Affiliation(s)
- Ian A Clark
- Biomedical Sciences and Biochemistry, Research School of Biology, Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
25
|
Al-Subaie AM. Case Report: Case report: Mixed infection of Plasmodium vivax and Plasmodium falciparum in a tertiary hospital. F1000Res 2021; 10:779. [PMID: 34621515 PMCID: PMC8456375 DOI: 10.12688/f1000research.53162.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 11/25/2022] Open
Abstract
Mixed infections with two or more species of
Plasmodium are frequently reported due to vector factors, parasite factors (formation of hypnozoites) and host factors (residing in endemic areas, travel to endemic areas, inadequately treated previous infection, lack of compliance to therapy). Here we report a case of a 33-year-old Saudi female who had a significant travel history, and a peripheral blood smear (PBS) revealed mixed infection with
P. falciparum and
P. vivax. The case was successfully treated with a combination therapy of artemisinin and primaquine with follow up testing at three, seven, 14, and 28 days. Mixed malaria infections are especially reported in travelers to endemic areas. Hence, adequate diagnosis and appropriate treatment of the cases contributes majorly to preventing relapse and controlling the disease. Travel consultations should be given to all travelers before their trips to endemic countries.
Collapse
Affiliation(s)
- Abeer M Al-Subaie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam AbdulRahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
26
|
Forero-Peña DA, Carrión-Nessi FS, Chavero M, Gamardo Á, Figuera L, Camejo-Ávila NA, Marcano MV, Hidalgo M, Arenas-Leal CJ, Villegas L, Grillet ME, Pacheco MA, Mora MSD, Escalante AA. The clinical-epidemiological profile of malaria patients from Southern Venezuela, a critical hotspot in Latin America. Malar J 2021; 20:375. [PMID: 34544438 PMCID: PMC8453994 DOI: 10.1186/s12936-021-03913-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/09/2021] [Indexed: 11/10/2022] Open
Abstract
Background Venezuela accounted for 55% of the cases and 73% of the malaria deaths in the Americas in 2019. Bolivar state, in the southeast, contributes > 60% of the country's Plasmodium vivax and Plasmodium falciparum cases every year. This study describes the clinical–epidemiological characteristics of clinical malaria patients in this high-transmission area. Methods A prospective study was conducted on patients seeking medical attention in three medical centres in the state capital, Ciudad Bolivar, between June and October 2018. Malaria diagnosis was carried out using microscopy following national standards. Malaria-positive patients were examined for clinical symptoms, and haematological tests were performed at the time of diagnosis. Patients were followed up by telephone to evaluate malaria recurrences. Results Out of 287 patients, 200 (69.7%) were positive for P. vivax, 69 (24%) for P. falciparum, and 18 (6.3%) had mixed (P. vivax/P. falciparum) infections. Patients' median age was 33 years (IQR 20), 168 (69%) were men, and 40% practiced gold mining as the main occupation. Fever (96.5%), chills (91.3%), and headaches (90.6%) were the most frequent symptoms. At least one symptom associated with severe malaria was observed in 69 out of 161 patients with complete clinical evaluation (42.9%). Plasmodium vivax infections were found in 42 out of 69 (60.9%) severe cases; by contrast, P. falciparum and mixed malaria caused 34.8% (24/69) and 4.4% (3/69) of infections, respectively. Two patients died of cerebral malaria. Mean hemoglobin was lower in the patients infected with P. falciparum than those infected with P. vivax. Regardless of the parasite causing the infection, patients presented high levels of total bilirubin, aminotransferases (AST, ALT), and lactate dehydrogenase (LDH). Out of the 142 patients followed up by phone for three months (49.5% of the 287 patients), 35 (24.7%) reported recurrences. Conclusions The high malaria prevalence among young male adults practicing gold mining suggests that this occupation is a significant risk factor. The unexpected high prevalence of P. vivax patients with at least one criteria of severe clinical disease is a matter of concern. Whether it is the result of a lack of timely diagnosis and effective treatment should be explored.
Collapse
Affiliation(s)
- David A Forero-Peña
- Biomedical Research and Therapeutic Vaccines Institute, Ciudad Bolivar, Venezuela. .,Department of Internal Medicine, "Ruiz Y Páez" University Hospital Complex, Ciudad Bolivar, Venezuela.
| | - Fhabián S Carrión-Nessi
- Biomedical Research and Therapeutic Vaccines Institute, Ciudad Bolivar, Venezuela.,"Dr. Francisco Battistini Casalta" Health Sciences School, University of Oriente - Bolivar Nucleus, Ciudad Bolivar, Venezuela
| | - Melynar Chavero
- Biomedical Research and Therapeutic Vaccines Institute, Ciudad Bolivar, Venezuela.,Department of Internal Medicine, "Ruiz Y Páez" University Hospital Complex, Ciudad Bolivar, Venezuela
| | - Ángel Gamardo
- Biomedical Research and Therapeutic Vaccines Institute, Ciudad Bolivar, Venezuela
| | - Luisamy Figuera
- Biomedical Research and Therapeutic Vaccines Institute, Ciudad Bolivar, Venezuela
| | | | - María V Marcano
- Biomedical Research and Therapeutic Vaccines Institute, Ciudad Bolivar, Venezuela
| | - Mariana Hidalgo
- Venezuelan Institute of Scientific Research (IVIC), Miranda, Venezuela
| | | | - Leopoldo Villegas
- Civil Association of Social Impact (ASOCIS), Tumeremo, Venezuela.,Global Development One (GDO), Maryland, USA
| | - María E Grillet
- Vector and Parasite Biology Laboratory, Tropical Ecology and Zoology Institute, Faculty of Sciences, Central University of Venezuela, Caracas, Venezuela
| | - M Andreína Pacheco
- Biology Department/Institute of Genomics and Evolutionary Medicine (iGEM), Temple University, Philadelphia, PA, 19122-1801, USA
| | - Marisol Sandoval-de Mora
- Department of Internal Medicine, "Ruiz Y Páez" University Hospital Complex, Ciudad Bolivar, Venezuela
| | - Ananías A Escalante
- Biology Department/Institute of Genomics and Evolutionary Medicine (iGEM), Temple University, Philadelphia, PA, 19122-1801, USA.
| |
Collapse
|
27
|
Pincelli A, Cardoso MA, Malta MB, Johansen IC, Corder RM, Nicolete VC, Soares IS, Castro MC, Ferreira MU. Low-level Plasmodium vivax exposure, maternal antibodies, and anemia in early childhood: Population-based birth cohort study in Amazonian Brazil. PLoS Negl Trop Dis 2021; 15:e0009568. [PMID: 34264946 PMCID: PMC8282015 DOI: 10.1371/journal.pntd.0009568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/16/2021] [Indexed: 11/26/2022] Open
Abstract
Background Malaria causes significant morbidity and mortality in children under 5 years of age in sub-Saharan Africa and the Asia-Pacific region. Neonates and young infants remain relatively protected from clinical disease and the transplacental transfer of maternal antibodies is hypothesized as one of the protective factors. The adverse health effects of Plasmodium vivax malaria in early childhood–traditionally viewed as a benign infection–remain largely neglected in relatively low-endemicity settings across the Amazon. Methodology/Principal findings Overall, 1,539 children participating in a birth cohort study in the main transmission hotspot of Amazonian Brazil had a questionnaire administered, and blood sampled at the two-year follow-up visit. Only 7.1% of them experienced malaria confirmed by microscopy during their first 2 years of life– 89.1% of the infections were caused by P. vivax. Young infants appear to be little exposed to, or largely protected from infection, but children >12 months of age become as vulnerable to vivax malaria as their mothers. Few (1.4%) children experienced ≥4 infections during the 2-year follow-up, accounting for 43.4% of the overall malaria burden among study participants. Antenatal malaria diagnosed by microscopy during pregnancy or by PCR at delivery emerged as a significant correlate of subsequent risk of P. vivax infection in the offspring (incidence rate ratio, 2.58; P = 0.002), after adjusting for local transmission intensity. Anti-P. vivax antibodies measured at delivery do not protect mothers from subsequent malaria; whether maternal antibodies transferred to the fetus reduce early malaria risk in children remains undetermined. Finally, recent and repeated vivax malaria episodes in early childhood are associated with increased risk of anemia at the age of 2 years in this relatively low-endemicity setting. Conclusions/Significance Antenatal infection increases the risk of vivax malaria in the offspring and repeated childhood P. vivax infections are associated with anemia at the age of 2 years. Plasmodium vivax malaria causes frequent hospital admissions of infants and toddlers in areas of intense transmission in the Asia-Pacific region, often due to severe anemia, but its epidemiology and burden have been understudied in children from other endemic settings. Here we characterize the cumulative impact of P. vivax infections in infants and toddlers exposed to relatively low levels of malaria transmission in the Brazilian Amazon. We have previously shown that vivax malaria in pregnancy is associated with increased risk of maternal anemia and impaired fetal growth in this population. Now we show that the adverse effects of malaria extend to early childhood. Children born to mothers who had one or more infections during pregnancy are at an elevated risk of P. vivax malaria in their early life, although the transfer of maternal antibodies to the fetus may provide some short-term protection. Children who are repeatedly infected with P. vivax since birth are more likely to be anemic at the age of 2 years. These findings further challenge the traditional view of vivax malaria as a relatively benign infection in pregnancy and early childhood in the Amazon.
Collapse
Affiliation(s)
- Anaclara Pincelli
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marly A. Cardoso
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Maíra B. Malta
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
- Graduate Program in Collective Health, Catholic University of Santos, Santos, Brazil
| | - Igor C. Johansen
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rodrigo M. Corder
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vanessa C. Nicolete
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S. Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcia C. Castro
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Marcelo U. Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- * E-mail:
| | | |
Collapse
|
28
|
Karoli R, Shakya S, Gupta N, Mittal V, Upadhyay AK. Clinical profile of malaria at a tertiary care teaching hospital in North India. Trop Parasitol 2021; 11:25-30. [PMID: 34195057 PMCID: PMC8213115 DOI: 10.4103/tp.tp_76_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/13/2020] [Accepted: 11/24/2020] [Indexed: 11/04/2022] Open
Abstract
Introduction: Despite recent reductions in the overall malaria case incidence, malaria remains an important public health issue. There has been a change in clinical and epidemiological profile of malaria in our country and vivax malaria known to be benign has been reported to cause severe complications. Therefore, the present study was conducted to delineate the clinical profile of malaria, proportion of severity, spectrum of complications, and presence of comorbidities among adult patients admitted at a tertiary health-care center in North India. Materials and Methods: This was an observational prospective study conducted in all adult patients (>15 years of age) diagnosed to have Plasmodium vivax malaria, Plasmodium falciparum, and mixed malarial infection at a tertiary care teaching hospital on the basis of peripheral smear or rapid diagnostic tests. Results: The study included 295 patients, the most common species was vivax (62%) followed by falciparum (29%) and mixed plasmodium spp. (9%). The mean age of the patients was 34.23 ± 15.7 years, with 64% male and 36% female. Out of all patients, 23% patients had at least one component of severe malaria. Severe anemia (hemoglobin <5 mg/dl), thrombocytopenia (platelet count <1 lac/cmm), and acute kidney injury were significantly greater in patients with P. vivax. Presence of comorbid conditions was observed in a significant proportion (32%) of patients. Conclusion: P. vivax is the plasmodium species which is responsible for most of the cases. Its potential to cause life-threatening illness is the cause of concern. The role of comorbid conditions in influencing the clinicaloutcome of malaria should be further explored.
Collapse
Affiliation(s)
- Ritu Karoli
- Department of Medicine, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Shobhit Shakya
- Department of Medicine, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Nikhil Gupta
- Department of Medicine, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Vineeta Mittal
- Department of Microbiology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Anil Kumar Upadhyay
- Department of Medicine, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
29
|
Kho S, Qotrunnada L, Leonardo L, Andries B, Wardani PAI, Fricot A, Henry B, Hardy D, Margyaningsih NI, Apriyanti D, Puspitasari AM, Prayoga P, Trianty L, Kenangalem E, Chretien F, Brousse V, Safeukui I, del Portillo HA, Fernandez-Becerra C, Meibalan E, Marti M, Price RN, Woodberry T, Ndour PA, Russell BM, Yeo TW, Minigo G, Noviyanti R, Poespoprodjo JR, Siregar NC, Buffet PA, Anstey NM. Evaluation of splenic accumulation and colocalization of immature reticulocytes and Plasmodium vivax in asymptomatic malaria: A prospective human splenectomy study. PLoS Med 2021; 18:e1003632. [PMID: 34038413 PMCID: PMC8154101 DOI: 10.1371/journal.pmed.1003632] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 04/19/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND A very large biomass of intact asexual-stage malaria parasites accumulates in the spleen of asymptomatic human individuals infected with Plasmodium vivax. The mechanisms underlying this intense tropism are not clear. We hypothesised that immature reticulocytes, in which P. vivax develops, may display high densities in the spleen, thereby providing a niche for parasite survival. METHODS AND FINDINGS We examined spleen tissue in 22 mostly untreated individuals naturally exposed to P. vivax and Plasmodium falciparum undergoing splenectomy for any clinical indication in malaria-endemic Papua, Indonesia (2015 to 2017). Infection, parasite and immature reticulocyte density, and splenic distribution were analysed by optical microscopy, flow cytometry, and molecular assays. Nine non-endemic control spleens from individuals undergoing spleno-pancreatectomy in France (2017 to 2020) were also examined for reticulocyte densities. There were no exclusion criteria or sample size considerations in both patient cohorts for this demanding approach. In Indonesia, 95.5% (21/22) of splenectomy patients had asymptomatic splenic Plasmodium infection (7 P. vivax, 13 P. falciparum, and 1 mixed infection). Significant splenic accumulation of immature CD71 intermediate- and high-expressing reticulocytes was seen, with concentrations 11 times greater than in peripheral blood. Accordingly, in France, reticulocyte concentrations in the splenic effluent were higher than in peripheral blood. Greater rigidity of reticulocytes in splenic than in peripheral blood, and their higher densities in splenic cords both suggest a mechanical retention process. Asexual-stage P. vivax-infected erythrocytes of all developmental stages accumulated in the spleen, with non-phagocytosed parasite densities 3,590 times (IQR: 2,600 to 4,130) higher than in circulating blood, and median total splenic parasite loads 81 (IQR: 14 to 205) times greater, accounting for 98.7% (IQR: 95.1% to 98.9%) of the estimated total-body P. vivax biomass. More reticulocytes were in contact with sinus lumen endothelial cells in P. vivax- than in P. falciparum-infected spleens. Histological analyses revealed 96% of P. vivax rings/trophozoites and 46% of schizonts colocalised with 92% of immature reticulocytes in the cords and sinus lumens of the red pulp. Larger splenic cohort studies and similar investigations in untreated symptomatic malaria are warranted. CONCLUSIONS Immature CD71+ reticulocytes and splenic P. vivax-infected erythrocytes of all asexual stages accumulate in the same splenic compartments, suggesting the existence of a cryptic endosplenic lifecycle in chronic P. vivax infection. Findings provide insight into P. vivax-specific adaptions that have evolved to maximise survival and replication in the spleen.
Collapse
Affiliation(s)
- Steven Kho
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| | | | - Leo Leonardo
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Benediktus Andries
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | | | - Aurelie Fricot
- UMR_S1134, BIGR, Inserm, Université de F-75015 Paris, and Laboratory of Excellence GR-Ex, Paris, France
| | - Benoit Henry
- UMR_S1134, BIGR, Inserm, Université de F-75015 Paris, and Laboratory of Excellence GR-Ex, Paris, France
| | - David Hardy
- Institut Pasteur, Experimental Neuropathology Unit, Paris, France
| | | | - Dwi Apriyanti
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | - Pak Prayoga
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Leily Trianty
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Enny Kenangalem
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Rumah Sakit Umum Daerah Kabupaten Mimika, Timika, Papua, Indonesia
| | - Fabrice Chretien
- Institut Pasteur, Experimental Neuropathology Unit, Paris, France
| | - Valentine Brousse
- UMR_S1134, BIGR, Inserm, Université de F-75015 Paris, and Laboratory of Excellence GR-Ex, Paris, France
| | - Innocent Safeukui
- Department of Biological Sciences, Notre Dame University, Notre Dame, Indiana, United States of America
| | - Hernando A. del Portillo
- ISGlobal, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute, Badalona, Spain
- Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Carmen Fernandez-Becerra
- ISGlobal, Hospital Clinic-Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute, Badalona, Spain
| | - Elamaran Meibalan
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Matthias Marti
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Wellcome Center for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
| | - Ric N. Price
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
- Center for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tonia Woodberry
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| | - Papa A. Ndour
- UMR_S1134, BIGR, Inserm, Université de F-75015 Paris, and Laboratory of Excellence GR-Ex, Paris, France
| | - Bruce M. Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Tsin W. Yeo
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| | - Gabriela Minigo
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| | | | - Jeanne R. Poespoprodjo
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Rumah Sakit Umum Daerah Kabupaten Mimika, Timika, Papua, Indonesia
- Department of Pediatrics, University of Gadjah Mada, Yogyakarta, Indonesia
| | - Nurjati C. Siregar
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
- Department of Anatomical Pathology, Rumah Sakit Cipto Mangunkusumo and Universitas Indonesia, Jakarta, Indonesia
| | - Pierre A. Buffet
- UMR_S1134, BIGR, Inserm, Université de F-75015 Paris, and Laboratory of Excellence GR-Ex, Paris, France
| | - Nicholas M. Anstey
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
| |
Collapse
|
30
|
Abstract
Cindy S Chu and co-authors review options for diagnosis, safe and radical cure, and relapse prevention of Plasmodium Vivax.
Collapse
Affiliation(s)
- Cindy S. Chu
- Shoklo Malaria Research Unit-Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Headington, Oxford, United Kingdom
- * E-mail:
| | - Nicholas J. White
- Shoklo Malaria Research Unit-Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
31
|
Popovici J, Tebben K, Witkowski B, Serre D. Primaquine for Plasmodium vivax radical cure: What we do not know and why it matters. Int J Parasitol Drugs Drug Resist 2021; 15:36-42. [PMID: 33529838 PMCID: PMC7851417 DOI: 10.1016/j.ijpddr.2020.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 11/26/2022]
Abstract
Plasmodium vivax radical cure requires the administration of a blood schizonticide for killing blood-stage parasites and the addition of a drug able to kill hypnozoites, the dormant parasite stages residing in the liver of infected patients. All drugs used clinically for killing hypnozoites are 8-aminoquinolines and among them, primaquine has been at the forefront of P. vivax case management for decades. We discuss here the possible factors that could lead to the emergence and selection of P. vivax primaquine resistant parasites and emphasize on how a better understanding of the mechanisms underlying primaquine treatment and hypnozoite biology is needed to prevent this catastrophic scenario from happening.
Collapse
Affiliation(s)
- Jean Popovici
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia; Malaria Translational Research Unit, Institut Pasteur, Paris & Institut Pasteur du Cambodge, Phnom Penh, Cambodia.
| | - Kieran Tebben
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia; Malaria Translational Research Unit, Institut Pasteur, Paris & Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
32
|
Bhutani A, Kaushik RM, Kaushik R. A study on multi-organ dysfunction syndrome in malaria using sequential organ failure assessment score. Trop Parasitol 2021; 10:86-94. [PMID: 33747874 PMCID: PMC7951073 DOI: 10.4103/tp.tp_12_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 12/23/2019] [Accepted: 05/30/2020] [Indexed: 11/12/2022] Open
Abstract
Purpose: The purpose of this study is to examine the prevalence, clinical spectrum, prognostic factors, and outcome of multi-organ dysfunction syndrome (MODS) in patients with malaria. Materials and Methods: One hundred and twenty-four patients with malaria, diagnosed by a positive peripheral blood film and rapid malaria test, were studied for MODS using the sequential organ failure assessment (SOFA) score. The severity of malaria was assessed by the WHO criteria. Results: Severe malaria was present in 54 (43.54%) patients. MODS was detected in 108 (87.09%) patients with malaria (Plasmodium falciparum - 57 [85.07%], Plasmodium vivax - 46 [88.46%] and mixed P. falciparum and P. vivax malaria - 5 [100%] cases). SOFA scores of MODS-and non-MODS-patients differed significantly (P < 0.001). No significant association was seen between MODS and type of malaria (P = 0.894). Mortality among malaria patients with MODS was 5.55% (6/108) (P. falciparum 8.77% [5/57] and P. vivax 2.17% [1/46]). The outcome of MODS was associated significantly with the severity of the SOFA score at admission (P = 0.011) but not with the type of malaria, malaria parasite index, and the number of organs involved (P > 0.05 each). The SOFA score at admission correlated significantly with the duration of hospitalization (P < 0.0001). Conclusions: MODS occurs with high frequency in malaria and is not dependent on the type of malaria. The outcome of MODS and recovery time depends on the severity of MODS. The SOFA score is useful in detecting MODS and ascertaining its severity and prognosis in malaria.
Collapse
Affiliation(s)
- Amish Bhutani
- Department of Medicine, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| | - Rajeev Mohan Kaushik
- Department of Medicine, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| | - Reshma Kaushik
- Department of Medicine, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, Uttarakhand, India
| |
Collapse
|
33
|
Walia D, Arora U, Baitha U, Kumar A, Ranjan P, Soneja M, Verma N, Khan MA, Aggarwal P, Biswas A, Wig N. Clinical spectrum and predictors of severe Plasmodium vivax infections at a tertiary care center in North India. Drug Discov Ther 2021; 14:330-335. [PMID: 33390562 DOI: 10.5582/ddt.2020.03110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Traditionally attributed only to Plasmodium falciparum, Plasmodium vivax has recently been reported to cause a significant burden of complicated malaria cases. The present study aimed to delineate the clinical spectrum and identify predictors for severe disease. This was a prospective observational cohort study conducted at a tertiary care hospital in North India. Patients with acute febrile illness (AFI) aged at least 14 years were included if they were diagnosed with vivax malaria based on rapid kits or peripheral smears. Clinical data and investigations during hospital stay was recorded. 439 cases of acute febrile illness were screened, of whom 50 (11%) were diagnosed with malaria including eight P. falciparum infections. Forty-two vivax malaria cases, 22 (52%) of whom were severe, were followed till discharge or death. The median age of the cohort was 24.5 years (Q1-Q3, 19-36 years), including a total of 29 males (69%). Severe malaria was more frequently associated with historical complaints of oliguria or dyspnea, and examination findings of pallor, splenomegaly or altered sensorium. The following five factors were identified to predict severe disease: prolonged illness over 7 days, symptoms of oliguria or dyspnea, examination findings of pallor or crepitations on auscultation. Malaria accounts for 1 in 10 cases of AFI at our North Indian tertiary care center and approximately half of them present with severe disease. Prolonged duration of disease prior to presentation is a modifiable predictor for severe disease and should be targeted for reducing morbidity.
Collapse
Affiliation(s)
- Dinesh Walia
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Umang Arora
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Upendra Baitha
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Arvind Kumar
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Piyush Ranjan
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Soneja
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Nishant Verma
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - Maroof Ahmad Khan
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Praveen Aggarwal
- Department of Emergency Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Ashutosh Biswas
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
34
|
|
35
|
Sangwan J, Minhas A, Mane P, Dhingra A. Pattern of severe malaria with special reference to Plasmodium vivax in pediatric population of the most aspirational region of North India. INTERNATIONAL JOURNAL OF ADVANCED MEDICAL AND HEALTH RESEARCH 2021. [DOI: 10.4103/ijamr.ijamr_21_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
36
|
Patel H, Dunican C, Cunnington AJ. Predictors of outcome in childhood Plasmodium falciparum malaria. Virulence 2020; 11:199-221. [PMID: 32063099 PMCID: PMC7051137 DOI: 10.1080/21505594.2020.1726570] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Plasmodium falciparum malaria is classified as either uncomplicated or severe, determining clinical management and providing a framework for understanding pathogenesis. Severe malaria in children is defined by the presence of one or more features associated with adverse outcome, but there is wide variation in the predictive value of these features. Here we review the evidence for the usefulness of these features, alone and in combination, to predict death and other adverse outcomes, and we consider the role that molecular biomarkers may play in augmenting this prediction. We also examine whether a more personalized approach to predicting outcome for specific presenting syndromes of severe malaria, particularly cerebral malaria, has the potential to be more accurate. We note a general need for better external validation in studies of outcome predictors and for the demonstration that predictors can be used to guide clinical management in a way that improves survival and long-term health.
Collapse
Affiliation(s)
- Harsita Patel
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Claire Dunican
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Aubrey J. Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
37
|
Penman BS, Gandon S. Adaptive immunity selects against malaria infection blocking mutations. PLoS Comput Biol 2020; 16:e1008181. [PMID: 33031369 PMCID: PMC7544067 DOI: 10.1371/journal.pcbi.1008181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/22/2020] [Indexed: 11/18/2022] Open
Abstract
The mutation responsible for Duffy negativity, which impedes Plasmodium vivax infection, has reached high frequencies in certain human populations. Conversely, mutations capable of blocking the more lethal P. falciparum have not succeeded in malarious zones. Here we present an evolutionary-epidemiological model of malaria which demonstrates that if adaptive immunity against the most virulent effects of malaria is gained rapidly by the host, mutations which prevent infection per se are unlikely to succeed. Our results (i) explain the rarity of strain-transcending P. falciparum infection blocking adaptations in humans; (ii) make the surprising prediction that mutations which block P. falciparum infection are most likely to be found in populations experiencing low or infrequent malaria transmission, and (iii) predict that immunity against some of the virulent effects of P. vivax malaria may be built up over the course of many infections.
Collapse
Affiliation(s)
- Bridget S. Penman
- Zeeman Institute and School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Sylvain Gandon
- CEFE, CNRS, University of Montpellier, Paul Valéry University of Montpellier, EPHE, IRD, Montpellier, France
| |
Collapse
|
38
|
Plasmodium spp. mixed infection leading to severe malaria: a systematic review and meta-analysis. Sci Rep 2020; 10:11068. [PMID: 32632180 PMCID: PMC7338391 DOI: 10.1038/s41598-020-68082-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/21/2020] [Indexed: 12/11/2022] Open
Abstract
Mixed Plasmodium malaria infections can lead to severe malaria. This systematic review and meta-analysis aimed to explore the prevalence of severe mixed Plasmodium malaria infection and to compare it with the prevalence of severe P. falciparum malaria mono-infection across the included studies. Original English-language research articles from PubMed, Scopus, and ISI Web of Science were identified and screened. Articles reporting the number of mixed infections and the number of severe mixed infections were used to determine the main outcome of this study, while the number of P. falciparum infections and the number of severe P. falciparum infections were used to determine the secondary outcome of this study. For the main outcome, the pooled prevalence and 95% confidence interval (CI) of severe mixed infections was analysed using STATA software version 15.0 (Stata Corp, College Station, TX, USA). For the secondary outcome, the rate of severe mixed infections compared to severe P. falciparum infections was analysed using the meta-analysis approach, and summary odds ratios (ORs) and 95% CIs were calculated. Random-effects models were used to produce the summary ORs. The Mantel–Haenszel method and calculated I2 were also reported to test whether there was heterogeneity among the included studies. Publication bias was also assessed using funnel plots. The meta-analysis of secondary outcomes was conducted using Review Manager 5.3 software (Cochrane Community). A total of 894,561 malaria patients were reported in all 16 included studies. Overall, a pooled analysis showed that 9% (2,006/35,768, 95% CI 7.0–12.0%) of patients with mixed Plasmodium infection had severe mixed infection. A meta-analysis of 14 studies demonstrated that patients with mixed Plasmodium infection (1,999/35,755) and patients with P. falciparum malaria (9,249/294,397) had an equal risk of developing severe malaria (OR 0.93, 95% CI 0.59–1.44). Both mixed infection and P. falciparum mono-infection showed a similar trend of complications in which severe anaemia, pulmonary failure, and renal impairment were the three most common complications found. However, patients with mixed infection had a higher proportion of severe anaemia and pulmonary complications than those with P. falciparum infection. Moreover, patients with mixed infection had a higher proportion of multiple organ failure than those with P. falciparum mono-infection. Mixed Plasmodium spp. infections were common but often unrecognized or underestimated, leading to severe complications among these malaria patients. Therefore, in routine clinical laboratories, using an accurate combination of diagnostic procedures to identify suspected patients with mixed infections is crucial for therapeutic decisions, prompt treatment, and effective patient management.
Collapse
|
39
|
Rei Yan SL, Wakasuqui F, Wrenger C. Point-of-care tests for malaria: speeding up the diagnostics at the bedside and challenges in malaria cases detection. Diagn Microbiol Infect Dis 2020; 98:115122. [PMID: 32711185 DOI: 10.1016/j.diagmicrobio.2020.115122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022]
Abstract
Malaria remains as one of the major public health problems worldwide. About 228 million cases occurred in 2018 only, with Africa bearing about 93% of the cases. Asymptomatic population carrying the various forms of the parasite Plasmodium in endemic areas plays an important role in the spread of the disease. To tackle this battle, more sensitive and precise detection kits for malaria are crucial to better control the number of new malaria cases. In this review, we not only discuss some of the available approaches to rapidly detect new malaria cases in endemic areas but also shed light on parallel problems that may affect the detection of individuals infected with the parasite, covering kelch 13 mutation, glucose 6-phosphate dehydrogenase deficiency, and hemoglobin disorders. Available approaches for malaria detection covered in this review are focused on point-of-care tests, including portable polymerase chain reaction and aptamers.
Collapse
Affiliation(s)
- Sun L Rei Yan
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil
| | - Felipe Wakasuqui
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
40
|
Oboh MA, Singh US, Ndiaye D, Badiane AS, Ali NA, Bharti PK, Das A. Presence of additional Plasmodium vivax malaria in Duffy negative individuals from Southwestern Nigeria. Malar J 2020; 19:229. [PMID: 32590997 PMCID: PMC7318376 DOI: 10.1186/s12936-020-03301-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/17/2020] [Indexed: 11/13/2022] Open
Abstract
Background Malaria in sub-Saharan Africa (sSA) is thought to be mostly caused by Plasmodium falciparum. Recently, growing reports of cases due to Plasmodium ovale, Plasmodium malariae, and Plasmodium vivax have been increasingly observed to play a role in malaria epidemiology in sSA. This in fact is due to the usage of very sensitive diagnostic tools (e.g. PCR), which have highlighted the underestimation of non-falciparum malaria in this sub-region. Plasmodium vivax was historically thought to be absent in sSA due to the high prevalence of the Duffy negativity in individuals residing in this sub-continent. Recent studies reporting detection of vivax malaria in Duffy-negative individuals from Mali, Mauritania, Cameroon challenge this notion. Methods Following previous report of P. vivax in Duffy-negative individuals in Nigeria, samples were further collected and assessed RDT and/or microscopy. Thereafter, malaria positive samples were subjected to conventional PCR method and DNA sequencing to confirm both single/mixed infections as well as the Duffy status of the individuals. Results Amplification of Plasmodium gDNA was successful in 59.9% (145/242) of the evaluated isolates and as expected P. falciparum was the most predominant (91.7%) species identified. Interestingly, four P. vivax isolates were identified either as single (3) or mixed (one P. falciparum/P. vivax) infection. Sequencing results confirmed all vivax isolates as truly vivax malaria and the patient were of Duffy-negative genotype. Conclusion Identification of additional vivax isolates among Duffy-negative individuals from Nigeria, substantiate the expanding body of evidence on the ability of P. vivax to infect RBCs that do not express the DARC gene. Hence, such genetic-epidemiological study should be conducted at the country level in order to evaluate the true burden of P. vivax in Nigeria.
Collapse
Affiliation(s)
- Mary Aigbiremo Oboh
- Medical Research Council Unit The Gambia at LSHTM, Fajara, P.O. Box 273, Banjul, Gambia.
| | - Upasana Shyamsunder Singh
- School of Earth and Environmental Sciences, University of Manchester, Manchester, UK.,Genomic Epidemiology Laboratory, Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India
| | - Daouda Ndiaye
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Aida Sadikh Badiane
- Parasitology and Mycology Laboratory, Université Cheikh Anta Diop, Dakar, Senegal
| | - Nazia Anwar Ali
- National Institute of Research in Tribal Health, Jabalpur, Madhya Pradesh, 482003, India
| | - Praveen Kumar Bharti
- National Institute of Research in Tribal Health, Jabalpur, Madhya Pradesh, 482003, India
| | - Aparup Das
- Genomic Epidemiology Laboratory, Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Jabalpur, India.
| |
Collapse
|
41
|
Dash M, Pande V, Sinha A. Putative circumsporozoite protein (CSP) of Plasmodium vivax is considerably distinct from the well-known CSP and plays a role in the protein ubiquitination pathway. Gene 2020; 721S:100024. [PMID: 32550551 PMCID: PMC7285988 DOI: 10.1016/j.gene.2019.100024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 11/29/2022]
Abstract
Amidst technical challenges which limit successful culture and genetic manipulation of P. vivax parasites, we used a computational approach to identify a critical target with evolutionary significance. The putative circumsporozoite protein on chromosome 13 of P. vivax (PvpuCSP)is distinct from the well-known vaccine candidate PfCSP. The aim of this study was to understand the role of PvpuCSP and its relatedness to the well-known CSP. The study revealed PvpuCSP as a membrane bound E3 ubiquitin ligase involved in ubiquitination. It has a species-specific tetra-peptide unit which is differentially repeated in various P. vivax strains. The PvpuCSP is different from CSP in terms of stage-specific expression and function. Since E3 ubiquitin ligases are known antimalarial drug targets targeting the proteasome pathway, PvpuCSP, with evolutionary connotation and a key role in orchestrating protein degradation in P. vivax, can be explored as a potential drug target. PvpuCSP is predicted as E3 ubiquitin ligase, a part of ubiquitination pathway. Tetra-peptide tandem repeat at C terminal of PvpuCSP is exclusive to P. vivax. Moderately expressed during all parasitic stages in host and vector Partially disordered protein with both structured domains and two distinct IDRs A transmembrane protein with highly conserved functional domain across Apicomplexa
Collapse
Affiliation(s)
- Manoswini Dash
- Division of Epidemiology and Clinical Research, ICMR-National Institute of Malaria Research, New Delhi, India
| | - Veena Pande
- Department of Biotechnology, Bhimtal Campus, Kumaun University, Nainital, Uttarakhand, India
| | - Abhinav Sinha
- Division of Epidemiology and Clinical Research, ICMR-National Institute of Malaria Research, New Delhi, India
| |
Collapse
|
42
|
Comparison of real time and malachite-green based loop-mediated isothermal amplification assays for the detection of Plasmodium vivax and P. falciparum. PLoS One 2020; 15:e0234263. [PMID: 32525900 PMCID: PMC7289405 DOI: 10.1371/journal.pone.0234263] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/21/2020] [Indexed: 11/26/2022] Open
Abstract
The current context of malaria elimination requires urgent development and implementation of highly sensitive and specific methods for prompt detection and treatment of malaria parasites. Such methods should overcome current delays in diagnosis, allow the detection of low-density infections and address the difficulties in accessing remote endemic communities. In this study, we assessed the performance of the RealAmp and malachite-green loop mediated isothermal amplification (MG-LAMP) methodologies, using microscopy and conventional nested-PCR as reference techniques. Both LAMP techniques were performed for Plasmodium genus, P. falciparum, and P. vivax identification using 136 whole blood samples collected from three communities located in the Peruvian Amazon basin. Turnaround time and costs of performing the LAMP assays were estimated and compared to that of microscopy and nested-PCR. Using nested-PCR as reference standard, we calculated the sensitivity, specificity and 95% confidence interval (CI) for all methods. RealAmp had a sensitivity of 92% (95% CI: 85–96.5%) and specificity of 100% (95% CI: 89.1–100%) for species detection; sensitivity and specificity of MG-LAMP were 94% (95% CI: 87.5–97.8%) and 100% (89.1–100%), respectively. Whereas microscopy showed 88.1% sensitivity (95% CI: 80.2–93.7%) and 100% specificity (95%: 89.1–100%). The turnaround time and costs of performing the LAMP assays were lower compared to those associated with nested-PCR but higher than those associated with microscopy. The two LAMP assays were shown to be more sensitive and simple to implement than microscopy. Both LAMP methodologies could be used as large-scale screening tests, but the MG-LAMP assay uses a simple, portable heat-block while the RealAmp requires a RealAmp machine or a real-time PCR machine. This makes the MG-LAMP an appropriate choice for malaria surveillance studies in endemic sites. Use of LAMP tests in active case detection of Plasmodium parasites could help to detect positive malaria cases early.
Collapse
|
43
|
Plasmodium vivax in the Era of the Shrinking P. falciparum Map. Trends Parasitol 2020; 36:560-570. [PMID: 32407682 PMCID: PMC7297627 DOI: 10.1016/j.pt.2020.03.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 01/13/2023]
Abstract
Plasmodium vivax is an important cause of malaria, associated with a significant public health burden. Whilst enhanced malaria-control activities have successfully reduced the incidence of Plasmodium falciparum malaria in many areas, there has been a consistent increase in the proportion of malaria due to P. vivax in regions where both parasites coexist. This article reviews the epidemiology and biology of P. vivax, how the parasite differs from P. falciparum, and the key features that render it more difficult to control and eliminate. Since transmission of the parasite is driven largely by relapses from dormant liver stages, its timely elimination will require widespread access to safe and effective radical cure.
Collapse
|
44
|
Graham H, Bakare AA, Ayede AI, Oyewole OB, Gray A, Neal E, Qazi SA, Duke T, Falade AG. Diagnosis of pneumonia and malaria in Nigerian hospitals: A prospective cohort study. Pediatr Pulmonol 2020; 55 Suppl 1:S37-S50. [PMID: 32074408 PMCID: PMC7318580 DOI: 10.1002/ppul.24691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/30/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Pneumonia and malaria are the leading causes of global childhood mortality. We describe the clinical presentation of children diagnosed with pneumonia and/or malaria, and identify possible missed cases and diagnostic predictors. METHODS Prospective cohort study involving children (aged 28 days to 15 years) admitted to 12 secondary-level hospitals in south-west Nigeria, from November 2015 to October 2017. We described children diagnosed with malaria and/or pneumonia on admission and identified potential missed cases using WHO criteria. We used logistic regression models to identify associations between clinical features and severe pneumonia and malaria diagnoses. RESULTS Of 16 432 admitted children, 16 184 (98.5%) had adequate data for analysis. Two-thirds (10 561, 65.4%) of children were diagnosed with malaria and/or pneumonia by the admitting doctor; 31.5% (567/1799) of those with pneumonia were also diagnosed with malaria. Of 1345 (8.3%) children who met WHO severe pneumonia criteria, 557 (41.4%) lacked a pneumonia diagnosis. Compared with "potential missed" diagnoses of severe pneumonia, children with "detected" severe pneumonia were more likely to receive antibiotics (odds ratio [OR], 4.03; 2.63-6.16, P < .001), and less likely to die (OR, 0.72; 0.51-1.02, P = .067). Of 2299 (14.2%) children who met WHO severe malaria criteria, 365 (15.9%) lacked a malaria diagnosis. Compared with "potential missed" diagnoses of severe malaria, children with "detected" severe malaria were less likely to die (OR, 0.59; 0.38-0.91, P = 0.017), with no observed difference in antimalarial administration (OR, 0.29; 0.87-1.93, P = .374). We identified predictors of severe pneumonia and malaria diagnosis. CONCLUSION Pneumonia should be considered in all severely unwell children with respiratory signs, regardless of treatment for malaria or other conditions.
Collapse
Affiliation(s)
- Hamish Graham
- Centre for International Child Health, MCRI, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,Department of Paediatrics, University College Hospital, Ibadan, Nigeria
| | - Ayobami A Bakare
- Department of Community Medicine, University College Hospital, Ibadan, Nigeria
| | - Adejumoke I Ayede
- Department of Paediatrics, University College Hospital, Ibadan, Nigeria.,Department of Paediatrics, University of Ibadan, Ibadan, Nigeria
| | - Oladapo B Oyewole
- Department of Paediatrics, University College Hospital, Ibadan, Nigeria
| | - Amy Gray
- Centre for International Child Health, MCRI, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Eleanor Neal
- Centre for International Child Health, MCRI, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia.,Infection & Immunity, MCRI, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Shamim A Qazi
- Department of Maternal, Newborn, Child and Adolescent Health, World Health Organization, Geneva, Switzerland
| | - Trevor Duke
- Centre for International Child Health, MCRI, Royal Children's Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Adegoke G Falade
- Department of Paediatrics, University College Hospital, Ibadan, Nigeria.,Department of Paediatrics, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
45
|
Taylor WRJ, Kheng S, Muth S, Tor P, Kim S, Bjorge S, Topps N, Kosal K, Sothea K, Souy P, Char CM, Vanna C, Ly P, Khieu V, Christophel E, Kerleguer A, Pantaleo A, Mukaka M, Menard D, Baird JK. Hemolytic Dynamics of Weekly Primaquine Antirelapse Therapy Among Cambodians With Acute Plasmodium vivax Malaria With or Without Glucose-6-Phosphate Dehydrogenase Deficiency. J Infect Dis 2020; 220:1750-1760. [PMID: 31549159 PMCID: PMC6804333 DOI: 10.1093/infdis/jiz313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
Background Hemoglobin (Hb) data are limited in Southeast Asian glucose-6-phosphate dehydrogenase (G6PD) deficient (G6PD−) patients treated weekly with the World Health Organization–recommended primaquine regimen (ie, 0.75 mg/kg/week for 8 weeks [PQ 0.75]). Methods We treated Cambodians who had acute Plasmodium vivax infection with PQ0.75 and a 3-day course of dihydroartemisinin/piperaquine and determined the Hb level, reticulocyte count, G6PD genotype, and Hb type. Results Seventy-five patients (male sex, 63) aged 5–63 years (median, 24 years) were enrolled. Eighteen were G6PD deficient (including 17 with G6PD Viangchan) and 57 were not G6PD deficient; 26 had HbE (of whom 25 were heterozygous), and 6 had α-/β-thalassemia. Mean Hb concentrations at baseline (ie, day 0) were similar between G6PD deficient and G6PD normal patients (12.9 g/dL [range, 9‒16.3 g/dL] and 13.26 g/dL [range, 9.6‒16 g/dL], respectively; P = .46). G6PD deficiency (P = <.001), higher Hb concentration at baseline (P = <.001), higher parasitemia level at baseline (P = .02), and thalassemia (P = .027) influenced the initial decrease in Hb level, calculated as the nadir level minus the baseline level (range, −5.8–0 g/dL; mean, −1.88 g/dL). By day 14, the mean difference from the day 7 level (calculated as the day 14 level minus the day 7 level) was 0.03 g/dL (range, −0.25‒0.32 g/dL). Reticulocyte counts decreased from days 1 to 3, peaking on day 7 (in the G6PD normal group) and day 14 (in the G6PD deficient group); reticulocytemia at baseline (P = .001), G6PD deficiency (P = <.001), and female sex (P = .034) correlated with higher counts. One symptomatic, G6PD-deficient, anemic male patient was transfused on day 4. Conclusions The first PQ0.75 exposure was associated with the greatest decrease in Hb level and 1 blood transfusion, followed by clinically insignificant decreases in Hb levels. PQ0.75 requires monitoring during the week after treatment. Safer antirelapse regimens are needed in Southeast Asia. Clinical Trials Registration ACTRN12613000003774.
Collapse
Affiliation(s)
- Walter R J Taylor
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia.,Service de Médecine Tropicale et Humanitaire, Hôpitaux Universitaires de Genève, Switzerland.,Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Sim Kheng
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sinoun Muth
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Pety Tor
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Saorin Kim
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Steven Bjorge
- World Health Organization (WHO) Cambodia Country Office, Phnom Penh, Cambodia
| | - Narann Topps
- World Health Organization (WHO) Cambodia Country Office, Phnom Penh, Cambodia
| | - Khem Kosal
- Pailin Referral Hospital, Pailin, Cambodia
| | | | - Phum Souy
- Anlong Veng Referral Hospital, Anlong Venh, Cambodia
| | - Chuor Meng Char
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Chan Vanna
- Pramoy Health Center, Veal Veng, Cambodia
| | - Po Ly
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Virak Khieu
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Eva Christophel
- WHO Western Pacific Regional Office, Manila, the Philippines
| | | | | | - Mavuto Mukaka
- Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand.,Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Didier Menard
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia.,Malaria Genetics and Resistance Group, Biology of Host-Parasite Interactions Unit, Institut Pasteur, Paris, France
| | - J Kevin Baird
- Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, United Kingdom.,Eijkman Oxford Clinical Research Unit, Eijkman Institute of Molecular Biology, Jakarta, Indonesia
| |
Collapse
|
46
|
Population structure and genetic diversity of Trichomonas vaginalis clinical isolates in Australia and Ghana. INFECTION GENETICS AND EVOLUTION 2020; 82:104318. [PMID: 32278146 DOI: 10.1016/j.meegid.2020.104318] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/30/2022]
Abstract
Population genetic studies of Trichomonas vaginalis have detected high genetic diversity associated with phenotypic differences in clinical presentations. In this study, microscopy and next generation-multi-locus sequence typing (NG-MLST) were used to identify and genetically characterise T. vaginalis isolates from patients in Australia and Ghana. Seventy-one polymorphic nucleotide sites, 36 different alleles, 48 sequence types, 24 of which were novel, were identified among 178 isolates, revealing a geneticallly diverse T. vaginalis population. Polymorphism was found at most loci, clustering genotypes into eight groups among both Australian and Ghanaian isolates, although there was some variation between countries. The number of alleles for each locus ranged from two to nine. Study results confirmed geographic expansion and diversity of the T. vaginalis population. Two-type populations in almost equal frequencies and a third unassigned group were identified in this study. Linkage disequilibrium was observed, suggesting T. vaginalis population is highly clonal. Multillocus disequilibrium was observed even when analysing clades separately, as well as widespread clonal genotypes, suggesting that there is no evidence of recent recombination. A more comprehensive study to assess the extent of genetic diversity and population structure of T. vaginalis and their potential impact on varied pathology observed among infected individuals is recommended.
Collapse
|
47
|
Douglas NM, Kenangalem E, Hasanuddin A, Anstey NM, Sugiarto P, Price RN, Poespoprodjo JR. Malaria-related hospitalization during childhood in Papua, Indonesia: A retrospective cohort study. PLoS One 2020; 15:e0228018. [PMID: 31995581 PMCID: PMC6988973 DOI: 10.1371/journal.pone.0228018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/05/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND In endemic regions, the age distribution of malaria varies according to the infecting Plasmodium species. We aimed to delineate the pattern of malaria-related hospitalization from birth in Timika, Papua-an area co-endemic for P. falciparum and P. vivax. METHODS Between April 2004 and December 2013, infants born at Mitra Masyarakat Hospital, or presenting within the first 7 days of life, were enrolled retrospectively into a cohort study and followed passively using routinely-collected hospital surveillance data. Outcomes were stratified by the presence or absence of Plasmodium parasitemia and included re-presentation to hospital, requirement for hospital admission and death. RESULTS Overall, 11,408 infants were enrolled into the cohort. Median follow-up was 4.3 (maximum 9.7) years. In total, 7,847 (68.9%) infants made 90,766 re-presentations to hospital, 18,105 (19.9%) of which were associated with Plasmodium parasitemia. The incidence of re-presentations with malaria during the first year of life was 213 per 1,000 person-years (py) for P. vivax and 79 per 1,000py for P. falciparum (Incidence Rate Ratio (IRR) = 2.69, 95% Confidence Interval (95%CI): 2.48-2.92). After the age of 5 years, the incidence of P. vivax had fallen to 77/1,000py and the incidence of P. falciparum had risen to 95/1,000py (IRR = 0.80, 95%CI: 0.73-0.88). Overall, 79.7% (14,431/18,105) of malaria re-presentations were recurrences rather than initial infections. Malaria accounted for 31.7% (2,126/3,120) of all hospital admissions. The infant mortality rate in this study was 52 deaths per 1,000 live births. Beyond the early neonatal period, 13.4% of deaths were associated with Plasmodium parasitemia. CONCLUSIONS In Papua, Indonesia, malaria is a major cause of hospital presentation and admission in early life. The initial predominance of P. vivax over P. falciparum inverts after five years of age. Malaria is directly associated with nearly one in seven deaths after the early neonatal period.
Collapse
Affiliation(s)
- Nicholas M. Douglas
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Division of Infectious Diseases, Royal Darwin Hospital, Darwin, Australia
- * E-mail:
| | - Enny Kenangalem
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Mimika District Health Authority, Timika, Papua, Indonesia
| | - Afdhal Hasanuddin
- Department of Paediatrics, Rumah Sakit Mitra Masyarakat, Timika, Papua, Indonesia
| | - Nicholas M. Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Division of Infectious Diseases, Royal Darwin Hospital, Darwin, Australia
| | - Paulus Sugiarto
- Department of Paediatrics, Rumah Sakit Mitra Masyarakat, Timika, Papua, Indonesia
| | - Ric N. Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jeanne Rini Poespoprodjo
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Department of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Paediatrics, Rumah Sakit Umum Daerah, Kabupaten Mimika, Timika, Papua, Indonesia
| |
Collapse
|
48
|
Tang J, Templeton TJ, Cao J, Culleton R. The Consequences of Mixed-Species Malaria Parasite Co-Infections in Mice and Mosquitoes for Disease Severity, Parasite Fitness, and Transmission Success. Front Immunol 2020; 10:3072. [PMID: 32038623 PMCID: PMC6987389 DOI: 10.3389/fimmu.2019.03072] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/16/2019] [Indexed: 11/13/2022] Open
Abstract
The distributions of human malaria parasite species overlap in most malarious regions of the world, and co-infections involving two or more malaria parasite species are common. Little is known about the consequences of interactions between species during co-infection for disease severity and parasite transmission success. Anti-malarial interventions can have disproportionate effects on malaria parasite species and may locally differentially reduce the number of species in circulation. Thus, it is important to have a clearer understanding of how the interactions between species affect disease and transmission dynamics. Controlled competition experiments using human malaria parasites are impossible, and thus we assessed the consequences of mixed-species infections on parasite fitness, disease severity, and transmission success using the rodent malaria parasite species Plasmodium chabaudi, Plasmodium yoelii, and Plasmodium vinckei. We compared the fitness of individual species within single species and co-infections in mice. We also assessed the disease severity of single vs. mixed infections in mice by measuring mortality rates, anemia, and weight loss. Finally, we compared the transmission success of parasites in single or mixed species infections by quantifying oocyst development in Anopheles stephensi mosquitoes. We found that co-infections of P. yoelii with either P. vinckei or P. chabaudi led to a dramatic increase in infection virulence, with 100% mortality observed in mixed species infections, compared to no mortality for P. yoelii and P. vinckei single infections, and 40% mortality for P. chabaudi single infections. The increased mortality in the mixed infections was associated with an inability to clear parasitaemia, with the non-P. yoelii parasite species persisting at higher parasite densities than in single infections. P. yoelii growth was suppressed in all mixed infections compared to single infections. Transmissibility of P. vinckei and P. chabaudi to mosquitoes was also reduced in the presence of P. yoelii in co-infections compared to single infections. The increased virulence of co-infections containing P. yoelii (reticulocyte restricted) and P. chabaudi or P. vinckei (predominantly normocyte restricted) may be due to parasite cell tropism and/or immune modulation of the host. We explain the reduction in transmission success of species in co-infections in terms of inter-species gamete incompatibility.
Collapse
Affiliation(s)
- Jianxia Tang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China.,Malaria Unit, Department of Pathology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Thomas J Templeton
- Department of Protozoology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Jun Cao
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Richard Culleton
- Malaria Unit, Department of Pathology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
49
|
Patriani D, Arguni E, Kenangalem E, Dini S, Sugiarto P, Hasanuddin A, Lampah DA, Douglas NM, Anstey NM, Simpson JA, Price RN, Poespoprodjo JR. Early and late mortality after malaria in young children in Papua, Indonesia. BMC Infect Dis 2019; 19:922. [PMID: 31666012 PMCID: PMC6820991 DOI: 10.1186/s12879-019-4497-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/23/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In southern Papua, Indonesia, malaria is highly prevalent in young children and is a significant cause of morbidity and early mortality. The association between malaria and delayed mortality is unknown. METHODS Routinely-collected hospital surveillance data from southern Papua, Indonesia, were used to assess the risk of recurrent malaria and mortality within 12 months of an initial presentation with malaria in all children younger than 5 years old attending the local hospital. Analysis was primarily by Kaplan Meier and Cox regression methods. RESULTS In total 15,716 children presenting with malaria between April 2004 and December 2013 were included in the analysis; 6184 (39.3%) with Plasmodium falciparum, 7499 (47.7%) with P. vivax, 203 (1.3%) with P. malariae, 3 with P. ovale and 1827 (11.6%) with mixed infections. Within 1 year, 48.4% (7620/15,716) of children represented a total of 16,957 times with malaria (range 1 to 11 episodes), with the incidence of malaria being greater in patients initially presenting with P. vivax infection (1334 [95%CI 1307-1361] per 1000 patient years) compared to those with P. falciparum infection (920 [896-944]). In total 266 (1.7%) children died within 1 year of their initial presentation, 129 (48.5%) within 30 days and 137 (51.5%) between 31 and 365 days. There was no significant difference in the mortality risk in patients infected with P. vivax versus P. falciparum either before 30 days (Hazard Ratio (HR) 1.02 [0.69,1.49]) or between 31 and 365 days (HR = 1.30 [0.90,1.88]). Children who died had a greater incidence of malaria, 2280 [95%CI 1946-2671] per 1000 patient years preceding their death, compared to 1141 [95%CI 1124-1158] per 1000 patient years in those surviving. CONCLUSIONS Children under-5 years old with P. vivax malaria, are at significant risk of multiple representations with malaria and of dying within 1 year of their initial presentation. Preventing recurrent malaria must be a public health priority in this vulnerable population.
Collapse
Affiliation(s)
- Dewi Patriani
- grid.8570.aDepartment of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Kesehatan no.1, Sekip, Yogyakarta, 55284 Indonesia
| | - Eggi Arguni
- grid.8570.aDepartment of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Kesehatan no.1, Sekip, Yogyakarta, 55284 Indonesia
| | - Enny Kenangalem
- Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, Papua 99910 Indonesia ,Mimika District Hospital, Jl. Yos Sudarso, Timika, Papua 99910 Indonesia
| | - Saber Dini
- 0000 0001 2179 088Xgrid.1008.9Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, 207 Bouverie Street, The University of Melbourne, Melbourne, Victoria 3010 Australia
| | - Paulus Sugiarto
- Mitra Masyarakat Hospital, Jl. SP2-SP5-Charitas, Timika, 99910 Indonesia
| | - Afdhal Hasanuddin
- Mitra Masyarakat Hospital, Jl. SP2-SP5-Charitas, Timika, 99910 Indonesia
| | - Daniel Adrian Lampah
- Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, Papua 99910 Indonesia
| | - Nicholas M. Douglas
- 0000 0000 8523 7955grid.271089.5Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, PO Box 41096, Casuarina, Darwin, NT 0811 Australia
| | - Nicholas M. Anstey
- 0000 0000 8523 7955grid.271089.5Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, PO Box 41096, Casuarina, Darwin, NT 0811 Australia ,grid.240634.7Division of Medicine, Royal Darwin Hospital, Darwin, NT 0810 Australia
| | - Julie Anne Simpson
- Mitra Masyarakat Hospital, Jl. SP2-SP5-Charitas, Timika, 99910 Indonesia
| | - Ric N. Price
- 0000 0000 8523 7955grid.271089.5Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, PO Box 41096, Casuarina, Darwin, NT 0811 Australia ,0000 0004 1936 8948grid.4991.5Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, OX37LJ, Oxford, United Kingdom ,0000 0004 1937 0490grid.10223.32Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jeanne Rini Poespoprodjo
- grid.8570.aDepartment of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Jl. Kesehatan no.1, Sekip, Yogyakarta, 55284 Indonesia ,Timika Malaria Research Programme, Papuan Health and Community Development Foundation, Jl. SP2-SP5, RSMM Area, Timika, Papua 99910 Indonesia ,Mimika District Hospital, Jl. Yos Sudarso, Timika, Papua 99910 Indonesia
| |
Collapse
|
50
|
Meltzer E, Rahav G, Schwartz E. Vivax Malaria Chemoprophylaxis: The Role of Atovaquone-Proguanil Compared to Other Options. Clin Infect Dis 2019; 66:1751-1755. [PMID: 29228132 DOI: 10.1093/cid/cix1077] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/04/2017] [Indexed: 01/01/2023] Open
Abstract
Background Atovaquone-proguanil is considered causal prophylaxis (inhibition of liver-stage schizonts) for Plasmodium falciparum; however, its causal prophylactic efficacy for Plasmodium vivax is not known. Travelers returning to nonendemic areas provide a unique opportunity to study P. vivax prophylaxis. Methods In a retrospective observational study, for 11 years, Israeli rafters who had traveled to the Omo River in Ethiopia, a highly malaria-endemic area, were followed for at least 1 year after their return. Malaria prophylaxis used during this period included mefloquine, doxycycline, primaquine, and atovaquone-proguanil. Prophylaxis failure was divided into early (within a month of exposure) and late malaria. Results Two hundred fifty-two travelers were included in the study. Sixty-two (24.6%) travelers developed malaria, 56 (91.9%) caused by P. vivax, with 54 (87.1%) cases considered as late malaria. Among travelers using atovaquone-proguanil, there were no cases of early P. falciparum or P. vivax malaria. However, 50.0% of atovaquone-proguanil users developed late vivax malaria, as did 46.5% and 43.5% of mefloquine and doxycycline users, respectively; only 2 (1.4%) primaquine users developed late malaria (P < .0001). Conclusions Short-course atovaquone-proguanil appears to provide causal (liver schizont stage) prophylaxis for P. vivax, but is ineffective against late, hypnozoite reactivation-related attacks. These findings suggest that primaquine should be considered as the chemoprophylactic agent of choice for areas with high co-circulation of P. falciparum and P. vivax.
Collapse
Affiliation(s)
- Eyal Meltzer
- Center for Geographic Medicine and Tropical Diseases, Tel Hashomer, Israel.,Department of Medicine C, Tel Hashomer, Israel
| | - Galia Rahav
- Infectious Diseases Unit, The Sheba Medical Center, Tel Hashomer, Israel.,Sackler School of Medicine, Tel Aviv University, Israel
| | - Eli Schwartz
- Center for Geographic Medicine and Tropical Diseases, Tel Hashomer, Israel.,Department of Medicine C, Tel Hashomer, Israel.,Sackler School of Medicine, Tel Aviv University, Israel
| |
Collapse
|