1
|
Okwu DG, Zoleko Manego R, Duparc S, Kremsner PG, Ramharter M, Mombo-Ngoma G. The non-artemisinin antimalarial drugs under development: a review. Clin Microbiol Infect 2025; 31:941-947. [PMID: 40120754 DOI: 10.1016/j.cmi.2025.03.009] [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: 10/18/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND In 2022, malaria caused approximately 249 million cases and 608,000 deaths, primarily in Africa. Current treatments target asexual blood-stage parasites, gametocytes, and liver hypnozoites. Standard guidelines recommend a 3-day course of artemisinin-based combination therapies as a first-line treatment of uncomplicated malaria and parenteral artesunate for severe malaria. However, the emergence of partial resistance to artemisinin derivatives threatens the treatment efficacy, highlighting the urgent need for novel antimalarial drugs. OBJECTIVES This review summarizes recent progress in the clinical development of antimalarials particularly non-artemisinin compounds under target product profile-1. SOURCES Data were gathered from Medicines for Malaria Venture (MMV)'s portfolio and clinical trial databases between 2020 and 2024. CONTENT Sixteen clinical trials were reviewed, including safety and efficacy studies involving healthy volunteers, experimentally infected volunteers, asymptomatic Plasmodium falciparum carriers and malaria patients. Six trials evaluated the safety and tolerability of MMV533, ZY19489, INE963, GSK701/MMV367 and intravenous KAE609 in healthy volunteers. Efficacy trials involving experimentally infected volunteers assessed ZY19489 and GSK701/MMV367, whereas studies on asymptomatic carriers tested ZY19489/ferroquine and cabamiquine/pyronaridine. Trials on malaria patients investigated combinations of ganaplacide/lumefantrine-SDF, cabamiquine/pyronaridine, both oral and intravenous cipargamin and INE963. IMPLICATIONS Although attrition remains a possibility, several promising candidate drugs with novel modes of action are advancing through clinical development. Many are expected to become available for treating uncomplicated and severe malaria within the next decade. These new antimalarials could significantly enhance malaria treatment, reduce resistance, and support global health efforts towards malaria control, elimination and, potentially, eradication.
Collapse
Affiliation(s)
- Dearie Glory Okwu
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon; Department of Implementation Research, Bernhard Nocht Institute for Tropical Medicine & I. Dep. of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Rella Zoleko Manego
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon; German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany; Centre for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Dep. of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | | | - Peter Gottfried Kremsner
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon; Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Tübingen, Germany
| | - Michael Ramharter
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon; German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany; Centre for Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Dep. of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Ghyslain Mombo-Ngoma
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon; Department of Implementation Research, Bernhard Nocht Institute for Tropical Medicine & I. Dep. of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany; German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.
| |
Collapse
|
2
|
Bestgen B, Jones S, Thathy V, Kuemmerle A, Barcelo C, Haouala A, Gossen D, Marx MW, Di Resta I, Szramowska M, Webster RA, Llewellyn S, Ritacco DA, Yeo T, Leroy D, Barber BE, Fidock DA, Griffin P, Lickliter J, Chalon S. Safety, tolerability, pharmacokinetics, and antimalarial activity of MMV533: a phase 1a first-in-human, randomised, ascending dose and food effect study, and a phase 1b Plasmodium falciparum volunteer infection study. THE LANCET. INFECTIOUS DISEASES 2025; 25:507-518. [PMID: 39708824 DOI: 10.1016/s1473-3099(24)00664-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Novel antimalarials are needed to address emerging resistance to artemisinin and partner drugs. We did two trials to evaluate safety, tolerability, pharmacokinetics, and activity against blood stage Plasmodium falciparum for the drug candidate MMV533. METHODS A phase 1a first-in-human (FIH) trial was conducted at Nucleus Network (Melbourne, VIC, Australia). Part 1 was a double-blind, randomised, placebo-controlled, sequential ascending dose study and part 2 was an open-label, randomised, two-period crossover, pilot food effect study. A phase 1b, open-label, volunteer infection study (VIS) was conducted at Nucleus Network (Herston, QLD, Australia). Eligible participants were adults aged 18-55 years, with a bodyweight of at least 50 kg and BMI of 18-32 kg/m2 and participants in the VIS were malaria-naive. In part 1 of the FIH study, six cohorts of up to eight participants were randomly assigned (3:1) to a single oral MMV533 dose (5, 10, 20, 50, 100, and 160 mg) or placebo using an automated system, with study staff and participants masked to treatment allocation, and follow-up until day 28. In part 2, MMV533 30 mg was administered open-label to one cohort of nine participants assigned by simple randomisation (1:1) to the fasted-fed (n=4) or fed-fasted (n=5) groups. After a 21-day washout period, fed and fasted groups crossed over with follow-up until day 42. In the VIS, seven participants were assigned using simple randomisation (1:1:1) to three dosing groups of 20 mg (n=3), 35 mg (n=2), or 100 mg (n=2) after parasitaemia was detected, with follow-up until day 28. The primary outcomes were treatment emergent adverse events and relationship to MMV533 for the FIH study assessed in the safety population, and in the VIS primary outcomes were parasite reduction ratio over 48 h (log10PRR48), parasite clearance half-life (PCT1/2), and lag phase assessed in the pharmacodynamic population. MMV533 pharmacokinetics was a secondary outcome for both studies, evaluated in the pharmacokinetic population. The studies are registered with ClinicalTrials.gov, NCT04323306 and NCT05205941 (completed). FINDINGS The FIH study was conducted between July 31, 2020, and Sept 27, 2022, and the VIS between March 31 and Aug 9, 2022. 335 adults were assessed for eligibility, 71 enrolled, and 69 randomly assigned (53 in part 1 and nine in part 2 of the FIH study, and seven in the VIS). 32 (45%) of 71 participants were female and 39 (55%) were male. In part 1, 24 (63%) of 38 participants had an adverse event after MMV533 administration with no apparent relationship to dose versus six (50%) of 12 after placebo. Treatment-related adverse events were reported for four (11%) participants receiving MMV533 and one (8%) receiving placebo, with no relationship to dose. In part 2, adverse events were reported for three (38%) of eight participants when fasted and four (44%) of nine when fed, with no apparent influence of food. Time to maximum plasma concentration was 4·0-6·0 h, and apparent half-life was 103·8-127·2 h. After a high-fat meal, the geometric mean ratio (fed:fasted) of MMV533 AUC0-last was 112·0 (90% CI 89·6-140·0). In the VIS for MMV533 100 mg, log10PRR48 was 2·27 (1·99-2·56), PCT1/2 was 6·36 h (5·64-7·28), and lag phase was 2 h. INTERPRETATION An acceptable safety and tolerability profile, confirmed parasiticidal activity, and a long half-life support progression of MMV533 into clinical trials in patients with malaria as a component of new antimalarial combination therapies. FUNDING MMV Medicines for Malaria Venture and Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
| | - Sam Jones
- MMV Medicines for Malaria Venture, Geneva, Switzerland
| | - Vandana Thathy
- Department of Microbiology and Immunology, Columbia University Irving Medical Centre, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Amina Haouala
- MMV Medicines for Malaria Venture, Geneva, Switzerland
| | | | | | | | | | | | | | - Dominic A Ritacco
- Department of Microbiology and Immunology, Columbia University Irving Medical Centre, New York, NY, USA
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Centre, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Didier Leroy
- MMV Medicines for Malaria Venture, Geneva, Switzerland
| | | | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Centre, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Paul Griffin
- The University of Queensland, Brisbane, QLD, Australia
| | | | | |
Collapse
|
3
|
Lynch SA, Abd-Rahman AN, Peters JM, Heunis JM, S E Gower J, Potter AJ, Webster R, Jennings H, Mathison S, Sahai N, Amante FH, Barber BE. Transmissibility of a new Plasmodium falciparum 3D7 bank for use in malaria volunteer infection studies evaluating transmission blocking interventions. Sci Rep 2025; 15:13094. [PMID: 40240504 PMCID: PMC12003780 DOI: 10.1038/s41598-025-97282-y] [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: 12/03/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Transmission blocking activity is an important characteristic of antimalarial drugs, and can be evaluated in malaria volunteer infection studies (VIS). We undertook a pilot VIS to evaluate the suitability of a recently manufactured Plasmodium falciparum 3D7 bank (3D7-MBE-008) for evaluating transmission blocking interventions. Four adults were inoculated with P. falciparum 3D7-MBE-008 infected erythrocytes and administered piperaquine on days 8 and 10 to clear asexual parasitemia while permitting gametocyte development. On day 25, participants were randomised (1:1) to receive either 0.25 mg/kg primaquine (primaquine group) or no intervention (control group). Transmissibility was assessed by enriched membrane feeding assays on days 25, 29, 32, and 39, with transmission intensity (proportion of mosquitoes infected) determined by 18S qPCR. All participants were infective on day 25, with a median 94% (range, 12-100%) of mosquitoes positive for oocysts, and 76% (range, 8-94%) positive for sporozoites. In the primaquine group, mosquito infectivity decreased substantially between days 25 and 29. In the control group, mosquito infectivity remained high up to day 32, and persisted to day 39 in one participant. The P. falciparum 3D7-MBE-008 parasite bank induced blood-stage infections that were highly transmissible to mosquitoes and is therefore suitable for evaluating transmission blocking interventions.Trial registration anzctr.org.au (registration number: ACTRN12622001097730), registered 08/08/2022.
Collapse
Affiliation(s)
- Sean A Lynch
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | | | - Jenny M Peters
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
| | - Juanita M Heunis
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
| | - Jeremy S E Gower
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Adam J Potter
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
| | - Rebecca Webster
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
| | - Helen Jennings
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
| | - Susan Mathison
- University of the Sunshine Coast Clinical Trials, Brisbane, QLD, Australia
| | - Nischal Sahai
- University of the Sunshine Coast Clinical Trials, Brisbane, QLD, Australia
| | - Fiona H Amante
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia
| | - Bridget E Barber
- QIMR Berghofer, 300 Herston Rd, Herston, Brisbane, QLD, 4006, Australia.
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
- University of the Sunshine Coast Clinical Trials, Brisbane, QLD, Australia.
| |
Collapse
|
4
|
Okombo J, Fidock DA. Towards next-generation treatment options to combat Plasmodium falciparum malaria. Nat Rev Microbiol 2025; 23:178-191. [PMID: 39367132 PMCID: PMC11832322 DOI: 10.1038/s41579-024-01099-x] [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] [Accepted: 08/23/2024] [Indexed: 10/06/2024]
Abstract
Malaria, which is caused by infection of red blood cells with Plasmodium parasites, can be fatal in non-immune individuals if left untreated. The recent approval of the pre-erythrocytic vaccines RTS, S/AS01 and R21/Matrix-M has ushered in hope of substantial reductions in mortality rates, especially when combined with other existing interventions. However, the efficacy of these vaccines is partial, and chemotherapy remains central to malaria treatment and control. For many antimalarial drugs, clinical efficacy has been compromised by the emergence of drug-resistant Plasmodium falciparum strains. Therefore, there is an urgent need for new antimalarial medicines to complement the existing first-line artemisinin-based combination therapies. In this Review, we discuss various opportunities to expand the present malaria treatment space, appraise the current antimalarial drug development pipeline and highlight examples of promising targets. We also discuss other approaches to circumvent antimalarial resistance and how potency against drug-resistant parasites could be retained.
Collapse
Affiliation(s)
- John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
5
|
Adam A, Besson D, Bryant R, Rees S, Willis PA, Burrows JN, Hooft van Huisjduijnen R, Laleu B, Norton L, Canan S, Hawryluk N, Robinson D, Palmer M, Samby KK. Global Health Priority Box─Proactive Pandemic Preparedness. ACS Infect Dis 2024; 10:4030-4039. [PMID: 39488746 DOI: 10.1021/acsinfecdis.4c00700] [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: 11/04/2024]
Abstract
The coronavirus pandemic outbreak of 2019 highlighted the critical importance of preparedness for current and future public health threats (https://www.mmv.org/mmv-open/global-health-priority-box/about-global-health-priority-box). While the main attention for the past few years has been on COVID-19 research, this focus has reduced global resources on research in other areas, including malaria and neglected tropical diseases (NTDs). Such a shift in focus puts at risk the hard-earned progress in global health achieved over the past two decades (https://www.who.int/news-room/spotlight/10-global-health-issues-to-track-in-2021). To address the urgent need for new drugs to combat drug-resistant malaria, emerging zoonotic diseases, and vector control, Medicines for Malaria Venture (MMV) and Innovative Vector Control Consortium (IVCC) assembled a collection of 240 compounds and, in August 2022, launched the Global Health Priority Box (GHPB). This collection of compounds has confirmed activity against emerging pathogens or vectors and is available free of charge. This valuable tool enables researchers worldwide to build on each other's work and save precious time and resources by providing a starting point for the further development of treatments and insecticides. Furthermore, this open access box aligns with two of the many priorities outlined by the World Health Organization (WHO) (https://www.who.int/news-room/spotlight/10-global-health-issues-to-track-in-2021).
Collapse
Affiliation(s)
- Anna Adam
- MMV Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | | | - Rob Bryant
- Brychem/Agranova, BR6 9AP Kent, United Kingdom
| | - Sarah Rees
- Innovative Vector Control Consortium, L3 5QA Liverpool, United Kingdom
| | - Paul A Willis
- MMV Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | | | | | - Benoît Laleu
- MMV Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | - Larry Norton
- Innovative Vector Control Consortium, L3 5QA Liverpool, United Kingdom
| | - Stacie Canan
- Bristol Myers Squibb, California 92121, San Diego, United States of America
| | - Natalie Hawryluk
- Bristol Myers Squibb, California 92121, San Diego, United States of America
| | - Dale Robinson
- Bristol Myers Squibb, California 92121, San Diego, United States of America
| | - Mike Palmer
- MMV Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | | |
Collapse
|
6
|
Sakura T, Ishii R, Yoshida E, Kita K, Kato T, Inaoka DK. Accelerating Antimalarial Drug Discovery with a New High-Throughput Screen for Fast-Killing Compounds. ACS Infect Dis 2024; 10:4115-4126. [PMID: 39561299 DOI: 10.1021/acsinfecdis.4c00328] [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: 11/21/2024]
Abstract
The urgent need for rapidly acting compounds in the development of antimalarial drugs underscores the significance of such compounds in overcoming resistance issues and improving patient adherence to antimalarial treatments. The present study introduces a high-throughput screening (HTS) approach using 1536-well plates, employing Plasmodium falciparum lactate dehydrogenase (PfLDH) combined with nitroreductase (NTR) and fluorescent probes to evaluate inhibition of the growth of the asexual blood stage of malaria parasites. This method was adapted to efficiently assess the speed of action profiling (SAP) in a 384-well plate format, streamlining the traditionally time-consuming screening process. By successfully screening numerous compounds, this approach identified fast-killing hits early in the screening process, addressing challenges associated with artemisinin-based combination therapies. The high-throughput SAP method is expected to be of value in continuously monitoring fast-killing properties during structure-activity relationship studies, expediting the identification and development of novel, rapidly acting antimalarial drugs within phenotypic drug discovery campaigns.
Collapse
Affiliation(s)
- Takaya Sakura
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
| | - Ryuta Ishii
- Department of Cellular Architecture Studies, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Eri Yoshida
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
- Department of Infection Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Teruhisa Kato
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Exploratory Research for Drug Discovery, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
| | - Daniel Ken Inaoka
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
- Department of Infection Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
7
|
Woolley SD, Grigg MJ, Marquart L, Gower JSE, Piera K, Nair AS, Amante FM, Rajahram GS, William T, Frazer DM, Chalon S, McCarthy JS, Anstey NM, Barber BE. Longitudinal changes in iron homeostasis in human experimental and clinical malaria. EBioMedicine 2024; 105:105189. [PMID: 38851058 PMCID: PMC11200279 DOI: 10.1016/j.ebiom.2024.105189] [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: 11/29/2023] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND The interaction between iron status and malaria is incompletely understood. We evaluated longitudinal changes in iron homeostasis in volunteers enrolled in malaria volunteer infection studies (VIS) and in Malaysian patients with falciparum and vivax malaria. METHODS We retrieved data and samples from 55 participants (19 female) enrolled in malaria VIS, and 171 patients (45 female) with malaria and 30 healthy controls (13 female) enrolled in clinical studies in Malaysia. Ferritin, hepcidin, erythropoietin, and soluble transferrin receptor (sTfR) were measured by ELISA. FINDINGS In the VIS, participants' parasitaemia was correlated with baseline mean corpuscular volume (MCV), but not iron status (ferritin, hepcidin or sTfR). Ferritin, hepcidin and sTfR all increased during the VIS. Ferritin and hepcidin normalised by day 28, while sTfR remained elevated. In VIS participants, baseline ferritin was associated with post-treatment increases in liver transaminase levels. In Malaysian patients with malaria, hepcidin and ferritin were elevated on admission compared to healthy controls, while sTfR increased following admission. By day 28, hepcidin had normalised; however, ferritin and sTfR both remained elevated. INTERPRETATION Our findings demonstrate that parasitaemia is associated with an individual's MCV rather than iron status. The persistent elevation in sTfR 4 weeks post-infection in both malaria VIS and clinical malaria may reflect a causal link between malaria and iron deficiency. FUNDING National Health and Medical Research Council (Program Grant 1037304, Project Grants 1045156 and 1156809; Investigator Grants 2016792 to BEB, 2016396 to JCM, 2017436 to MJG); US National Institute of Health (R01-AI116472-03); Malaysian Ministry of Health (BP00500420).
Collapse
Affiliation(s)
- Stephen D Woolley
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia; Clinical Sciences Department, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Academic Department of Military Medicine, Royal Centre for Defence Medicine, Birmingham, United Kingdom
| | - Matthew J Grigg
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia; Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Louise Marquart
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Public Health, University of Queensland, Brisbane, Australia
| | - Jeremy S E Gower
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kim Piera
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
| | - Arya Sheela Nair
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Fiona M Amante
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Giri S Rajahram
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia; Department of Medicine, Queen Elizabeth II Hospital, Kota Kinabalu, Malaysia; Clinical Research Centre, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia
| | - Timothy William
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia; Clinical Research Centre, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia; Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - David M Frazer
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - James S McCarthy
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia; Victorian Infectious Diseases Institute, Peter Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia; Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Bridget E Barber
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia; Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia; Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia; Infectious Diseases Department, Royal Brisbane and Women's Hospital, Brisbane, Australia.
| |
Collapse
|
8
|
Barber BE, Webster R, Potter AJ, Llewellyn S, Sahai N, Leelasena I, Mathison S, Kuritz K, Flynn J, Chalon S, Marrast AC, Gobeau N, Moehrle JJ. Characterising the blood-stage antimalarial activity of pyronaridine in healthy volunteers experimentally infected with Plasmodium falciparum. Int J Antimicrob Agents 2024; 64:107196. [PMID: 38734217 DOI: 10.1016/j.ijantimicag.2024.107196] [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: 11/11/2023] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
With the spread of artemisinin resistance throughout Southeast Asia and now in Africa, the antimalarial drug pyronaridine is likely to become an increasingly important component of new antimalarial drug regimens. However, the antimalarial activity of pyronaridine in humans has not been completely characterised. This volunteer infection study aimed to determine the pharmacokinetic/pharmacodynamic (PK/PD) relationship of pyronaridine in malaria naïve adults. Volunteers were inoculated with Plasmodium falciparum-infected erythrocytes on day 0 and administered different single oral doses of pyronaridine on day 8. Parasitaemia and concentrations of pyronaridine were measured and standard safety assessments performed. Curative artemether-lumefantrine therapy was administered if parasite regrowth occurred, or on day 47 ± 2. Outcomes were parasite clearance kinetics, PK and PK/PD parameters from modelling. Ten participants were inoculated and administered 360 mg (n = 4), 540 mg (n = 4) or 720 mg (n = 1) pyronaridine. One participant was withdrawn without receiving pyronaridine. The time to maximum pyronaridine concentration was 1-2 h, the elimination half-life was 8-9 d, and the parasite clearance half-life was approximately 5 h. Parasite regrowth occurred with 360 mg (4/4 participants) and 540 mg (2/4 participants). Key efficacy parameters including the minimum inhibitory concentration (5.5 ng/mL) and minimum parasiticidal concentration leading to 90% of maximum effect (MPC90: 8 ng/mL) were derived from the PK/PD model. Adverse events considered related to pyronaridine were predominantly mild to moderate gastrointestinal symptoms. There were no serious adverse events. Data obtained in this study will support the use of pyronaridine in new antimalarial combination therapies by informing partner drug selection and dosing considerations.
Collapse
Affiliation(s)
- Bridget E Barber
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; University of the Sunshine Coast Clinical Trials, Morayfield, QLD, Australia; Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Rebecca Webster
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Adam J Potter
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Stacey Llewellyn
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Nischal Sahai
- University of the Sunshine Coast Clinical Trials, Morayfield, QLD, Australia
| | - Indika Leelasena
- University of the Sunshine Coast Clinical Trials, Morayfield, QLD, Australia
| | - Susan Mathison
- University of the Sunshine Coast Clinical Trials, Morayfield, QLD, Australia
| | | | - Julia Flynn
- Medicines for Malaria Venture, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
9
|
Achan J, Barry A, Leroy D, Kamara G, Duparc S, Kaszubska W, Gandhi P, Buffet B, Tshilab P, Ogutu B, Taylor T, Krishna S, Richardson N, Ramachandruni H, Rietveld H. Defining the next generation of severe malaria treatment: a target product profile. Malar J 2024; 23:174. [PMID: 38835069 DOI: 10.1186/s12936-024-04986-z] [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: 02/09/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Severe malaria is a life-threatening infection, particularly affecting children under the age of 5 years in Africa. Current treatment with parenteral artemisinin derivatives is highly efficacious. However, artemisinin partial resistance is widespread in Southeast Asia, resulting in delayed parasite clearance after therapy, and has emerged independently in South America, Oceania, and Africa. Hence, new treatments for severe malaria are needed, and it is prudent to define their characteristics now. This manuscript focuses on the target product profile (TPP) for new treatments for severe malaria. It also highlights preparedness when considering ways of protecting the utility of artemisinin-based therapies. TARGET PRODUCT PROFILE Severe malaria treatments must be highly potent, with rapid onset of antiparasitic activity to clear the infection as quickly as possible to prevent complications. They should also have a low potential for drug resistance selection, given the high parasite burden in patients with severe malaria. Combination therapies are needed to deter resistance selection and dissemination. Partner drugs which are approved for uncomplicated malaria treatment would provide the most rapid development pathway for combinations, though new candidate molecules should be considered. Artemisinin combination approaches to severe malaria would extend the lifespan of current therapy, but ideally, completely novel, non-artemisinin-based combination therapies for severe malaria should be developed. These should be advanced to at least phase 2 clinical trials, enabling rapid progression to patient use should current treatment fail clinically. New drug combinations for severe malaria should be available as injectable formulations for rapid and effective treatment, or as rectal formulations for pre-referral intervention in resource-limited settings. CONCLUSION Defining the TPP is a key step to align responses across the community to proactively address the potential for clinical failure of artesunate in severe malaria. In the shorter term, artemisinin-based combination therapies should be developed using approved or novel drugs. In the longer term, novel combination treatments should be pursued. Thus, this TPP aims to direct efforts to preserve the efficacy of existing treatments while improving care and outcomes for individuals affected by this life-threatening disease.
Collapse
Affiliation(s)
| | - Aïssata Barry
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Didier Leroy
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | - George Kamara
- Médecins Sans Frontières, Magburaka District Hospital, Freetown, Sierra Leone
| | - Stephan Duparc
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | - Wiweka Kaszubska
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | | | - Bénédicte Buffet
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland
| | | | - Bernhards Ogutu
- Centre for Clinical Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Terrie Taylor
- Queen Elizabeth Central Hospital and Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Sanjeev Krishna
- Institut Für Tropenmedizin, Eberhard Karls Universität Tübingen, and German Center for Infection Research (Dzif), Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
- Clinical Academic Group, Institute for Infection and Immunity, St. George's University of London, London, UK
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Hanu Ramachandruni
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland.
| | - Hans Rietveld
- Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, CH-1215, Geneva 15, Switzerland.
| |
Collapse
|
10
|
Elias KM, Khan SR, Stadler E, Schlub TE, Cromer D, Polizzotto MN, Kent SJ, Turner T, Davenport MP, Khoury DS. Viral clearance as a surrogate of clinical efficacy for COVID-19 therapies in outpatients: a systematic review and meta-analysis. THE LANCET. MICROBE 2024; 5:e459-e467. [PMID: 38583464 DOI: 10.1016/s2666-5247(23)00398-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 04/09/2024]
Abstract
BACKGROUND Surrogates of antiviral efficacy are needed for COVID-19. We aimed to investigate the relationship between the virological effect of treatment and clinical efficacy as measured by progression to severe disease in outpatients treated for mild-to-moderate COVID-19. METHODS In this systematic review and meta-analysis, we searched PubMed, Scopus, and medRxiv from database inception to Aug 16, 2023, for randomised placebo-controlled trials that tested virus-directed treatments (ie, any monoclonal antibodies, convalescent plasma, or antivirals) in non-hospitalised individuals with COVID-19. We only included studies that reported both clinical outcomes (ie, rate of disease progression to hospitalisation or death) and virological outcomes (ie, viral load within the first 7 days of treatment). We extracted summary data from eligible reports, with discrepancies resolved through discussion. We used an established meta-regression model with random effects to assess the association between clinical efficacy and virological treatment effect, and calculated I2 to quantify residual study heterogeneity. FINDINGS We identified 1718 unique studies, of which 22 (with a total of 16 684 participants) met the inclusion criteria, and were in primarily unvaccinated individuals. Risk of bias was assessed as low in 19 of 22 studies for clinical outcomes, whereas for virological outcomes, a high risk of bias was assessed in 11 studies, some risk in ten studies, and a low risk in one study. The unadjusted relative risk of disease progression for each extra log10 copies per mL reduction in viral load in treated compared with placebo groups was 0·12 (95% CI 0·04-0·34; p<0·0001) on day 3, 0·20 (0·08-0·50; p=0·0006) on day 5, and 0·53 (0·30-0·94; p=0·030) on day 7. The residual heterogeneity in our meta-regression was estimated as low (I2=0% [0-53] on day 3, 0% [0-71] on day 5, and 0% [0-43] on day 7). INTERPRETATION Despite the aggregation of studies with differing designs, and evidence of risk of bias in some virological outcomes, this review provides evidence that treatment-induced acceleration of viral clearance within the first 5 days after treatment is a potential surrogate of clinical efficacy to prevent hospitalisation with COVID-19. This work supports the use of viral clearance as an early phase clinical trial endpoint of therapeutic efficacy. FUNDING Australian Government Department of Health, Medical Research Future Fund, and Australian National Health and Medical Research Council.
Collapse
Affiliation(s)
- Karen M Elias
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Shanchita R Khan
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Eva Stadler
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Timothy E Schlub
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia; Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Mark N Polizzotto
- Clinical Hub for Interventional Research and John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Canberra, ACT, Australia; Canberra Regional Cancer Centre, The Canberra Hospital, Canberra, ACT, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tari Turner
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
11
|
Lindblom JR, Zhang X, Lehane AM. A pH Fingerprint Assay to Identify Inhibitors of Multiple Validated and Potential Antimalarial Drug Targets. ACS Infect Dis 2024; 10:1185-1200. [PMID: 38499199 PMCID: PMC11019546 DOI: 10.1021/acsinfecdis.3c00588] [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: 10/31/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 03/20/2024]
Abstract
New drugs with novel modes of action are needed to safeguard malaria treatment. In recent years, millions of compounds have been tested for their ability to inhibit the growth of asexual blood-stage Plasmodium falciparum parasites, resulting in the identification of thousands of compounds with antiplasmodial activity. Determining the mechanisms of action of antiplasmodial compounds informs their further development, but remains challenging. A relatively high proportion of compounds identified as killing asexual blood-stage parasites show evidence of targeting the parasite's plasma membrane Na+-extruding, H+-importing pump, PfATP4. Inhibitors of PfATP4 give rise to characteristic changes in the parasite's internal [Na+] and pH. Here, we designed a "pH fingerprint" assay that robustly identifies PfATP4 inhibitors while simultaneously allowing the detection of (and discrimination between) inhibitors of the lactate:H+ transporter PfFNT, which is a validated antimalarial drug target, and the V-type H+ ATPase, which was suggested as a possible target of the clinical candidate ZY19489. In our pH fingerprint assays and subsequent secondary assays, ZY19489 did not show evidence for the inhibition of pH regulation by the V-type H+ ATPase, suggesting that it has a different mode of action in the parasite. The pH fingerprint assay also has the potential to identify protonophores, inhibitors of the acid-loading Cl- transporter(s) (for which the molecular identity(ies) remain elusive), and compounds that act through inhibition of either the glucose transporter PfHT or glycolysis. The pH fingerprint assay therefore provides an efficient starting point to match a proportion of antiplasmodial compounds with their mechanisms of action.
Collapse
Affiliation(s)
| | | | - Adele M. Lehane
- Research School of Biology, Australian National University, Canberra, Australian Capital
Territory 2600, Australia
| |
Collapse
|
12
|
Schäfer TM, Pessanha de Carvalho L, Inoue J, Kreidenweiss A, Held J. The problem of antimalarial resistance and its implications for drug discovery. Expert Opin Drug Discov 2024; 19:209-224. [PMID: 38108082 DOI: 10.1080/17460441.2023.2284820] [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: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Malaria remains a devastating infectious disease with hundreds of thousands of casualties each year. Antimalarial drug resistance has been a threat to malaria control and elimination for many decades and is still of concern today. Despite the continued effectiveness of current first-line treatments, namely artemisinin-based combination therapies, the emergence of drug-resistant parasites in Southeast Asia and even more alarmingly the occurrence of resistance mutations in Africa is of great concern and requires immediate attention. AREAS COVERED A comprehensive overview of the mechanisms underlying the acquisition of drug resistance in Plasmodium falciparum is given. Understanding these processes provides valuable insights that can be harnessed for the development and selection of novel antimalarials with reduced resistance potential. Additionally, strategies to mitigate resistance to antimalarial compounds on the short term by using approved drugs are discussed. EXPERT OPINION While employing strategies that utilize already approved drugs may offer a prompt and cost-effective approach to counter antimalarial drug resistance, it is crucial to recognize that only continuous efforts into the development of novel antimalarial drugs can ensure the successful treatment of malaria in the future. Incorporating resistance propensity assessment during this developmental process will increase the likelihood of effective and enduring malaria treatments.
Collapse
Affiliation(s)
| | | | - Juliana Inoue
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
13
|
Woolley SD, Grigg MJ, Marquart L, Gower J, Piera K, Nair AS, Amante FM, Rajahram GS, William T, Frazer DM, Chalon S, McCarthy JS, Anstey NM, Barber BE. Longitudinal changes in iron homeostasis in human experimental and clinical malaria. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.19.23300265. [PMID: 38196596 PMCID: PMC10775340 DOI: 10.1101/2023.12.19.23300265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Background The interaction between iron deficiency and malaria is incompletely understood. We evaluated longitudinal changes in iron homeostasis in volunteers enrolled in malaria volunteer infection studies (VIS) and in Malaysian patients with falciparum and vivax malaria. Methods We retrieved samples and associated data from 55 participants enrolled in malaria VIS, and 171 malaria patients and 30 healthy controls enrolled in clinical studies in Malaysia. Ferritin, hepcidin, erythropoietin, and soluble transferrin receptor (sTfR) were measured by ELISA. Results In the VIS, participants' parasitaemia was correlated with baseline mean corpuscular volume (MCV), but not iron status (ferritin, hepcidin or sTfR). Ferritin, hepcidin and sTfR all increased during the VIS. Ferritin and hepcidin normalised by day 28, while sTfR remained elevated. In VIS participants, baseline iron status (ferritin) was associated with post-treatment increases in liver transaminase levels. In Malaysian malaria patients, hepcidin and ferritin were elevated on admission compared to healthy controls, while sTfR increased following admission. Hepcidin normalised by day 28; however, ferritin and sTfR both remained elevated 4 weeks following admission. Conclusion Our findings demonstrate that parasitaemia is associated with an individual's MCV rather than iron status. The persistent elevation in sTfR 4 weeks post-infection in both malaria VIS and clinical malaria may reflect a causal link between malaria and iron deficiency.
Collapse
Affiliation(s)
- Stephen D Woolley
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Clinical Sciences Department, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Academic Department of Military Medicine, Royal Centre for Defence Medicine, Birmingham, United Kingdom
| | - Matthew J Grigg
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Louise Marquart
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- School of Public Health, University of Queensland, Brisbane, Australia
| | - Jeremy Gower
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kim Piera
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
| | - Arya Sheela Nair
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Fiona M Amante
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Giri S Rajahram
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
- Department of Medicine, Queen Elizabeth II Hospital, Kota Kinabalu, Malaysia
- Clinical Research Centre, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia
| | - Timothy William
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
- Department of Medicine, Queen Elizabeth II Hospital, Kota Kinabalu, Malaysia
- Subang Jaya Medical Centre, Subang Jaya, Malaysia
| | - David M Frazer
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - James S McCarthy
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Victorian Infectious Diseases Institute, Peter Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
| | - Bridget E Barber
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Malaysia
- Infectious Diseases Department, Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
14
|
Siqueira-Neto JL, Wicht KJ, Chibale K, Burrows JN, Fidock DA, Winzeler EA. Antimalarial drug discovery: progress and approaches. Nat Rev Drug Discov 2023; 22:807-826. [PMID: 37652975 PMCID: PMC10543600 DOI: 10.1038/s41573-023-00772-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 09/02/2023]
Abstract
Recent antimalarial drug discovery has been a race to produce new medicines that overcome emerging drug resistance, whilst considering safety and improving dosing convenience. Discovery efforts have yielded a variety of new molecules, many with novel modes of action, and the most advanced are in late-stage clinical development. These discoveries have led to a deeper understanding of how antimalarial drugs act, the identification of a new generation of drug targets, and multiple structure-based chemistry initiatives. The limited pool of funding means it is vital to prioritize new drug candidates. They should exhibit high potency, a low propensity for resistance, a pharmacokinetic profile that favours infrequent dosing, low cost, preclinical results that demonstrate safety and tolerability in women and infants, and preferably the ability to block Plasmodium transmission to Anopheles mosquito vectors. In this Review, we describe the approaches that have been successful, progress in preclinical and clinical development, and existing challenges. We illustrate how antimalarial drug discovery can serve as a model for drug discovery in diseases of poverty.
Collapse
Affiliation(s)
| | - Kathryn J Wicht
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | - Kelly Chibale
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | | | - David A Fidock
- Department of Microbiology and Immunology and Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
15
|
Alaithan H, Kumar N, Islam MZ, Liappis AP, Nava VE. Novel Therapeutics for Malaria. Pharmaceutics 2023; 15:1800. [PMID: 37513987 PMCID: PMC10383744 DOI: 10.3390/pharmaceutics15071800] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Malaria is a potentially fatal disease caused by protozoan parasites of the genus Plasmodium. It is responsible for significant morbidity and mortality in endemic countries of the tropical and subtropical world, particularly in Africa, Southeast Asia, and South America. It is estimated that 247 million malaria cases and 619,000 deaths occurred in 2021 alone. The World Health Organization's (WHO) global initiative aims to reduce the burden of disease but has been massively challenged by the emergence of parasitic strains resistant to traditional and emerging antimalarial therapy. Therefore, development of new antimalarial drugs with novel mechanisms of action that overcome resistance in a safe and efficacious manner is urgently needed. Based on the evolving understanding of the physiology of Plasmodium, identification of potential targets for drug intervention has been made in recent years, resulting in more than 10 unique potential anti-malaria drugs added to the pipeline for clinical development. This review article will focus on current therapies as well as novel targets and therapeutics against malaria.
Collapse
Affiliation(s)
- Haitham Alaithan
- Veterans Affairs Medical Center, Washington, DC 20422, USA
- Department of Medicine, George Washington University, Washington, DC 20037, USA
| | - Nirbhay Kumar
- Department of Global Health, Milken Institute of Public Health, George Washington University, Washington, DC 20037, USA
| | - Mohammad Z Islam
- Department of Pathology and Translational Pathology, Louisiana State University Health Science Center, Shreveport, LA 71103, USA
| | - Angelike P Liappis
- Veterans Affairs Medical Center, Washington, DC 20422, USA
- Department of Medicine, George Washington University, Washington, DC 20037, USA
| | - Victor E Nava
- Veterans Affairs Medical Center, Washington, DC 20422, USA
- Department of Pathology, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
16
|
Umumararungu T, Nkuranga JB, Habarurema G, Nyandwi JB, Mukazayire MJ, Mukiza J, Muganga R, Hahirwa I, Mpenda M, Katembezi AN, Olawode EO, Kayitare E, Kayumba PC. Recent developments in antimalarial drug discovery. Bioorg Med Chem 2023; 88-89:117339. [PMID: 37236020 DOI: 10.1016/j.bmc.2023.117339] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Although malaria remains a big burden to many countries that it threatens their socio-economic stability, particularly in the countries where malaria is endemic, there have been great efforts to eradicate this disease with both successes and failures. For example, there has been a great improvement in malaria prevention and treatment methods with a net reduction in infection and mortality rates. However, the disease remains a global threat in terms of the number of people affected because it is one of the infectious diseases that has the highest prevalence rate, especially in Africa where the deadly Plasmodium falciparum is still widely spread. Methods to fight malaria are being diversified, including the use of mosquito nets, the target candidate profiles (TCPs) and target product profiles (TPPs) of medicine for malarial venture (MMV) strategy, the search for newer and potent drugs that could reverse chloroquine resistance, and the use of adjuvants such as rosiglitazone and sevuparin. Although these adjuvants have no antiplasmodial activity, they can help to alleviate the effects which result from plasmodium invasion such as cytoadherence. The list of new antimalarial drugs under development is long, including the out of ordinary new drugs MMV048, CDRI-97/78 and INE963 from South Africa, India and Novartis, respectively.
Collapse
Affiliation(s)
- Théoneste Umumararungu
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda.
| | - Jean Bosco Nkuranga
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Gratien Habarurema
- Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Rwanda
| | - Jean Baptiste Nyandwi
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Marie Jeanne Mukazayire
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Janvier Mukiza
- Department of Mathematical Science and Physical Education, School of Education, College of Education, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Raymond Muganga
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Innocent Hahirwa
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Matabishi Mpenda
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Alain Nyirimigabo Katembezi
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda; Rwanda Food and Drugs Authority, Nyarutarama Plaza, KG 9 Avenue, Kigali, Rwanda
| | - Emmanuel Oladayo Olawode
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, 18301 N Miami Ave #1, Miami, FL 33169, USA
| | - Egide Kayitare
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| | - Pierre Claver Kayumba
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Rwanda
| |
Collapse
|
17
|
Kanai M, Hagenah LM, Ashley EA, Chibale K, Fidock DA. Keystone Malaria Symposium 2022: a vibrant discussion of progress made and challenges ahead from drug discovery to treatment. Trends Parasitol 2022; 38:711-718. [PMID: 35864072 PMCID: PMC9631389 DOI: 10.1016/j.pt.2022.06.005] [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: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
In recent years, the field of malaria research has made substantial progress in the areas of antimalarial drug resistance and discovery. These efforts are essential to combatting the devastating impact of malaria, which, in 2020, resulted in an estimated 241 million cases and 627 000 deaths. Recent advances in this area were presented at a Keystone Symposium entitled ‘Malaria: Confronting Challenges from Drug Discovery to Treatment’, held in person in Breckenridge, Colorado, in April 2022. Herein, we present a summary of the proceedings of this vibrant scientific exchange, which brought together a superb group of faculty, postdocs, and students from around the globe.
Collapse
Affiliation(s)
- Mariko Kanai
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Laura M Hagenah
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Elizabeth A Ashley
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Mahosot Hospital, Vientiane, Laos.
| | - Kelly Chibale
- Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, South Africa; Department of Chemistry, University of Cape Town, Rondebosch, South Africa.
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
18
|
Elemental Analysis, Phytochemical Screening and Evaluation of Antioxidant, Antibacterial and Anticancer Activity of Pleurotus ostreatus through In Vitro and In Silico Approaches. Metabolites 2022; 12:metabo12090821. [PMID: 36144225 PMCID: PMC9502197 DOI: 10.3390/metabo12090821] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/24/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
Oyster mushrooms form an integral part of many diets owing to their characteristic aroma, delicious taste and nutraceutical value. In this study, we examined oyster mushrooms by direct arc optical emission spectroscopy for the presence of various biologically important elements. Furthermore, we screened phytochemicals present in Pleurotus ostreatus by applying GC-MS. Additionally, the antioxidant, antibacterial and anticancer activities of the ethanolic extract of Pleurotus ostreatus were studied. Moreover, we docked the phytochemicals and examined their binding affinities with EGFR, PR and NF-κB proteins, which are overexpressed in breast cancer. The elemental analysis showed the presence of Fe, K, Na, Ca, Mg, Cr and Sr in the spectrum. Moreover, GC-MS data revealed the presence of 32 biologically active compounds in oyster mushrooms. The ethanolic extract displayed remarkable free radical scavenging activity (~50%) against DPPH. The mushroom has shown promising antibacterial activity against both Gram-positive (S. aureus) and Gram-negative bacteria (Pseudomonasaeruginosa, Proteus vulgaris and Proteus mirabilis). The present study also revealed that oyster mushrooms possess significant anticancer activity. The ethanolic extract inhibited the growth and proliferation of MCF-7 cells. It also induced cell shrinkage, membrane blebbing and nuclear fragmentation, resulting in apoptosis of malignant cells. The molecular docking analysis showed that ligand 15 (Linoleic acid ethyl ester), ligand 27 (Ergosta-5,7,9(11),22-tetraen-3-ol, (3. beta.,22E), ligand 28 (Stigmasta-5,22-dien-3-ol, acetate, (3. beta.,22Z), ligand 30 (Ergosta-5,7,22-Trien-3-Ol, (3. Beta.,22E) and ligand 32 (gamma. Sitosterol) exhibited better binding affinities with EGFR, PR and NF-κB proteins. This result provides a strong ground for confirmation of the in vitro anticancer effect of Pleurotus ostreatus. From the present in vitro and in silico studies, it can be concluded that Pleurotus ostreatus is a useful source of essential elements and reservoir of bioactive compounds which confer its significant antioxidant, antibacterial and anticancer properties.
Collapse
|
19
|
Daubenberger C, Burrows JN. Volunteer infection studies accelerate the clinical development of novel drugs against malaria. THE LANCET. INFECTIOUS DISEASES 2022; 22:753-754. [PMID: 35247323 DOI: 10.1016/s1473-3099(21)00722-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 06/14/2023]
Affiliation(s)
- Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | | |
Collapse
|
20
|
Radohery GFR, Gower J, Barber BE, Kansagra K, Möhrle JJ, Davenport MP, McCarthy JS, Khoury DS, Rebelo M. Effect of novel antimalarial ZY-19489 on Plasmodium falciparum viability in a volunteer infection study. THE LANCET. INFECTIOUS DISEASES 2022; 22:760-761. [PMID: 35643094 DOI: 10.1016/s1473-3099(22)00294-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Affiliation(s)
| | - Jeremy Gower
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Bridget E Barber
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | | | - Miles P Davenport
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and the Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Maria Rebelo
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Instituto de Medicina Molecular Joao Lobo Antunes, Lisbon 1649-028, Portugal.
| |
Collapse
|