1
|
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: 0] [Impact Index Per Article: 0] [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
|
2
|
Mogaka S, Molu H, Kagasi E, Ogila K, Waihenya R, Onditi F, Ozwara H. Senna occidentalis (L.) Link root extract inhibits Plasmodium growth in vitro and in mice. BMC Complement Med Ther 2023; 23:71. [PMID: 36879244 PMCID: PMC9987147 DOI: 10.1186/s12906-023-03854-8] [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: 08/12/2022] [Accepted: 01/20/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Senna occidentalis (L.) Link has been used worldwide in traditional treatment of many diseases and conditions including snakebite. In Kenya, a decoction from the plant roots taken orally, is used as a cure for malaria. Several studies have demonstrated that extracts from the plant possess antiplasmodial activity, in vitro. However, the safety and curative potency of the plant root against established malaria infection is yet to be scientifically validated, in vivo. On the other hand, there are reports on variation in bioactivity of extracts obtained from this plant species, depending on the plant part used and place of origin among other factors. In this study, we demonstrated the antiplasmodial activity of Senna occidentalis roots extract in vitro, and in mice. METHODS Methanol, ethyl acetate, chloroform, hexane and water extracts of S. occidentalis root were tested for in vitro antiplasmodial activity against Plasmodium falciparum, strain 3D7. Cytotoxicity of the most active solvent extracts was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the curative potency in Plasmodium berghei infected mice evaluated by Rane's test. RESULTS All of the solvent extracts tested in this study inhibited the propagation of P. falciparum, strain 3D7, in vitro, with polar extracts being more active than non-polar ones. Methanolic extracts had the highest activity (IC50 = 1.76) while hexane extract displayed the lowest activity (IC50 = 18.47). At the tested concentrations, methanolic and aqueous extracts exhibited high selectivity index against P. falciparum strain 3D7 (SI > 10) in the cytotoxicity assay. Further, the extracts significantly suppressed the propagation of P. berghei parasites (P < 0.05) in vivo and increased the survival time of the infected mice (P < 0.0001). CONCLUSIONS Senna occidentalis (L.) Link root extract inhibits the propagation of malaria parasites in vitro and in BALB/c mice.
Collapse
Affiliation(s)
- Simeon Mogaka
- Department of Tropical and Infectious Diseases, Institute of Primate Research, P.O Box 24481, Karen, Nairobi, 00502, Kenya.
- Department of Zoology, Jomo Kenyatta University of Agriculture and Technology, P.O Box 62000-00200, Nairobi, Kenya.
| | - Halkano Molu
- Department of Tropical and Infectious Diseases, Institute of Primate Research, P.O Box 24481, Karen, Nairobi, 00502, Kenya
| | - Esther Kagasi
- Department of Tropical and Infectious Diseases, Institute of Primate Research, P.O Box 24481, Karen, Nairobi, 00502, Kenya
| | - Kenneth Ogila
- Department of Zoology, Jomo Kenyatta University of Agriculture and Technology, P.O Box 62000-00200, Nairobi, Kenya
| | - Rebeccah Waihenya
- Department of Zoology, Jomo Kenyatta University of Agriculture and Technology, P.O Box 62000-00200, Nairobi, Kenya
| | - Faith Onditi
- Department of Tropical and Infectious Diseases, Institute of Primate Research, P.O Box 24481, Karen, Nairobi, 00502, Kenya
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hastings Ozwara
- Department of Tropical and Infectious Diseases, Institute of Primate Research, P.O Box 24481, Karen, Nairobi, 00502, Kenya
| |
Collapse
|
3
|
Webster R, Mitchell H, Peters JM, Heunis J, O'Neill B, Gower J, Lynch S, Jennings H, Amante FH, Llewellyn S, Marquart L, Potter AJ, Birrell GW, Edstein MD, Shanks GD, McCarthy JS, Barber BE. Transmission Blocking Activity of Low-dose Tafenoquine in Healthy Volunteers Experimentally Infected With Plasmodium falciparum. Clin Infect Dis 2023; 76:506-512. [PMID: 35731843 DOI: 10.1093/cid/ciac503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Blocking the transmission of parasites from humans to mosquitoes is a key component of malaria control. Tafenoquine exhibits activity against all stages of the malaria parasite and may have utility as a transmission blocking agent. We aimed to characterize the transmission blocking activity of low-dose tafenoquine. METHODS Healthy adults were inoculated with Plasmodium falciparum 3D7-infected erythrocytes on day 0. Piperaquine was administered on days 9 and 11 to clear asexual parasitemia while allowing gametocyte development. A single 50-mg oral dose of tafenoquine was administered on day 25. Transmission was determined by enriched membrane feeding assays predose and at 1, 4, and 7 days postdose. Artemether-lumefantrine was administered following the final assay. Outcomes were the reduction in mosquito infection and gametocytemia after tafenoquine and safety parameters. RESULTS Six participants were enrolled, and all were infective to mosquitoes before tafenoquine, with a median 86% (range, 22-98) of mosquitoes positive for oocysts and 57% (range, 4-92) positive for sporozoites. By day 4 after tafenoquine, the oocyst and sporozoite positivity rate had reduced by a median 35% (interquartile range [IQR]: 16-46) and 52% (IQR: 40-62), respectively, and by day 7, 81% (IQR 36-92) and 77% (IQR 52-98), respectively. The decline in gametocyte density after tafenoquine was not significant. No significant participant safety concerns were identified. CONCLUSIONS Low-dose tafenoquine (50 mg) reduces P. falciparum transmission to mosquitoes, with a delay in effect.
Collapse
Affiliation(s)
- Rebecca Webster
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Hayley Mitchell
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jenny M Peters
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Juanita Heunis
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Brighid O'Neill
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jeremy Gower
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sean Lynch
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Helen Jennings
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Fiona H Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | - Adam J Potter
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Geoffrey W Birrell
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Michael D Edstein
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - G Dennis Shanks
- Australian Defence Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | |
Collapse
|
4
|
Methylene Blue-Based Combination Therapy with Amodiaquine Prevents Severe Malaria in an Experimental Rodent Model. Pharmaceutics 2022; 14:pharmaceutics14102031. [PMID: 36297466 PMCID: PMC9611243 DOI: 10.3390/pharmaceutics14102031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Untreated malaria can progress rapidly to severe forms (<24 h). Moreover, resistance to antimalarial drugs is a threat to global efforts to protect people from malaria. Given this, it is clear that new chemotherapy must be developed. We contribute new data about using methylene blue (MB) to cure malaria and cerebral malaria in a combined therapy with common antimalarial drugs, including mefloquine (MQ) and amodiaquine (AQ). A C57BL6/J mouse model was used in an experimental cerebral malaria model. Mice were infected with Plasmodium berghei ANKA on Day 0 (D0) and the treatment started on D3 (nearly 1% parasitaemia) with AQ, MQ or MB alone or in combination with AQ or MQ. AQ, MQ and MB alone were unable to prevent cerebral malaria as part of a late chemotherapy. MB-based combination therapies were efficient even if treatment began at a late stage. We found a significant difference in survival rate (p < 0.0001) between MBAQ and the untreated group, but also with the AQ (p = 0.0024) and MB groups (p < 0.0001). All the infected mice treated with MB in combination with AQ were protected from cerebral malaria. Partial protection was demonstrated with MB associated with MQ. In this group, a significant difference was found between MBMQ and the untreated group (p < 0.0001), MQ (p = 0.0079) and MB (p = 0.0039). MB associated with AQ would be a good candidate for preventing cerebral malaria.
Collapse
|
5
|
McCarthy JS, Yalkinoglu Ö, Odedra A, Webster R, Oeuvray C, Tappert A, Bezuidenhout D, Giddins MJ, Dhingra SK, Fidock DA, Marquart L, Webb L, Yin X, Khandelwal A, Bagchus WM. Safety, pharmacokinetics, and antimalarial activity of the novel plasmodium eukaryotic translation elongation factor 2 inhibitor M5717: a first-in-human, randomised, placebo-controlled, double-blind, single ascending dose study and volunteer infection study. THE LANCET. INFECTIOUS DISEASES 2021; 21:1713-1724. [PMID: 34715032 PMCID: PMC8612936 DOI: 10.1016/s1473-3099(21)00252-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/02/2021] [Accepted: 04/13/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND M5717 is the first plasmodium translation elongation factor 2 inhibitor to reach clinical development as an antimalarial. We aimed to characterise the safety, pharmacokinetics, and antimalarial activity of M5717 in healthy volunteers. METHODS This first-in-human study was a two-part, single-centre clinical trial done in Brisbane, QLD, Australia. Part one was a double-blind, randomised, placebo-controlled, single ascending dose study in which participants were enrolled into one of nine dose cohorts (50, 100, 200, 400, 600, 1000, 1250, 1800, or 2100 mg) and randomly assigned (3:1) to M5717 or placebo. A sentinel dosing strategy was used for each dose cohort whereby two participants (one assigned to M5717 and one assigned to placebo) were initially randomised and dosed. Randomisation schedules were generated electronically by independent, unblinded statisticians. Part two was an open-label, non-randomised volunteer infection study using the Plasmodium falciparum induced blood-stage malaria model in which participants were enrolled into three dose cohorts. Healthy men and women of non-childbearing potential aged 18-55 years were eligible for inclusion; individuals in the volunteer infection study were required to be malaria naive. Safety and tolerability (primary outcome of the single ascending dose study and secondary outcome of the volunteer infection study) were assessed by frequency and severity of adverse events. The pharmacokinetic profile of M5717 was also characterised (primary outcome of the volunteer infection study and secondary outcome of the single ascending dose study). Parasite clearance kinetics (primary outcome of the volunteer infection study) were assessed by the parasite reduction ratio and the corresponding parasite clearance half-life; the incidence of recrudescence up to day 28 was determined (secondary outcome of the volunteer infection study). Recrudescent parasites were tested for genetic mutations (exploratory outcome). The trial is registered with ClinicalTrials.gov (NCT03261401). FINDINGS Between Aug 28, 2017, and June 14, 2019, 221 individuals were assessed for eligibility, of whom 66 men were enrolled in the single ascending dose study (eight per cohort for 50-1800 mg cohorts, randomised three M5717 to one placebo, and two in the 2100 mg cohort, randomised one M5717 to one placebo) and 22 men were enrolled in the volunteer infection study (six in the 150 mg cohort and eight each in the 400 mg and 800 mg cohorts). No adverse event was serious; all M5717-related adverse events were mild or moderate in severity and transient, with increased frequency observed at doses above 1250 mg. In the single ascending dose study, treatment-related adverse events occurred in three of 17 individuals in the placebo group; no individual in the 50 mg, 100 mg, or 200 mg groups; one of six individuals in each of the 400 mg, 1000 mg, and 1250 mg groups; two of six individuals in the 600 mg group; and in all individuals in the 1800 mg and 2100 mg groups. In the volunteer infection study, M5717-related adverse events occurred in no participants in the 150 mg or 800 mg groups and in one of eight participants in the 400 mg group. Transient oral hypoesthesia (in three participants) and blurred vision (in four participants) were observed in the 1800 mg or 2100 mg groups and constituted an unknown risk; thus, further dosing was suspended after dosing of the two sentinel individuals in the 2100 mg cohort. Maximum blood concentrations occurred 1-7 h after dosing, and a long half-life was observed (146-193 h at doses ≥200 mg). Parasite clearance occurred in all participants and was biphasic, characterised by initial slow clearance lasting 35-55 h (half-life 231·1 h [95% CI 40·9 to not reached] for 150 mg, 60·4 h [38·6 to 138·6] for 400 mg, and 24·7 h [20·4 to 31·3] for 800 mg), followed by rapid clearance (half-life 3·5 h [3·1 to 4·0] for 150 mg, 3·9 h [3·3 to 4·8] for 400 mg, and 5·5 h [4·8 to 6·4] for 800 mg). Recrudescence occurred in three (50%) of six individuals dosed with 150 mg and two (25%) of eight individuals dosed with 400 mg. Genetic mutations associated with resistance were detected in four cases of parasite recrudescence (two individuals dosed with 150 mg and two dosed with 400 mg). INTERPRETATION The safety, pharmacokinetics, and antimalarial activity of M5717 support its development as a component of a single-dose antimalarial combination therapy or for malaria prophylaxis. FUNDING Wellcome Trust and the healthcare business of Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
- 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
| | | | - Anand Odedra
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Liverpool School of Tropical Medicine, Liverpool, UK
| | - Rebecca Webster
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Claude Oeuvray
- The Global Health Institute of Merck (an affiliate of Merck KGaA), Eysin, Switzerland
| | - Aliona Tappert
- the healthcare business of Merck KGaA, Darmstadt, Germany
| | | | - Marla J Giddins
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Satish K Dhingra
- Department of Microbiology and Immunology, 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; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Xiaoyan Yin
- Global Statistics for NDD, Immunology, Endocrinology, Fertility & Others, EMD Serono, Billerica, MA, USA
| | | | | |
Collapse
|
6
|
Antimalarial drug candidates in phase I and II drug development: a scoping review. Antimicrob Agents Chemother 2021; 66:e0165921. [PMID: 34843390 PMCID: PMC8846400 DOI: 10.1128/aac.01659-21] [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] [Indexed: 11/20/2022] Open
Abstract
The emergence and spread of parasite resistance to currently available antimalarials has highlighted the importance of developing novel antimalarials. This scoping review provides an overview of antimalarial drug candidates undergoing phase I and II studies between 1 January 2016 and 28 April 2021. PubMed, Web of Science, Embase, clinical trial registries, and reference lists were searched for relevant studies. Information regarding antimalarial compound details, clinical trial characteristics, study population, and drug pharmacokinetics and pharmacodynamics (PK-PD) were extracted. A total of 50 studies were included, of which 24 had published their results and 26 were unpublished. New antimalarial compounds were evaluated as monotherapy (28 studies, 14 drug candidates) and combination therapy (9 studies, 10 candidates). Fourteen active compounds were identified in the current antimalarial drug development pipeline together with 11 compounds that are inactive, 6 due to insufficient efficacy. PK-PD data were available from 24 studies published as open-access articles. Four unpublished studies have made their results publicly available on clinical trial registries. The terminal elimination half-life of new antimalarial compounds ranged from 14.7 to 483 h. The log10 parasite reduction ratio over 48 h and parasite clearance half-life for Plasmodium falciparum following a single-dose monotherapy were 1.55 to 4.1 and 3.4 to 9.4 h, respectively. The antimalarial drug development landscape has seen a number of novel compounds, with promising PK-PD properties, evaluated in phase I and II studies over the past 5 years. Timely public disclosure of PK-PD data is crucial for informative decision-making and drug development strategy.
Collapse
|
7
|
McCarthy JS, Donini C, Chalon S, Woodford J, Marquart L, Collins KA, Rozenberg FD, Fidock DA, Cherkaoui-Rbati MH, Gobeau N, Möhrle JJ. A Phase 1, Placebo-controlled, Randomized, Single Ascending Dose Study and a Volunteer Infection Study to Characterize the Safety, Pharmacokinetics, and Antimalarial Activity of the Plasmodium Phosphatidylinositol 4-Kinase Inhibitor MMV390048. Clin Infect Dis 2021; 71:e657-e664. [PMID: 32239164 PMCID: PMC7744986 DOI: 10.1093/cid/ciaa368] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/01/2020] [Indexed: 02/01/2023] Open
Abstract
Background MMV390048 is the first Plasmodium phosphatidylinositol 4-kinase inhibitor to reach clinical development as a new antimalarial. We aimed to characterize the safety, pharmacokinetics, and antimalarial activity of a tablet formulation of MMV390048. Methods A 2-part, phase 1 trial was conducted in healthy adults. Part 1 was a double-blind, randomized, placebo-controlled, single ascending dose study consisting of 3 cohorts (40, 80, 120 mg MMV390048). Part 2 was an open-label volunteer infection study using the Plasmodium falciparum induced blood-stage malaria model consisting of 2 cohorts (40 mg and 80 mg MMV390048). Results Twenty four subjects were enrolled in part 1 (n = 8 per cohort, randomized 3:1 MMV390048:placebo) and 15 subjects were enrolled in part 2 (40 mg [n = 7] and 80 mg [n = 8] cohorts). One subject was withdrawn from part 2 (80 mg cohort) before dosing and was not included in analyses. No serious or severe adverse events were attributed to MMV390048. The rate of parasite clearance was greater in subjects administered 80 mg compared to those administered 40 mg (clearance half-life 5.5 hours [95% confidence interval {CI}, 5.2–6.0 hours] vs 6.4 hours [95% CI, 6.0–6.9 hours]; P = .005). Pharmacokinetic/pharmacodynamic modeling estimated a minimum inhibitory concentration of 83 ng/mL and a minimal parasiticidal concentration that would achieve 90% of the maximum effect of 238 ng/mL, and predicted that a single 120-mg dose would achieve an adequate clinical and parasitological response with 92% certainty. Conclusions The safety, pharmacokinetics, and pharmacodynamics of MMV390048 support its further development as a partner drug of a single-dose combination therapy for malaria. Clinical Trials Registration NCT02783820 (part 1); NCT02783833 (part 2).
Collapse
Affiliation(s)
- James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | - John Woodford
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Felix D Rozenberg
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA.,Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | | | | | | |
Collapse
|
8
|
Hughes E, Wallender E, Mohamed Ali A, Jagannathan P, Savic RM. Malaria PK/PD and the Role Pharmacometrics Can Play in the Global Health Arena: Malaria Treatment Regimens for Vulnerable Populations. Clin Pharmacol Ther 2021; 110:926-940. [PMID: 33763871 PMCID: PMC8518425 DOI: 10.1002/cpt.2238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/05/2021] [Indexed: 12/23/2022]
Abstract
Malaria is an infectious disease which disproportionately effects children and pregnant women. These vulnerable populations are often excluded from clinical trials resulting in one‐size‐fits‐all treatment regimens based on those established for a nonpregnant adult population. Pharmacokinetic/pharmacodynamic (PK/PD) models can be used to optimize dose selection as they define the drug exposure‐response relationship. Additionally, these models are able to identify patient characteristics that cause alterations in the expected PK/PD profiles and through simulations can recommend changes to dosing which compensate for the differences. In this review, we examine how PK/PD models have been applied to optimize antimalarial dosing recommendations for young children, including those who are malnourished, pregnant women, and individuals receiving concomitant therapies such as those for HIV treatment. The malaria field has had great success in utilizing PK/PD models as a foundation to update treatment guidelines and propose the next generation of dosing regimens to investigate in clinical trials. We propose how the malaria field can continue to use modeling to improve therapies by further integrating PK data into clinical studies and including data on drug resistance and host immunity in PK/PD models. Finally, we suggest that other disease areas can achieve similar success in applying pharmacometrics to improve outcomes by implementing three key principals.
Collapse
Affiliation(s)
- Emma Hughes
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Erika Wallender
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, California, USA
| | - Ali Mohamed Ali
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | | | - Radojka M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
9
|
Parasite-Host Dynamics throughout Antimalarial Drug Development Stages Complicate the Translation of Parasite Clearance. Antimicrob Agents Chemother 2021; 65:AAC.01539-20. [PMID: 33526486 PMCID: PMC8097426 DOI: 10.1128/aac.01539-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/20/2021] [Indexed: 11/21/2022] Open
Abstract
Ensuring continued success against malaria depends on a pipeline of new antimalarials. Antimalarial drug development utilizes preclinical murine and experimental human malaria infection studies to evaluate drug efficacy. Ensuring continued success against malaria depends on a pipeline of new antimalarials. Antimalarial drug development utilizes preclinical murine and experimental human malaria infection studies to evaluate drug efficacy. A sequential approach is typically adapted, with results from each stage informing the design of the next stage of development. The validity of this approach depends on confidence that results from murine malarial studies predict the outcome of clinical trials in humans. Parasite clearance rates following treatment are key parameters of drug efficacy. To investigate the validity of forward predictions, we developed a suite of mathematical models to capture parasite growth and drug clearance along the drug development pathway and estimated parasite clearance rates. When comparing the three infection experiments, we identified different relationships of parasite clearance with dose and different maximum parasite clearance rates. In Plasmodium berghei-NMRI mouse infections, we estimated a maximum parasite clearance rate of 0.2 (1/h); in Plasmodium falciparum-SCID mouse infections, 0.05 (1/h); and in human volunteer infection studies with P. falciparum, we found a maximum parasite clearance rate of 0.12 (1/h) and 0.18 (1/h) after treatment with OZ439 and MMV048, respectively. Sensitivity analysis revealed that host-parasite driven processes account for up to 25% of variance in parasite clearance for medium-high doses of antimalarials. Although there are limitations in translating parasite clearance rates across these experiments, they provide insight into characterizing key parameters of drug action and dose response and assist in decision-making regarding dosage for further drug development.
Collapse
|
10
|
Defining the Antimalarial Activity of Cipargamin in Healthy Volunteers Experimentally Infected with Blood-Stage Plasmodium falciparum. Antimicrob Agents Chemother 2021; 65:AAC.01423-20. [PMID: 33199389 PMCID: PMC7849011 DOI: 10.1128/aac.01423-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022] Open
Abstract
The spiroindolone cipargamin, a new antimalarial compound that inhibits Plasmodium ATP4, is currently in clinical development. This study aimed to characterize the antimalarial activity of cipargamin in healthy volunteers experimentally infected with blood-stage Plasmodium falciparum. The spiroindolone cipargamin, a new antimalarial compound that inhibits Plasmodium ATP4, is currently in clinical development. This study aimed to characterize the antimalarial activity of cipargamin in healthy volunteers experimentally infected with blood-stage Plasmodium falciparum. Eight subjects were intravenously inoculated with parasite-infected erythrocytes and received a single oral dose of 10 mg cipargamin 7 days later. Blood samples were collected to monitor the development and clearance of parasitemia and plasma cipargamin concentrations. Parasite regrowth was treated with piperaquine monotherapy to clear asexual parasites, while allowing gametocyte transmissibility to mosquitoes to be investigated. An initial rapid decrease in parasitemia occurred in all participants following cipargamin dosing, with a parasite clearance half-life of 3.99 h. As anticipated from the dose selected, parasite regrowth occurred in all 8 subjects 3 to 8 days after dosing and allowed the pharmacokinetic/pharmacodynamic relationship to be determined. Based on the limited data from the single subtherapeutic dose cohort, a MIC of 11.6 ng/ml and minimum parasiticidal concentration that achieves 90% of maximum effect of 23.5 ng/ml were estimated, and a single 95-mg dose (95% confidence interval [CI], 50 to 270) was predicted to clear 109 parasites/ml. Low gametocyte densities were detected in all subjects following piperaquine treatment, which did not transmit to mosquitoes. Serious adverse liver function changes were observed in three subjects, which led to premature study termination. The antimalarial activity characterized in this study supports the further clinical development of cipargamin as a new treatment for P. falciparum malaria, although the hepatic safety profile of the compound warrants further evaluation. (This study has been registered at ClinicalTrials.gov under identifier NCT02543086.)
Collapse
|
11
|
Andrews KA, Owen JS, McCarthy J, Wesche D, Gobeau N, Grasela TH, Möhrle JJ. Retrospective Analysis Using Pharmacokinetic/Pharmacodynamic Modeling and Simulation Offers Improvements in Efficiency of the Design of Volunteer Infection Studies for Antimalarial Drug Development. Clin Transl Sci 2020; 14:712-719. [PMID: 33326705 PMCID: PMC7993277 DOI: 10.1111/cts.12934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/13/2020] [Indexed: 01/23/2023] Open
Abstract
Volunteer infection studies using the induced blood stage malaria (IBSM) model have been shown to facilitate antimalarial drug development. Such studies have traditionally been undertaken in single-dose cohorts, as many as necessary to obtain the dose-response relationship. To enhance ethical and logistic aspects of such studies, and to reduce the number of cohorts needed to establish the dose-response relationship, we undertook a retrospective in silico analysis of previously accrued data to improve study design. A pharmacokinetic (PK)/pharmacodynamic (PD) model was developed from initial fictive-cohort data for OZ439 (mixing the data of the three single-dose cohorts as: n = 2 on 100 mg, 2 on 200 mg, and 4 on 500 mg). A three-compartment model described OZ439 PKs. Net growth of parasites was modeled using a Gompertz function and drug-induced parasite death using a Hill function. Parameter estimates for the PK and PD models were comparable for the multidose single-cohort vs. the pooled analysis of all cohorts. Simulations based on the multidose single-cohort design described the complete data from the original IBSM study. The novel design allows for the ascertainment of the PK/PD relationship early in the study, providing a basis for rational dose selection for subsequent cohorts and studies.
Collapse
Affiliation(s)
- Kayla Ann Andrews
- Cognigen Corporation, a SimulationsPlus Company, Buffalo, New York, USA.,Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Joel S Owen
- Cognigen Corporation, a SimulationsPlus Company, Buffalo, New York, USA
| | - James McCarthy
- The Royal Melbourne Hospital, The University of Melbourne at the Doherty Institute, Melbourne, Australia
| | - David Wesche
- Certara Strategic Consulting, Princeton, New Jersey, USA
| | | | | | | |
Collapse
|
12
|
Wockner LF, Hoffmann I, Webb L, Mordmüller B, Murphy SC, Kublin JG, O'Rourke P, McCarthy JS, Marquart L. Growth Rate of Plasmodium falciparum: Analysis of Parasite Growth Data From Malaria Volunteer Infection Studies. J Infect Dis 2020; 221:963-972. [PMID: 31679015 PMCID: PMC7198127 DOI: 10.1093/infdis/jiz557] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
Background Growth rate of malaria parasites in the blood of infected subjects is an important
measure of efficacy of drugs and vaccines. Methods We used log-linear and sine-wave models to estimate the parasite growth rate of the 3D7
strain of Plasmodium falciparum using data from 177 subjects from 14
induced blood stage malaria (IBSM) studies conducted at QIMR Berghofer. We estimated
parasite multiplication rate per 48 hour (PMR48), PMR per life-cycle
(PMRLC), and parasite life-cycle duration. We compared these parameters to
those from studies conducted elsewhere with infections induced by IBSM (n=66),
sporozoites via mosquito bite (n=336) or injection (n=51). Results The parasite growth rate of 3D7 in QIMR Berghofer studies was 0.75/day (95% CI:
0.73–0.77/day), PMR48 was 31.9 (95% CI: 28.7–35.4),
PMRLC was 16.4 (95% CI: 15.1–17.8) and parasite life-cycle was 38.8
hour (95% CI: 38.3–39.2 hour). These parameters were similar to estimates from
IBSM studies elsewhere (0.71/day, 95% CI: 0.67–0.75/day; PMR48 26.6,
95% CI: 22.2–31.8), but significantly higher (P < 0.001)
than in sporozoite studies (0.47/day, 95% CI: 0.43–0.50/day; PMR48
8.6, 95% CI: 7.3–10.1). Conclusions Parasite growth rates were similar across different IBSM studies and higher than
infections induced by sporozoite.
Collapse
Affiliation(s)
- Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Isabell Hoffmann
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Lachlan Webb
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - James G Kublin
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
13
|
Zhang LL, Li JL, Ji MX, Tian D, Wang LY, Chen C, Tian M. Attenuated P. falciparum Parasite Shows Cytokine Variations in Humanized Mice. Front Immunol 2020; 11:1801. [PMID: 33013831 PMCID: PMC7516016 DOI: 10.3389/fimmu.2020.01801] [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: 04/24/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
A recently developed humanized mouse has been used to assess the immune response evoked against the isolated attenuated C9 parasite clone (C9-M; carrying a single insertion disrupting the open reading frame (ORF) of PF3D7_1305500) of Plasmodium falciparum. Significant human RBC engraftment was achieved by ameliorating the residual non-adaptive immune response using clodronate-loaded liposome treatment. Controlled reactive professional phagocytic leukocytes in immunodeficient mice allowed for sizeable human blood chimerism and injected huRBCs acted as bona fide host cells for P. falciparum. huRBC-reconstituted immunodeficient mice received infectious challenge with attenuated P. falciparum C9 parasite mutants (C9-M), complemented (C9-C), and wild type (NF54) progenitors to study the role of immune effectors in the clearance of the parasite from mouse circulation. C9-M and NF54 parasites grew and developed in the huRBC-reconstituted humanized NSG mice. Further, the presence of mutant parasites in deep-seated tissues suggests the escape of parasites from the host's immune responses and thus extended the survival of the parasite. Our results suggest an evasion mechanism that may have been employed by the parasite to survive the mouse's residual non-adaptive immune responses. Collectively, our data suggest that huRBCs reconstituted NSG mice infected with attenuated P. falciparum is a valuable tool to explore the role of C9 mutation in the growth and survival of parasite mutants and their response to the host's immune responses. This mouse might help in identifying novel chemotherapeutic targets to develop new anti-malarial drugs.
Collapse
Affiliation(s)
- Lei-Lei Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Jin-Long Li
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ming-Xin Ji
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Dan Tian
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Li-Yan Wang
- Department of Operating Room, The Second Hospital of Jilin University, Changchun, China
| | - Chen Chen
- Department of Operating Room, The Second Hospital of Jilin University, Changchun, China
| | - Miao Tian
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Chughlay MF, Akakpo S, Odedra A, Csermak-Renner K, Djeriou E, Winnips C, Leboulleux D, Gaur AH, Shanks GD, McCarthy J, Chalon S. Liver Enzyme Elevations in Plasmodium falciparum Volunteer Infection Studies: Findings and Recommendations. Am J Trop Med Hyg 2020; 103:378-393. [PMID: 32314694 PMCID: PMC7356411 DOI: 10.4269/ajtmh.19-0846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Malaria volunteer infection studies (VISs) accelerate new drug and vaccine development. In the induced blood-stage malaria (IBSM) model, volunteers are inoculated with erythrocytes infected with Plasmodium falciparum. Observations of elevated liver enzymes in the IBSM model with new chemical entities (NCEs) promoted an analysis of available data. Data were reviewed from eight IBSM studies of seven different NCEs, plus two studies with the registered antimalarial piperaquine conducted between June 2013 and January 2017 at QIMR Berghofer, Brisbane, Australia. Alanine aminotransferase (ALT) was elevated (> 2.5 times the upper limit of normal [×ULN]) in 20/114 (17.5%) participants. Of these, 8.9% (10/114) had moderate increases (> 2.5–5 × ULN), noted in seven studies of six different NCEs ± piperaquine or piperaquine alone, and 8.9% (10/114) had severe elevations (> 5 × ULN), occurring in six studies of six different NCEs ± piperaquine. Aspartate aminotransferase (AST) was elevated (> 2.5 × ULN) in 11.4% (13/114) of participants, across six of the 10 studies. Bilirubin was > 2 × ULN in one participant. Published data from other VIS models, using sporozoite inoculation by systemic administration or mosquito feeding, also showed moderate/severe liver enzyme elevations. In conclusion, liver enzyme elevations in IBSM studies are most likely multifactorial and could be caused by the model conditions, that is, malaria infection/parasite density and/or effective parasite clearance, or by participant-specific risk factors, acetaminophen administration, or direct hepatotoxicity of the test drug. We make recommendations that may mitigate the risk of liver enzyme elevations in future VISs and propose measures to assist their interpretation, should they occur.
Collapse
Affiliation(s)
| | | | - Anand Odedra
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | | | | | - Aditya H Gaur
- St. Jude Children's Research Hospital, Memphis, Tennessee
| | - G Dennis Shanks
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - James McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | |
Collapse
|
15
|
Burgert L, Rottmann M, Wittlin S, Gobeau N, Krause A, Dingemanse J, Möhrle JJ, Penny MA. Ensemble modeling highlights importance of understanding parasite-host behavior in preclinical antimalarial drug development. Sci Rep 2020; 10:4410. [PMID: 32157151 PMCID: PMC7064600 DOI: 10.1038/s41598-020-61304-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/20/2020] [Indexed: 11/23/2022] Open
Abstract
Emerging drug resistance and high-attrition rates in early and late stage drug development necessitate accelerated development of antimalarial compounds. However, systematic and meaningful translation of drug efficacy and host-parasite dynamics between preclinical testing stages is missing. We developed an ensemble of mathematical within-host parasite growth and antimalarial action models, fitted to extensive data from four antimalarials with different modes of action, to assess host-parasite interactions in two preclinical drug testing systems of murine parasite P. berghei in mice, and human parasite P. falciparum in immune-deficient mice. We find properties of the host-parasite system, namely resource availability, parasite maturation and virulence, drive P. berghei dynamics and drug efficacy, whereas experimental constraints primarily influence P. falciparum infection and drug efficacy. Furthermore, uninvestigated parasite behavior such as dormancy influences parasite recrudescence following non-curative treatment and requires further investigation. Taken together, host-parasite interactions should be considered for meaningful translation of pharmacodynamic properties between murine systems and for predicting human efficacious treatment.
Collapse
Affiliation(s)
- Lydia Burgert
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Andreas Krause
- Idorsia Pharmaceuticals Ltd, Clinical Pharmacology, Allschwil, Switzerland
| | - Jasper Dingemanse
- Idorsia Pharmaceuticals Ltd, Clinical Pharmacology, Allschwil, Switzerland
| | - Jörg J Möhrle
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Medicines for Malaria Venture, Geneva, Switzerland
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| |
Collapse
|
16
|
Charman SA, Andreu A, Barker H, Blundell S, Campbell A, Campbell M, Chen G, Chiu FCK, Crighton E, Katneni K, Morizzi J, Patil R, Pham T, Ryan E, Saunders J, Shackleford DM, White KL, Almond L, Dickins M, Smith DA, Moehrle JJ, Burrows JN, Abla N. An in vitro toolbox to accelerate anti-malarial drug discovery and development. Malar J 2020; 19:1. [PMID: 31898492 PMCID: PMC6941357 DOI: 10.1186/s12936-019-3075-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/14/2019] [Indexed: 01/08/2023] Open
Abstract
Background Modelling and simulation are being increasingly utilized to support the discovery and development of new anti-malarial drugs. These approaches require reliable in vitro data for physicochemical properties, permeability, binding, intrinsic clearance and cytochrome P450 inhibition. This work was conducted to generate an in vitro data toolbox using standardized methods for a set of 45 anti-malarial drugs and to assess changes in physicochemical properties in relation to changing target product and candidate profiles. Methods Ionization constants were determined by potentiometric titration and partition coefficients were measured using a shake-flask method. Solubility was assessed in biorelevant media and permeability coefficients and efflux ratios were determined using Caco-2 cell monolayers. Binding to plasma and media proteins was measured using either ultracentrifugation or rapid equilibrium dialysis. Metabolic stability and cytochrome P450 inhibition were assessed using human liver microsomes. Sample analysis was conducted by LC–MS/MS. Results Both solubility and fraction unbound decreased, and permeability and unbound intrinsic clearance increased, with increasing Log D7.4. In general, development compounds were somewhat more lipophilic than legacy drugs. For many compounds, permeability and protein binding were challenging to assess and both required the use of experimental conditions that minimized the impact of non-specific binding. Intrinsic clearance in human liver microsomes was varied across the data set and several compounds exhibited no measurable substrate loss under the conditions used. Inhibition of cytochrome P450 enzymes was minimal for most compounds. Conclusions This is the first data set to describe in vitro properties for 45 legacy and development anti-malarial drugs. The studies identified several practical methodological issues common to many of the more lipophilic compounds and highlighted areas which require more work to customize experimental conditions for compounds being designed to meet the new target product profiles. The dataset will be a valuable tool for malaria researchers aiming to develop PBPK models for the prediction of human PK properties and/or drug–drug interactions. Furthermore, generation of this comprehensive data set within a single laboratory allows direct comparison of properties across a large dataset and evaluation of changing property trends that have occurred over time with changing target product and candidate profiles.
Collapse
Affiliation(s)
- Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| | - Alice Andreu
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Helena Barker
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Scott Blundell
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Anna Campbell
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Michael Campbell
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Francis C K Chiu
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Elly Crighton
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Julia Morizzi
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Rahul Patil
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Thao Pham
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Eileen Ryan
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Jessica Saunders
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Karen L White
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Lisa Almond
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Maurice Dickins
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | | | - Joerg J Moehrle
- Medicines for Malaria Venture, PO Box 1826, 20 Route de Pré-Bois, CH-1215, Geneva 15, Switzerland
| | - Jeremy N Burrows
- Medicines for Malaria Venture, PO Box 1826, 20 Route de Pré-Bois, CH-1215, Geneva 15, Switzerland
| | - Nada Abla
- Medicines for Malaria Venture, PO Box 1826, 20 Route de Pré-Bois, CH-1215, Geneva 15, Switzerland
| |
Collapse
|
17
|
Atmar RL, Keitel WA. Searching for Improved Flu Vaccines-The Time Is Now. J Infect Dis 2020; 221:1-4. [PMID: 31665360 DOI: 10.1093/infdis/jiz545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Robert L Atmar
- Departments of Medicine and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wendy A Keitel
- Departments of Medicine and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
18
|
A Single-Dose Combination Study with the Experimental Antimalarials Artefenomel and DSM265 To Determine Safety and Antimalarial Activity against Blood-Stage Plasmodium falciparum in Healthy Volunteers. Antimicrob Agents Chemother 2019; 64:AAC.01371-19. [PMID: 31685476 PMCID: PMC7187626 DOI: 10.1128/aac.01371-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022] Open
Abstract
Artefenomel and DSM265 are two new compounds that have been shown to be well tolerated and effective when administered as monotherapy malaria treatment. This study aimed to determine the safety, pharmacokinetics, and pharmacodynamics of artefenomel and DSM265 administered in combination to healthy subjects in a volunteer infection study using the Plasmodium falciparum-induced blood-stage malaria model. Thirteen subjects were inoculated with parasite-infected erythrocytes on day 0 and received a single oral dose of artefenomel and DSM265 on day 7. Cohort 1 (n = 8) received 200 mg artefenomel plus 100 mg DSM265, and cohort 2 (n = 5) received 200 mg artefenomel plus 50 mg DSM265. Blood samples were collected to measure parasitemia, gametocytemia, and artefenomel-DSM265 plasma concentrations. There were no treatment-related adverse events. The pharmacokinetic profiles of artefenomel and DSM265 were similar to those of the compounds when administered as monotherapy, suggesting no pharmacokinetic interactions. A reduction in parasitemia occurred in all subjects following treatment (log10 parasite reduction ratios over 48 h [PRR48] of 2.80 for cohort 1 and 2.71 for cohort 2; parasite clearance half-lives of 5.17 h for cohort 1 and 5.33 h for cohort 2). Recrudescence occurred in 5/8 subjects in cohort 1 between days 19 and 28 and in 5/5 subjects in cohort 2 between days 15 and 22. Low-level gametocytemia (1 to 330 female gametocytes/ml) was detected in all subjects from day 14. The results of this single-dosing combination study support the further clinical development of the use of artefenomel and DSM265 in combination as a treatment for falciparum malaria. (This study has been registered at ClinicalTrials.gov under identifier NCT02389348.).
Collapse
|
19
|
Yadav DK, Kumar S, Teli MK, Yadav R, Chaudhary S. Molecular Targets for Malarial Chemotherapy: A Review. Curr Top Med Chem 2019; 19:861-873. [DOI: 10.2174/1568026619666190603080000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 11/22/2022]
Abstract
The malaria parasite resistance to the existing drugs is a serious problem to the currently used
antimalarials and, thus, highlights the urgent need to develop new and effective anti-malarial molecules.
This could be achieved either by the identification of the new drugs for the validated targets or by further
refining/improving the existing antimalarials; or by combining previously effective agents with
new/existing drugs to have a synergistic effect that counters parasite resistance; or by identifying novel
targets for the malarial chemotherapy. In this review article, a comprehensive collection of some of the
novel molecular targets has been enlisted for the antimalarial drugs. The targets which could be deliberated
for developing new anti-malarial drugs could be: membrane biosynthesis, mitochondrial system,
apicoplasts, parasite transporters, shikimate pathway, hematin crystals, parasite proteases, glycolysis,
isoprenoid synthesis, cell cycle control/cycline dependent kinase, redox system, nucleic acid metabolism,
methionine cycle and the polyamines, folate metabolism, the helicases, erythrocyte G-protein, and
farnesyl transferases. Modern genomic tools approaches such as structural biology and combinatorial
chemistry, novel targets could be identified followed by drug development for drug resistant strains providing
wide ranges of novel targets in the development of new therapy. The new approaches and targets
mentioned in the manuscript provide a basis for the development of new unique strategies for antimalarial
therapy with limited off-target effects in the near future.
Collapse
Affiliation(s)
- Dharmendra K. Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Surendra Kumar
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Mahesh K. Teli
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Ravikant Yadav
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur-302017, India
| | - Sandeep Chaudhary
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur-302017, India
| |
Collapse
|
20
|
Ozonide Antimalarial Activity in the Context of Artemisinin-Resistant Malaria. Trends Parasitol 2019; 35:529-543. [PMID: 31176584 DOI: 10.1016/j.pt.2019.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/02/2019] [Accepted: 05/01/2019] [Indexed: 12/18/2022]
Abstract
The ozonides are one of the most advanced drug classes in the antimalarial development pipeline and were designed to improve on limitations associated with current front-line artemisinin-based therapies. Like the artemisinins, the pharmacophoric peroxide bond of ozonides is essential for activity, and it appears that these antimalarials share a similar mode of action, raising the possibility of cross-resistance. Resistance to artemisinins is associated with Plasmodium falciparum mutations that allow resistant parasites to escape short-term artemisinin-mediated damage (elimination half-life ~1 h). Importantly, some ozonides (e.g., OZ439) have a sustained in vivo drug exposure profile, providing a major pharmacokinetic advantage over the artemisinin derivatives. Here, we describe recent progress made towards understanding ozonide antimalarial activity and discuss ozonide utility within the context of artemisinin resistance.
Collapse
|
21
|
UCT943, a Next-Generation Plasmodium falciparum PI4K Inhibitor Preclinical Candidate for the Treatment of Malaria. Antimicrob Agents Chemother 2018; 62:AAC.00012-18. [PMID: 29941635 PMCID: PMC6125526 DOI: 10.1128/aac.00012-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/07/2018] [Indexed: 01/12/2023] Open
Abstract
The 2-aminopyridine MMV048 was the first drug candidate inhibiting Plasmodium phosphatidylinositol 4-kinase (PI4K), a novel drug target for malaria, to enter clinical development. In an effort to identify the next generation of PI4K inhibitors, the series was optimized to improve properties such as solubility and antiplasmodial potency across the parasite life cycle, leading to the 2-aminopyrazine UCT943. The compound displayed higher asexual blood stage, transmission-blocking, and liver stage activities than MMV048 and was more potent against resistant Plasmodium falciparum and Plasmodium vivax clinical isolates. Excellent in vitro antiplasmodial activity translated into high efficacy in Plasmodium berghei and humanized P. falciparum NOD-scid IL-2Rγ null mouse models. The high passive permeability and high aqueous solubility of UCT943, combined with low to moderate in vivo intrinsic clearance, resulted in sustained exposure and high bioavailability in preclinical species. In addition, the predicted human dose for a curative single administration using monkey and dog pharmacokinetics was low, ranging from 50 to 80 mg. As a next-generation Plasmodium PI4K inhibitor, UCT943, based on the combined preclinical data, has the potential to form part of a single-exposure radical cure and prophylaxis (SERCaP) to treat, prevent, and block the transmission of malaria.
Collapse
|
22
|
Rufener R, Ritler D, Zielinski J, Dick L, da Silva ET, da Silva Araujo A, Joekel DE, Czock D, Goepfert C, Moraes AM, de Souza MVN, Müller J, Mevissen M, Hemphill A, Lundström-Stadelmann B. Activity of mefloquine and mefloquine derivatives against Echinococcus multilocularis. Int J Parasitol Drugs Drug Resist 2018; 8:331-340. [PMID: 29933218 PMCID: PMC6020078 DOI: 10.1016/j.ijpddr.2018.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/27/2022]
Abstract
The cestode E. multilocularis causes the disease alveolar echinococcosis (AE) in humans. The continuously proliferating metacestode (larval stage) of the parasite infects mostly the liver and exhibits tumor-like growth. Current chemotherapeutical treatment options rely on benzimidazoles, which are rarely curative and have to be applied daily and life-long. This can result in considerable hepatotoxicity and thus treatment discontinuation. Therefore, novel drugs against AE are urgently needed. The anti-malarial mefloquine was previously shown to be active against E. multilocularis metacestodes in vitro, and in mice infected by intraperitoneal inoculation of metacestodes when administered at 100 mg/kg by oral gavage twice a week for 12 weeks. In the present study, the same dosage regime was applied in mice infected via oral uptake of eggs representing the natural route of infection. After 12 weeks of treatment, the presence of parasite lesions was assessed in a liver squeeze chamber and by PCR, and a significantly reduced parasite load was found in mefloquine-treated animals. Assessment of mefloquine plasma concentrations by HPLC and modeling using a two-compartment pharmacokinetic model with first-order absorption showed that >90% of the expected steady-state levels (Cmin 1.15 mg/L, Cmax 2.63 mg/L) were reached. These levels are close to concentrations achieved in humans during long-term weekly dosage of 250 mg (dose applied for malaria prophylaxis). In vitro structure-activity relationship analysis of mefloquine and ten derivatives revealed that none of the derivatives exhibited stronger activities than mefloquine. Activity was only observed, when the 2-piperidylmethanol group of mefloquine was replaced by an amino group-containing residue and when the trifluoromethyl residue on position 8 of the quinoline structure was present. This is in line with the anti-malarial activity of mefloquine and it implies that the mode of action in E. multilocularis might be similar to the one against malaria.
Collapse
Affiliation(s)
- Reto Rufener
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland
| | - Dominic Ritler
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland
| | - Jana Zielinski
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, 3012, Bern, Switzerland
| | - Luca Dick
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland
| | - Emerson Teixeira da Silva
- Fundação Oswaldo Cruz, Instituto de Tecnologia em Fármacos - Far Manguinhos, 21041-250, Rio de Janeiro, Brazil
| | - Adriele da Silva Araujo
- Fundação Oswaldo Cruz, Instituto de Tecnologia em Fármacos - Far Manguinhos, 21041-250, Rio de Janeiro, Brazil
| | - Deborah Elisabeth Joekel
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
| | - David Czock
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Christine Goepfert
- Institute of Animal Pathology COMPATH, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, Switzerland
| | - Adriana Marques Moraes
- Fundação Oswaldo Cruz, Instituto de Tecnologia em Fármacos - Far Manguinhos, 21041-250, Rio de Janeiro, Brazil
| | | | - Joachim Müller
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland
| | - Meike Mevissen
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, 3012, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland
| | - Britta Lundström-Stadelmann
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012, Bern, Switzerland.
| |
Collapse
|
23
|
De Rycker M, Baragaña B, Duce SL, Gilbert IH. Challenges and recent progress in drug discovery for tropical diseases. Nature 2018; 559:498-506. [PMID: 30046073 PMCID: PMC6129172 DOI: 10.1038/s41586-018-0327-4] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
Infectious tropical diseases have a huge effect in terms of mortality and morbidity, and impose a heavy economic burden on affected countries. These diseases predominantly affect the world's poorest people. Currently available drugs are inadequate for the majority of these diseases, and there is an urgent need for new treatments. This Review discusses some of the challenges involved in developing new drugs to treat these diseases and highlights recent progress. While there have been notable successes, there is still a long way to go.
Collapse
Affiliation(s)
- Manu De Rycker
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Beatriz Baragaña
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK
| | - Suzanne L Duce
- Medicines Monitoring Unit (MEMO), Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Ian H Gilbert
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
24
|
Abstract
The last two decades have seen a surge in antimalarial drug development with product development partnerships taking a leading role. Resistance of Plasmodium falciparum to the artemisinin derivatives, piperaquine and mefloquine in Southeast Asia means new antimalarials are needed with some urgency. There are at least 13 agents in clinical development. Most of these are blood schizonticides for the treatment of uncomplicated falciparum malaria, under evaluation either singly or as part of two-drug combinations. Leading candidates progressing through the pipeline are artefenomel-ferroquine and lumefantrine-KAF156, both in Phase 2b. Treatment of severe malaria continues to rely on two parenteral drugs with ancient forebears: artesunate and quinine, with sevuparin being evaluated as an adjuvant therapy. Tafenoquine is under review by stringent regulatory authorities for approval as a single-dose treatment for Plasmodium vivax relapse prevention. This represents an advance over standard 14-day primaquine regimens; however, the risk of acute haemolytic anaemia in patients with glucose-6-phosphate dehydrogenase deficiency remains. For disease prevention, several of the newer agents show potential but are unlikely to be recommended for use in the main target groups of pregnant women and young children for some years. Latest predictions are that the malaria burden will continue to be high in the coming decades. This fact, coupled with the repeated loss of antimalarials to resistance, indicates that new antimalarials will be needed for years to come. Failure of the artemisinin-based combinations in Southeast Asia has stimulated a reappraisal of current approaches to combination therapy for malaria with incorporation of three or more drugs in a single treatment under consideration.
Collapse
Affiliation(s)
- Elizabeth A Ashley
- Myanmar Oxford Clinical Research Unit, Yangon, Myanmar.
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | | |
Collapse
|
25
|
Paquet T, Le Manach C, Cabrera DG, Younis Y, Henrich PP, Abraham TS, Lee MCS, Basak R, Ghidelli-Disse S, Lafuente-Monasterio MJ, Bantscheff M, Ruecker A, Blagborough AM, Zakutansky SE, Zeeman AM, White KL, Shackleford DM, Mannila J, Morizzi J, Scheurer C, Angulo-Barturen I, Martínez MS, Ferrer S, Sanz LM, Gamo FJ, Reader J, Botha M, Dechering KJ, Sauerwein RW, Tungtaeng A, Vanachayangkul P, Lim CS, Burrows J, Witty MJ, Marsh KC, Bodenreider C, Rochford R, Solapure SM, Jiménez-Díaz MB, Wittlin S, Charman SA, Donini C, Campo B, Birkholtz LM, Hanson KK, Drewes G, Kocken CHM, Delves MJ, Leroy D, Fidock DA, Waterson D, Street LJ, Chibale K. Antimalarial efficacy of MMV390048, an inhibitor of Plasmodium phosphatidylinositol 4-kinase. Sci Transl Med 2018; 9:9/387/eaad9735. [PMID: 28446690 DOI: 10.1126/scitranslmed.aad9735] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
As part of the global effort toward malaria eradication, phenotypic whole-cell screening revealed the 2-aminopyridine class of small molecules as a good starting point to develop new antimalarial drugs. Stemming from this series, we found that the derivative, MMV390048, lacked cross-resistance with current drugs used to treat malaria. This compound was efficacious against all Plasmodium life cycle stages, apart from late hypnozoites in the liver. Efficacy was shown in the humanized Plasmodium falciparum mouse model, and modest reductions in mouse-to-mouse transmission were achieved in the Plasmodium berghei mouse model. Experiments in monkeys revealed the ability of MMV390048 to be used for full chemoprotection. Although MMV390048 was not able to eliminate liver hypnozoites, it delayed relapse in a Plasmodium cynomolgi monkey model. Both genomic and chemoproteomic studies identified a kinase of the Plasmodium parasite, phosphatidylinositol 4-kinase, as the molecular target of MMV390048. The ability of MMV390048 to block all life cycle stages of the malaria parasite suggests that this compound should be further developed and may contribute to malaria control and eradication as part of a single-dose combination treatment.
Collapse
Affiliation(s)
- Tanya Paquet
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Claire Le Manach
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Yassir Younis
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Philipp P Henrich
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.,The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Tara S Abraham
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, Suite 368, Philadelphia, PA 19107, USA
| | - Marcus C S Lee
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Rajshekhar Basak
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, CT 06520-8114, USA
| | - Sonja Ghidelli-Disse
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - María José Lafuente-Monasterio
- Malaria Disease Performance Unit, Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Marcus Bantscheff
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Andrea Ruecker
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | | | | | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, 2280 GH Rijswijk, Netherlands
| | - Karen L White
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Janne Mannila
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Admescope Ltd., Typpitie 1, 90620 Oulu, Finland
| | - Julia Morizzi
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Christian Scheurer
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland.,University of Basel, 4003 Basel, Switzerland
| | - Iñigo Angulo-Barturen
- Malaria Disease Performance Unit, Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - María Santos Martínez
- Malaria Disease Performance Unit, Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Santiago Ferrer
- Malaria Disease Performance Unit, Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Laura María Sanz
- Malaria Disease Performance Unit, Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Francisco Javier Gamo
- Malaria Disease Performance Unit, Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Janette Reader
- Department of Biochemistry, Centre for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Mariette Botha
- Department of Biochemistry, Centre for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Koen J Dechering
- TropIQ Health Sciences, Transistorweg 5, 6534 AT Nijmegen, Netherlands
| | - Robert W Sauerwein
- TropIQ Health Sciences, Transistorweg 5, 6534 AT Nijmegen, Netherlands.,Radboud University Medical Center, Department of Medical Microbiology, 6500 HB Nijmegen, Netherlands
| | - Anchalee Tungtaeng
- Department of Veterinary Medicine, Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Pattaraporn Vanachayangkul
- Department of Immunology and Medicine, Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Chek Shik Lim
- Novartis Institute for Tropical Diseases Pte. Ltd., 10 Biopolis Road, #05-01 Chromos, Singapore 138670, Singapore
| | - Jeremy Burrows
- Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Michael J Witty
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.,Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Kennan C Marsh
- AbbVie, 1 North Waukegan Road, North Chicago, IL 60064-6104, USA
| | - Christophe Bodenreider
- Novartis Institute for Tropical Diseases Pte. Ltd., 10 Biopolis Road, #05-01 Chromos, Singapore 138670, Singapore
| | - Rosemary Rochford
- Departments of Immunology and Microbiology and Environmental and Occupational Health, University of Colorado Denver, Aurora, CO 80045, USA
| | - Suresh M Solapure
- Nagarjuna Gardens, 60 Feet Road, Sahakaranagar, Bangalore 560092, India
| | - María Belén Jiménez-Díaz
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1020 Locust Street, Suite 368, Philadelphia, PA 19107, USA
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland.,University of Basel, 4003 Basel, Switzerland
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Cristina Donini
- Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Brice Campo
- Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Centre for Sustainable Malaria Control, University of Pretoria, Pretoria, South Africa
| | - Kirsten K Hanson
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Gerard Drewes
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, 2280 GH Rijswijk, Netherlands
| | - Michael J Delves
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| | - Didier Leroy
- Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.,Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - David Waterson
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.,Medicines for Malaria Venture, International Center Cointrin, Route de Pré-Bois 20, 1215 Geneva, Switzerland
| | - Leslie J Street
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa. .,South African Medical Research Council Drug Discovery and Development Research Unit, and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
26
|
Moreno-Sabater A, Pérignon JL, Mazier D, Lavazec C, Soulard V. Humanized mouse models infected with human Plasmodium species for antimalarial drug discovery. Expert Opin Drug Discov 2017; 13:131-140. [DOI: 10.1080/17460441.2018.1410136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Alicia Moreno-Sabater
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
- Assistance Publique - Hopitaux de Paris - Hôpitaux Universitaires Paris-Est - Site Saint-Antoine, Paris, Île-de-France France
| | | | - Dominique Mazier
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| | - Catherine Lavazec
- Institut Cochin – INSERM U1016, Paris, Île-de-France France
- CNRS - UMR8104, Paris, France
- Universite Paris Descartes, Paris, Île-de-France France
| | - Valerie Soulard
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| |
Collapse
|
27
|
Abstract
Basic science holds enormous power for revealing the biological mechanisms of disease and, in turn, paving the way toward new, effective interventions. Recognizing this power, the 2011 Research Agenda for Malaria Eradication included key priorities in fundamental research that, if attained, could help accelerate progress toward disease elimination and eradication. The Malaria Eradication Research Agenda (malERA) Consultative Panel on Basic Science and Enabling Technologies reviewed the progress, continuing challenges, and major opportunities for future research. The recommendations come from a literature of published and unpublished materials and the deliberations of the malERA Refresh Consultative Panel. These areas span multiple aspects of the Plasmodium life cycle in both the human host and the Anopheles vector and include critical, unanswered questions about parasite transmission, human infection in the liver, asexual-stage biology, and malaria persistence. We believe an integrated approach encompassing human immunology, parasitology, and entomology, and harnessing new and emerging biomedical technologies offers the best path toward addressing these questions and, ultimately, lowering the worldwide burden of malaria.
Collapse
|
28
|
Abstract
Since the turn of the century, a remarkable expansion has been achieved in the range and effectiveness of products and strategies available to prevent, treat, and control malaria, including advances in diagnostics, drugs, vaccines, and vector control. These advances have once again put malaria elimination on the agenda. However, it is clear that even with the means available today, malaria control and elimination pose a formidable challenge in many settings. Thus, currently available resources must be used more effectively, and new products and approaches likely to achieve these goals must be developed. This paper considers tools (both those available and others that may be required) to achieve and maintain malaria elimination. New diagnostics are needed to direct treatment and detect transmission potential; new drugs and vaccines to overcome existing resistance and protect against clinical and severe disease, as well as block transmission and prevent relapses; and new vector control measures to overcome insecticide resistance and more powerfully interrupt transmission. It is also essential that strategies for combining new and existing approaches are developed for different settings to maximise their longevity and effectiveness in areas with continuing transmission and receptivity. For areas where local elimination has been recently achieved, understanding which measures are needed to maintain elimination is necessary to prevent rebound and the reestablishment of transmission. This becomes increasingly important as more countries move towards elimination.
Collapse
|
29
|
Andrews KA, Wesche D, McCarthy J, Möhrle JJ, Tarning J, Phillips L, Kern S, Grasela T. Model-Informed Drug Development for Malaria Therapeutics. Annu Rev Pharmacol Toxicol 2017; 58:567-582. [PMID: 28992431 PMCID: PMC7198115 DOI: 10.1146/annurev-pharmtox-010715-103429] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Malaria is a critical public health problem resulting in substantial morbidity and
mortality, particularly in developing countries. Owing to the development of resistance
toward current therapies, novel approaches to accelerate the development efforts of new
malaria therapeutics are urgently needed. There have been significant advancements in the
development of in vitro and in vivo experiments that generate data used to inform
decisions about the potential merit of new compounds. A comprehensive disease-drug model
capable of integrating discrete data from different preclinical and clinical components
would be a valuable tool across all stages of drug development. This could have an
enormous impact on the otherwise slow and resource-intensive process of traditional
clinical drug development.
Collapse
Affiliation(s)
- Kayla Ann Andrews
- Cognigen Corporation, a subsidiary of Simulations Plus, Buffalo, New York 14221, USA; , , .,Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York 14214, USA
| | - David Wesche
- Bill and Melinda Gates Foundation, Seattle, Washington 98109, USA; ,
| | - James McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,School of Medicine, University of Queensland, Brisbane, Australia;
| | - Jörg J Möhrle
- Medicines for Malaria Venture, Geneva 1215, Switzerland;
| | - Joel Tarning
- 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 OX3 7FZ, United Kingdom
| | - Luann Phillips
- Cognigen Corporation, a subsidiary of Simulations Plus, Buffalo, New York 14221, USA; , ,
| | - Steven Kern
- Bill and Melinda Gates Foundation, Seattle, Washington 98109, USA; ,
| | - Thaddeus Grasela
- Cognigen Corporation, a subsidiary of Simulations Plus, Buffalo, New York 14221, USA; , ,
| |
Collapse
|
30
|
Shanks GD, Möhrle JJ. Treating malaria: new drugs for a new era. THE LANCET. INFECTIOUS DISEASES 2017; 17:1223-1224. [PMID: 28916444 DOI: 10.1016/s1473-3099(17)30475-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 01/08/2023]
Affiliation(s)
- G Dennis Shanks
- Australian Army Malaria Institute, Brisbane, QLD 4051, Australia.
| | | |
Collapse
|
31
|
Affiliation(s)
- Robert E. Sinden
- Department of Life Sciences, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
McCarthy JS, Lotharius J, Rückle T, Chalon S, Phillips MA, Elliott S, Sekuloski S, Griffin P, Ng CL, Fidock DA, Marquart L, Williams NS, Gobeau N, Bebrevska L, Rosario M, Marsh K, Möhrle JJ. Safety, tolerability, pharmacokinetics, and activity of the novel long-acting antimalarial DSM265: a two-part first-in-human phase 1a/1b randomised study. THE LANCET. INFECTIOUS DISEASES 2017; 17:626-635. [PMID: 28363636 PMCID: PMC5446412 DOI: 10.1016/s1473-3099(17)30171-8] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/10/2017] [Accepted: 02/16/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND DSM265 is a novel antimalarial that inhibits plasmodial dihydroorotate dehydrogenase, an enzyme essential for pyrimidine biosynthesis. We investigated the safety, tolerability, and pharmacokinetics of DSM265, and tested its antimalarial activity. METHODS Healthy participants aged 18-55 years were enrolled in a two-part study: part 1, a single ascending dose (25-1200 mg), double-blind, randomised, placebo-controlled study, and part 2, an open-label, randomised, active-comparator controlled study, in which participants were inoculated with Plasmodium falciparum induced blood-stage malaria (IBSM) and treated with DSM265 (150 mg) or mefloquine (10 mg/kg). Primary endpoints were DSM265 safety, tolerability, and pharmacokinetics. Randomisation lists were created using a validated, automated system. Both parts were registered with the Australian New Zealand Clinical Trials Registry, number ACTRN12613000522718 (part 1) and number ACTRN12613000527763 (part 2). FINDINGS In part 1, 73 participants were enrolled between April 12, 2013, and July 14, 2015 (DSM265, n=55; placebo, n=18). In part 2, nine participants were enrolled between Sept 30 and Nov 25, 2013 (150 mg DSM265, n=7; 10 mg/kg mefloquine, n=2). In part 1, 117 adverse events were reported; no drug-related serious or severe events were reported. The most common drug-related adverse event was headache. The mean DSM265 peak plasma concentration (Cmax) ranged between 1310 ng/mL and 34 800 ng/mL and was reached in a median time (tmax) between 1·5 h and 4 h, with a mean elimination half-life between 86 h and 118 h. In part 2, the log10 parasite reduction ratio at 48 h in the DSM265 (150 mg) group was 1·55 (95% CI 1·42-1·67) and in the mefloquine (10 mg/kg) group was 2·34 (2·17-2·52), corresponding to a parasite clearance half-life of 9·4 h (8·7-10·2) and 6·2 h (5·7-6·7), respectively. The median minimum inhibitory concentration of DSM265 in blood was estimated as 1040 ng/mL (range 552-1500), resulting in a predicted single efficacious dose of 340 mg. Parasite clearance was significantly faster in participants who received mefloquine than in participants who received DSM265 (p<0·0001). INTERPRETATION The good safety profile, long elimination half-life, and antimalarial effect of DSM265 supports its development as a partner drug in a single-dose antimalarial combination treatment. FUNDING Wellcome Trust, UK Department for International Development, Global Health Innovative Technology Fund, Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Q-Pharm Pty Ltd, Herston, QLD, Australia.
| | | | | | | | | | | | | | | | | | | | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | | | | | - Maria Rosario
- Takeda Development Center Americas, Inc, Cambridge, MA, USA
| | | | | |
Collapse
|
33
|
Burrows JN, Duparc S, Gutteridge WE, Hooft van Huijsduijnen R, Kaszubska W, Macintyre F, Mazzuri S, Möhrle JJ, Wells TNC. New developments in anti-malarial target candidate and product profiles. Malar J 2017; 16:26. [PMID: 28086874 PMCID: PMC5237200 DOI: 10.1186/s12936-016-1675-x] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/30/2016] [Indexed: 11/10/2022] Open
Abstract
A decade of discovery and development of new anti-malarial medicines has led to a renewed focus on malaria elimination and eradication. Changes in the way new anti-malarial drugs are discovered and developed have led to a dramatic increase in the number and diversity of new molecules presently in pre-clinical and early clinical development. The twin challenges faced can be summarized by multi-drug resistant malaria from the Greater Mekong Sub-region, and the need to provide simplified medicines. This review lists changes in anti-malarial target candidate and target product profiles over the last 4 years. As well as new medicines to treat disease and prevent transmission, there has been increased focus on the longer term goal of finding new medicines for chemoprotection, potentially with long-acting molecules, or parenteral formulations. Other gaps in the malaria armamentarium, such as drugs to treat severe malaria and endectocides (that kill mosquitoes which feed on people who have taken the drug), are defined here. Ultimately the elimination of malaria requires medicines that are safe and well-tolerated to be used in vulnerable populations: in pregnancy, especially the first trimester, and in those suffering from malnutrition or co-infection with other pathogens. These updates reflect the maturing of an understanding of the key challenges in producing the next generation of medicines to control, eliminate and ultimately eradicate malaria.
Collapse
Affiliation(s)
- Jeremy N Burrows
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Stephan Duparc
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | | | | | - Wiweka Kaszubska
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Fiona Macintyre
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | | | - Jörg J Möhrle
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland
| | - Timothy N C Wells
- Medicines for Malaria Venture, Route de Pré Bois 20, 1215, Geneva 15, Switzerland.
| |
Collapse
|
34
|
McCarthy JS, Rückle T, Djeriou E, Cantalloube C, Ter-Minassian D, Baker M, O'Rourke P, Griffin P, Marquart L, Hooft van Huijsduijnen R, Möhrle JJ. A Phase II pilot trial to evaluate safety and efficacy of ferroquine against early Plasmodium falciparum in an induced blood-stage malaria infection study. Malar J 2016; 15:469. [PMID: 27624471 PMCID: PMC5022189 DOI: 10.1186/s12936-016-1511-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/31/2016] [Indexed: 01/13/2023] Open
Abstract
Background Ferroquine (SSR97193) is a candidate anti-malarial currently undergoing clinical trials for malaria. To better understand its pharmacokinetic (PK) and pharmacodynamic (PD) parameters the compound was tested in the experimentally induced blood stage malaria infection model in volunteers. Methods Male and non-pregnant female aged 18–50 years were screened for this phase II, controlled, single-centre clinical trial. Subjects were inoculated with ~1800 viable Plasmodium falciparum 3D7A-infected human erythrocytes, and treated with a single-dose of 800 mg ferroquine. Blood samples were taken at defined time-points to measure PK and PD parameters. The blood concentration of ferroquine and its active metabolite, SSR97213, were measured on dry blood spot samples by ultra-performance liquid chromatography with tandem mass spectrometry (LC-MS/MS). Parasitaemia and emergence of gametocytes were monitored by quantitative PCR. Safety was determined by recording adverse events and monitoring clinical laboratory assessments during the course of the study. Results Eight subjects were enrolled into the study, inoculated with infected erythrocytes and treated with 800 mg ferroquine. Ferroquine was rapidly absorbed with maximal exposure after 4–8 and 4–12 h exposure for SSR97213. Non-compartmental PK analysis resulted in estimates for half-lives of 10.9 and 23.8 days for ferroquine and SSR97213, respectively. Parasite clearance as reported by parasite reduction ratio was 162.9 (95 % CI 141–188) corresponding to a parasite clearance half-life of 6.5 h (95 % CI: 6.4–6.7 h). PK/PD modelling resulted in a predicted minimal parasiticidal concentration of 20 ng/mL, and the single dosing tested in this study was predicted to maintain an exposure above this threshold for 454 h (37.8 days). Although ferroquine was overall well tolerated, transient elevated transaminase levels were observed in three subjects. Paracetamol was the only concomitant treatment among the two out of these three subjects that may have played a role in the elevated transaminases levels. No clinically significant ECG abnormalities were observed. Conclusions The parameters and PK/PD model derived from this study pave the way to the further rational development of ferroquine as an anti-malarial partner drug. The safety of ferroquine has to be further explored in controlled human trials. Trial registration anzctr.org.au (registration number: ACTRN12613001040752), registered 18/09/2013 Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1511-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Thomas Rückle
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215, Meyrin, Geneva, Switzerland
| | - Elhadj Djeriou
- Sanofi Aventis Recherche Développement, Chilly-Mazarin, France
| | | | | | - Mark Baker
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215, Meyrin, Geneva, Switzerland.,Novartis Consumer Health SA, 2 route de l'Etraz, Case Postale 1279, 1260, Nyon, Switzerland
| | - Peter O'Rourke
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Paul Griffin
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.,University of Queensland, Brisbane, Australia.,Mater Health Services, Brisbane, Australia.,Q-Pharm Pty Ltd, Brisbane, Australia
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Jörg J Möhrle
- Medicines for Malaria Venture, Route de Pré-Bois 20, 1215, Meyrin, Geneva, Switzerland.
| |
Collapse
|
35
|
Krause A, Dingemanse J, Mathis A, Marquart L, Möhrle JJ, McCarthy JS. Pharmacokinetic/pharmacodynamic modelling of the antimalarial effect of Actelion-451840 in an induced blood stage malaria study in healthy subjects. Br J Clin Pharmacol 2016; 82:412-21. [PMID: 27062080 PMCID: PMC4972157 DOI: 10.1111/bcp.12962] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/10/2016] [Accepted: 03/30/2016] [Indexed: 01/22/2023] Open
Abstract
Aims The aim of this study was to use data from an experimental induced blood stage malaria clinical trial to characterize the antimalarial activity of the new compound Actelion‐451840 using pharmacokinetic/pharmacodynamic (PK/PD) modelling. Then, using simulations from the model, the dose and dosing regimen necessary to achieve cure of infection were derived. Methods Eight healthy male subjects were infected with blood stage P. falciparum. After 7 days, a single dose of 500 mg of Actelion‐451840 was administered under fed conditions. Parasite and drug concentrations were sampled frequently. Parasite growth and the relation to drug exposure were estimated using PK/PD modelling. Simulations were then undertaken to derive estimates of the likelihood of achieving cure in different scenarios. Results Actelion‐451840 was safe and well tolerated. Single dose treatment markedly reduced the level of P. falciparum parasitaemia, with a weighted average parasite reduction rate of 73.6 (95% CI 56.1, 96.5) and parasite clearance half‐life of 7.7 h (95% CI 7.3, 8.3). A two compartment PK/PD model with a steep concentration−kill effect predicted maximum effect with a sustained concentration of 10–15 ng ml−1 and cure achieved in 90% of subjects with six once daily doses of 300 mg once daily. Conclusions Actelion‐451840 shows clinical efficacy against P. falciparum infections. The PK/PD model developed from a single proof‐of‐concept study with eight healthy subjects enabled prediction of therapeutic effects, with cure rates with seven daily doses predicted to be equivalent to artesunate monotherapy. Larger doses or more frequent dosing are not predicted to achieve more rapid cure.
Collapse
Affiliation(s)
- Andreas Krause
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Jasper Dingemanse
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Alexandre Mathis
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Louise Marquart
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | |
Collapse
|