1
|
Lambert M, Jambon S, Bouhlel MA, Depauw S, Vrevin J, Blanck S, Marot G, Figeac M, Preudhomme C, Quesnel B, Boykin DW, David‐Cordonnier M. Induction of AML cell differentiation using HOXA9/DNA binding inhibitors as a potential therapeutic option for HOXA9-dependent AML. Hemasphere 2024; 8:e77. [PMID: 38716146 PMCID: PMC11072194 DOI: 10.1002/hem3.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/15/2024] [Accepted: 04/06/2024] [Indexed: 06/06/2024] Open
Abstract
The mainstay of acute myeloid leukemia (AML) treatment still relies on traditional chemotherapy, with a survival rate of approximately 30% for patients under 65 years of age and as low as 5% for those beyond. This unfavorable prognosis primarily stems from frequent relapses, resistance to chemotherapy, and limited approved targeted therapies for specific AML subtypes. Around 70% of all AML cases show overexpression of the transcription factor HOXA9, which is associated with a poor prognosis, increased chemoresistance, and higher relapse rates. However, direct targeting of HOXA9 in a clinical setting has not been achieved yet. The dysregulation caused by the leukemic HOXA9 transcription factor primarily results from its binding activity to DNA, leading to differentiation blockade. Our previous investigations have identified two HOXA9/DNA binding competitors, namely DB1055 and DB818. We assessed their antileukemic effects in comparison to HOXA9 knockdown or cytarabine treatment. Using human AML cell models, DB1055 and DB818 induced in vitro cell growth reduction, death, differentiation, and common transcriptomic deregulation but did not impact human CD34+ bone marrow cells. Furthermore, DB1055 and DB818 exhibited potent antileukemic activities in a human THP-1 AML in vivo model, leading to the differentiation of monocytes into macrophages. In vitro assays also demonstrated the efficacy of DB1055 and DB818 against AML blasts from patients, with DB1055 successfully reducing leukemia burden in patient-derived xenografts in NSG immunodeficient mice. Our findings indicate that inhibiting HOXA9/DNA interaction using DNA ligands may offer a novel differentiation therapy for the future treatment of AML patients dependent on HOXA9.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
- Université Sorbonne Paris NordBobignyFrance
| | - Samy Jambon
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - Mohamed A. Bouhlel
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - Sabine Depauw
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - Julie Vrevin
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - Samuel Blanck
- Univ. Lille, CHU Lille, ULR 2694—METRICSLilleFrance
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UAR 2014—PLBS, BililleLilleFrance
| | - Guillemette Marot
- Univ. Lille, CHU Lille, ULR 2694—METRICSLilleFrance
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UAR 2014—PLBS, BililleLilleFrance
- Inria, MODAL: Models for Data Analysis and LearningLilleFrance
| | - Martin Figeac
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, FranceLilleFrance
| | - Claude Preudhomme
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - David W. Boykin
- Department of ChemistryGeorgia State UniversityAtlantaGeorgiaUSA
| | - Marie‐Hélène David‐Cordonnier
- Univ. Lille, CNRS, Inserm, CHU Lille, IRCL, UMR9020‐U1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| |
Collapse
|
2
|
Venturelli A, Tagliazucchi L, Lima C, Venuti F, Malpezzi G, Magoulas GE, Santarem N, Calogeropoulou T, Cordeiro-da-Silva A, Costi MP. Current Treatments to Control African Trypanosomiasis and One Health Perspective. Microorganisms 2022; 10:microorganisms10071298. [PMID: 35889018 PMCID: PMC9321528 DOI: 10.3390/microorganisms10071298] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Human African Trypanosomiasis (HAT, sleeping sickness) and Animal African Trypanosomiasis (AAT) are neglected tropical diseases generally caused by the same etiological agent, Trypanosoma brucei. Despite important advances in the reduction or disappearance of HAT cases, AAT represents a risky reservoir of the infections. There is a strong need to control AAT, as is claimed by the European Commission in a recent document on the reservation of antimicrobials for human use. Control of AAT is considered part of the One Health approach established by the FAO program against African Trypanosomiasis. Under the umbrella of the One Health concepts, in this work, by analyzing the pharmacological properties of the therapeutic options against Trypanosoma brucei spp., we underline the need for clearer and more defined guidelines in the employment of drugs designed for HAT and AAT. Essential requirements are addressed to meet the challenge of drug use and drug resistance development. This approach shall avoid inter-species cross-resistance phenomena and retain drugs therapeutic activity.
Collapse
Affiliation(s)
- Alberto Venturelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - Lorenzo Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
- Doctorate School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Clara Lima
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Federica Venuti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - Giulia Malpezzi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
| | - George E. Magoulas
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (G.E.M.); (T.C.)
| | - Nuno Santarem
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Theodora Calogeropoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (G.E.M.); (T.C.)
| | - Anabela Cordeiro-da-Silva
- Host-Parasite Interactions Group, Institute of Research and Innovation in Health, University of Porto, 4099-002 Porto, Portugal; (C.L.); (N.S.); (A.C.-d.-S.)
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4099-002 Porto, Portugal
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.V.); (L.T.); (F.V.); (G.M.)
- Correspondence:
| |
Collapse
|
3
|
Dickie EA, Giordani F, Gould MK, Mäser P, Burri C, Mottram JC, Rao SPS, Barrett MP. New Drugs for Human African Trypanosomiasis: A Twenty First Century Success Story. Trop Med Infect Dis 2020; 5:tropicalmed5010029. [PMID: 32092897 PMCID: PMC7157223 DOI: 10.3390/tropicalmed5010029] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/23/2022] Open
Abstract
The twentieth century ended with human African trypanosomiasis (HAT) epidemics raging across many parts of Africa. Resistance to existing drugs was emerging, and many programs aiming to contain the disease had ground to a halt, given previous success against HAT and the competing priorities associated with other medical crises ravaging the continent. A series of dedicated interventions and the introduction of innovative routes to develop drugs, involving Product Development Partnerships, has led to a dramatic turnaround in the fight against HAT caused by Trypanosoma brucei gambiense. The World Health Organization have been able to optimize the use of existing tools to monitor and intervene in the disease. A promising new oral medication for stage 1 HAT, pafuramidine maleate, ultimately failed due to unforeseen toxicity issues. However, the clinical trials for this compound demonstrated the possibility of conducting such trials in the resource-poor settings of rural Africa. The Drugs for Neglected Disease initiative (DNDi), founded in 2003, has developed the first all oral therapy for both stage 1 and stage 2 HAT in fexinidazole. DNDi has also brought forward another oral therapy, acoziborole, potentially capable of curing both stage 1 and stage 2 disease in a single dosing. In this review article, we describe the remarkable successes in combating HAT through the twenty first century, bringing the prospect of the elimination of this disease into sight.
Collapse
Affiliation(s)
- Emily A. Dickie
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK; (E.A.D.); (F.G.); (M.K.G.)
| | - Federica Giordani
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK; (E.A.D.); (F.G.); (M.K.G.)
| | - Matthew K. Gould
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK; (E.A.D.); (F.G.); (M.K.G.)
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland; (P.M.); (C.B.)
| | - Christian Burri
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland; (P.M.); (C.B.)
- University of Basel, Petersplatz 1, 4000 Basel, Switzerland
| | - Jeremy C. Mottram
- York Biomedical Research Institute, Department of Biology, University of York, Wentworth Way, Heslington, York YO10 5DD, UK;
| | - Srinivasa P. S. Rao
- Novartis Institute for Tropical Diseases, 5300 Chiron Way, Emeryville, CA 94608, USA;
| | - Michael P. Barrett
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK; (E.A.D.); (F.G.); (M.K.G.)
- Correspondence:
| |
Collapse
|
4
|
Andrade CH, Neves BJ, Melo-Filho CC, Rodrigues J, Silva DC, Braga RC, Cravo PVL. In Silico Chemogenomics Drug Repositioning Strategies for Neglected Tropical Diseases. Curr Med Chem 2019. [DOI: 10.2174/0929867325666180309114824] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Only ~1% of all drug candidates against Neglected Tropical Diseases (NTDs)
have reached clinical trials in the last decades, underscoring the need for new, safe and effective
treatments. In such context, drug repositioning, which allows finding novel indications
for approved drugs whose pharmacokinetic and safety profiles are already known,
emerging as a promising strategy for tackling NTDs. Chemogenomics is a direct descendent
of the typical drug discovery process that involves the systematic screening of chemical
compounds against drug targets in high-throughput screening (HTS) efforts, for the identification
of lead compounds. However, different to the one-drug-one-target paradigm, chemogenomics
attempts to identify all potential ligands for all possible targets and diseases. In
this review, we summarize current methodological development efforts in drug repositioning
that use state-of-the-art computational ligand- and structure-based chemogenomics approaches.
Furthermore, we highlighted the recent progress in computational drug repositioning
for some NTDs, based on curation and modeling of genomic, biological, and chemical data.
Additionally, we also present in-house and other successful examples and suggest possible solutions
to existing pitfalls.
Collapse
Affiliation(s)
- Carolina Horta Andrade
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Bruno Junior Neves
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Cleber Camilo Melo-Filho
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Juliana Rodrigues
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Diego Cabral Silva
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Rodolpho Campos Braga
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Pedro Vitor Lemos Cravo
- Laboratory of Cheminformatics, Centro Universitario de Anapolis (UniEVANGELICA), Anapolis, GO, 75083-515, Brazil
| |
Collapse
|
5
|
Burri C, Yeramian PD, Allen JL, Merolle A, Serge KK, Mpanya A, Lutumba P, Mesu VKBK, Bilenge CMM, Lubaki JPF, Mpoto AM, Thompson M, Munungu BF, Manuel F, Josenando T, Bernhard SC, Olson CA, Blum J, Tidwell RR, Pohlig G. Efficacy, Safety, and Dose of Pafuramidine, a New Oral Drug for Treatment of First Stage Sleeping Sickness, in a Phase 2a Clinical Study and Phase 2b Randomized Clinical Studies. PLoS Negl Trop Dis 2016; 10:e0004362. [PMID: 26881924 PMCID: PMC4755713 DOI: 10.1371/journal.pntd.0004362] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/14/2015] [Indexed: 11/19/2022] Open
Abstract
Background Sleeping sickness (human African trypanosomiasis [HAT]) is caused by protozoan parasites and characterized by a chronic progressive course, which may last up to several years before death. We conducted two Phase 2 studies to determine the efficacy and safety of oral pafuramidine in African patients with first stage HAT. Methods The Phase 2a study was an open-label, non-controlled, proof-of-concept study where 32 patients were treated with 100 mg of pafuramidine orally twice a day (BID) for 5 days at two trypanosomiasis reference centers (Angola and the Democratic Republic of the Congo [DRC]) between August 2001 and November 2004. The Phase 2b study compared pafuramidine in 41 patients versus standard pentamidine therapy in 40 patients. The Phase 2b study was open-label, parallel-group, controlled, randomized, and conducted at two sites in the DRC between April 2003 and February 2007. The Phase 2b study was then amended to add an open-label sequence (Phase 2b-2), where 30 patients received pafuramidine for 10 days. The primary efficacy endpoint was parasitologic cure at 24 hours (Phase 2a) or 3 months (Phase 2b) after treatment completion. The primary safety outcome was the rate of occurrence of World Health Organization Toxicity Scale Grade 3 or higher adverse events. All subjects provided written informed consent. Findings/Conclusion Pafuramidine for the treatment of first stage HAT was comparable in efficacy to pentamidine after 10 days of dosing. The cure rates 3 months post-treatment were 79% in the 5-day pafuramidine, 100% in the 7-day pentamidine, and 93% in the 10-day pafuramidine groups. In Phase 2b, the percentage of patients with at least 1 treatment-emergent adverse event was notably higher after pentamidine treatment (93%) than pafuramidine treatment for 5 days (25%) and 10 days (57%). These results support continuation of the development program for pafuramidine into Phase 3. Sleeping sickness (human African trypanosomiasis [HAT]) is caused by parasites, and has a chronic progressive course that may last from several months to several years before death occurs. The present studies were done to assess the effectiveness and safety of oral pafuramidine versus intramuscular pentamidine (the standard treatment), in patients with first stage HAT. The results indicated that, several months after treatment, pafuramidine administered for 10 days was as effective as pentamidine administered for 7 days, and it had a better safety profile than pentamidine. With further study, pafuramidine could be a promising alternative for patients with first stage HAT. In addition, the design of the studies can be used a guide for future studies for identification and delivery of treatment to affected individuals in rural Africa.
Collapse
Affiliation(s)
- Christian Burri
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Patrick D Yeramian
- The Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
| | - James L Allen
- Immtech Pharmaceuticals, Inc., Vernon Hills, Illinois, United States of America
| | | | | | - Alain Mpanya
- Lisumbi Health Centre, Kinshasa, Democratic Republic of the Congo
| | - Pascal Lutumba
- Institut National de Recherche Biomédicale and Tropical Medicine Department, Kinshasa University, Kinshasa, Democratic Republic of the Congo
| | - Victor Kande Betu Ku Mesu
- Programme des Maladies Tropicales Négligées, Ministère de la Santé Publique Kinshasa, Democratic Republic of the Congo
| | | | | | - Alfred Mpoo Mpoto
- Hôspital Evangélique de Vanga, Vanga, Democratic Republic of the Congo
| | - Mark Thompson
- Federally Qualified Community Health Center, Elgin, Illinois, United States of America
| | | | - Francisco Manuel
- Instituto de Combate e de Controlo das Tripanossomíases, Luanda, Angola
| | | | - Sonja C Bernhard
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Carol A Olson
- Infectious Diseases, Global Product Development, PPD, Rockville, Maryland, United States of America
| | - Johannes Blum
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Richard R Tidwell
- University of North Carolina, Department of Pathology and Laboratory Medicine, School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Gabriele Pohlig
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Pohlig G, Bernhard SC, Blum J, Burri C, Mpanya A, Lubaki JPF, Mpoto AM, Munungu BF, N’tombe PM, Deo GKM, Mutantu PN, Kuikumbi FM, Mintwo AF, Munungi AK, Dala A, Macharia S, Bilenge CMM, Mesu VKBK, Franco JR, Dituvanga ND, Tidwell RR, Olson CA. Efficacy and Safety of Pafuramidine versus Pentamidine Maleate for Treatment of First Stage Sleeping Sickness in a Randomized, Comparator-Controlled, International Phase 3 Clinical Trial. PLoS Negl Trop Dis 2016; 10:e0004363. [PMID: 26882015 PMCID: PMC4755561 DOI: 10.1371/journal.pntd.0004363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/14/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Sleeping sickness (human African trypanosomiasis [HAT]) is a neglected tropical disease with limited treatment options that currently require parenteral administration. In previous studies, orally administered pafuramidine was well tolerated in healthy patients (for up to 21 days) and stage 1 HAT patients (for up to 10 days), and demonstrated efficacy comparable to pentamidine. METHODS This was a Phase 3, multi-center, randomized, open-label, parallel-group, active control study where 273 male and female patients with first stage Trypanosoma brucei gambiense HAT were treated at six sites: one trypanosomiasis reference center in Angola, one hospital in South Sudan, and four hospitals in the Democratic Republic of the Congo between August 2005 and September 2009 to support the registration of pafuramidine for treatment of first stage HAT in collaboration with the United States Food and Drug Administration. Patients were treated with either 100 mg of pafuramidine orally twice a day for 10 days or 4 mg/kg pentamidine intramuscularly once daily for 7 days to assess the efficacy and safety of pafuramidine versus pentamidine. Pregnant and lactating women as well as adolescents were included. The primary efficacy endpoint was the combined rate of clinical and parasitological cure at 12 months. The primary safety outcome was the frequency and severity of adverse events. The study was registered on the International Clinical Trials Registry Platform at www.clinicaltrials.gov with the number ISRCTN85534673. FINDINGS/CONCLUSIONS The overall cure rate at 12 months was 89% in the pafuramidine group and 95% in the pentamidine group; pafuramidine was non-inferior to pentamidine as the upper bound of the 95% confidence interval did not exceed 15%. The safety profile of pafuramidine was superior to pentamidine; however, 3 patients in the pafuramidine group had glomerulonephritis or nephropathy approximately 8 weeks post-treatment. Two of these events were judged as possibly related to pafuramidine. Despite good tolerability observed in preceding studies, the development program for pafuramidine was discontinued due to delayed post-treatment toxicity.
Collapse
Affiliation(s)
- Gabriele Pohlig
- Swiss Tropical and Public Health Institute, Pharmaceutical Medicine Unit, Swiss Centre for International Health, Basel, Switzerland
| | - Sonja C. Bernhard
- Swiss Tropical and Public Health Institute, Pharmaceutical Medicine Unit, Swiss Centre for International Health, Basel, Switzerland
- Pharmacy & Clinical Pharmacology at the Division of Clinical Pharmacology, University of Basel, Basel, Switzerland
| | - Johannes Blum
- Swiss Tropical and Public Health Institute, Medical Services and Diagnostic, Basel, Switzerland
| | - Christian Burri
- Pharmacy & Clinical Pharmacology at the Division of Clinical Pharmacology, University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Department of Medicines Research, Basel, Switzerland
| | - Alain Mpanya
- Programme Nationale de Lutte contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | | | | | | | | | | | - Pierre Nsele Mutantu
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Florent Mbo Kuikumbi
- Programme Nationale de Lutte contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | | | | | - Amadeu Dala
- Instituto de Combate e de Controlo das Tripanossomíases, Luanda, Angola
| | | | | | - Victor Kande Betu Ku Mesu
- Programme des Maladies Tropicales Négligées, Ministère de la Santé Publique Kinshasa, Democratic Republic of the Congo
| | - Jose Ramon Franco
- World Health Organisation Geneva, Department of Control of Neglected Diseases, Geneva, Switzerland
| | | | - Richard R. Tidwell
- University of North Carolina, Department of Pathology and Lab Medicine, School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Carol A. Olson
- Sapphire Oak Consultants, LLC, Lindenhurst, Illinois, United States of America
| |
Collapse
|
7
|
Abstract
Over the past 17 years, the number of reported cases of human African trypanosomiasis (HAT) has declined by over 90%, a significant result since the disease was highlighted as a public health problem by the WHO in 1995. However, if the goal of eliminating HAT by 2020 is to be achieved, then new treatments need to be identified and developed. A plethora of compound collections has been screened against Trypanosoma brucei spp, the etiological agents of HAT, resulting in three compounds progressing to clinical development. However, due to the high attrition rates in drug discovery, it is essential that research continues to identify novel molecules. Failure to do so, will result in the absence of molecules in the pipeline to fall back on should the current clinical trials be unsuccessful. This could seriously compromise control efforts to date, resulting in a resurgence in the number of HAT cases.
Collapse
Affiliation(s)
- Amy J Jones
- a Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Brisbane, 4111, Australia
| | - Vicky M Avery
- a Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Brisbane, 4111, Australia
| |
Collapse
|
8
|
Thuita JK, Wolf KK, Murilla GA, Bridges AS, Boykin DW, Mutuku JN, Liu Q, Jones SK, Gem CO, Ching S, Tidwell RR, Wang MZ, Paine MF, Brun R. Chemotherapy of second stage human African trypanosomiasis: comparison between the parenteral diamidine DB829 and its oral prodrug DB868 in vervet monkeys. PLoS Negl Trop Dis 2015; 9:e0003409. [PMID: 25654243 PMCID: PMC4318582 DOI: 10.1371/journal.pntd.0003409] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 11/12/2014] [Indexed: 11/18/2022] Open
Abstract
Human African trypanosomiasis (HAT, sleeping sickness) ranks among the most neglected tropical diseases based on limited availability of drugs that are safe and efficacious, particularly against the second stage (central nervous system [CNS]) of infection. In response to this largely unmet need for new treatments, the Consortium for Parasitic Drug Development developed novel parenteral diamidines and corresponding oral prodrugs that have shown cure of a murine model of second stage HAT. As a rationale for selection of one of these compounds for further development, the pharmacokinetics and efficacy of intramuscular (IM) active diamidine 2,5-bis(5-amidino-2-pyridyl)furan (DB829; CPD-0802) and oral prodrug2,5-bis[5-(N-methoxyamidino)-2-pyridyl]furan (DB868) were compared in the vervet monkey model of second stage HAT. Treatment was initiated 28 days post-infection of monkeys with T. b. rhodesiense KETRI 2537. Results showed that IM DB829 at 5 mg/kg/day for 5 consecutive days, 5 mg/kg/day every other day for 5 doses, or 2.5 mg/kg/day for 5 consecutive days cured all monkeys (5/5). Oral DB868 was less successful, with no cures (0/2) at 3 mg/kg/day for 10 days and cure rates of 1/4 at 10 mg/kg/day for 10 days and 20 mg/kg/day for 10 days; in total, only 2/10 monkeys were cured with DB868 dose regimens. The geometric mean plasma Cmax of IM DB829 at 5 mg/kg following the last of 5 doses was 25-fold greater than that after 10 daily oral doses of DB868 at 20 mg/kg. These data suggest that the active diamidine DB829, administered IM, should be considered for further development as a potential new treatment for second stage HAT. Treatment of human African trypanosomiasis (HAT, sleeping sickness) suffers from a shortage of medicines that are both effective, especially against the second (late) stage of the disease, and safe for patients. The development of new HAT medicines also has been significantly influenced by the perceived need for easily administered oral medicines to reduce the need for hospitalization of patients in resource-poor settings where HAT typically occurs. However, the clinical status of second stage patients is likely to dictate the need for their hospitalization, thus both oral and parenterally administered medicines would be utilised effectively. Therefore, in an effort to develop new medicines that meet efficacy and safety requirements, we evaluated a novel injectable diamidine 2,5-bis(5-amidino-2-pyridyl)furan (DB829; CPD-0802) and its oral prodrug formulation 2,5-bis[5-(N-methoxyamidino)-2-pyridyl]furan (DB868) in the vervet monkey model of second stage HAT. Treatment with either compound was initiated 28 days post-infection of monkeys with T. b. rhodesiense KETRI 2537. DB829 was dosed at 5 mg/kg/day for 5 consecutive days, 5 mg/kg/day every other day for 5 doses or 2.5 mg/kg/day for 5 consecutive days intramuscularly (IM) while DB868 was administered at 20, 10 or 3 mg/kg/day for 10 consecutive days orally. Clinical and parasitological monitoring was carried out for at least 300 days before the monkeys were declared cured. All IM DB829 and oral DB868 dose regimens were well tolerated. In addition, all monkeys (5/5) treated with IM DB829 were confirmed cured. In contrast, oral DB868 cured only 1/4 monkeys at either 10 or 20 mg/kg and did not cure any monkey when dosed at 3 mg/kg. These results indicate that IM DB829 is a suitable compound for further development as treatment for second stage HAT.
Collapse
Affiliation(s)
- John K. Thuita
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
- * E-mail: ,
| | - Kristina K. Wolf
- University of North Carolina Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Grace A. Murilla
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Arlene S. Bridges
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David W. Boykin
- Department of Chemistry, Georgia State University, Atlanta, Georgia, United States of America
| | - James N. Mutuku
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Qiang Liu
- University of North Carolina Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Susan K. Jones
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Charles O. Gem
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Shelley Ching
- SVC Associates, Inc., Apex, North Carolina, United States of America
| | - Richard R. Tidwell
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Michael Z. Wang
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas, United States of America
| | - Mary F. Paine
- University of North Carolina Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Reto Brun
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
9
|
Ríos Martínez CH, Lagartera L, Kaiser M, Dardonville C. Antiprotozoal activity and DNA binding of N-substituted N-phenylbenzamide and 1,3-diphenylurea bisguanidines. Eur J Med Chem 2014; 81:481-91. [PMID: 24865793 DOI: 10.1016/j.ejmech.2014.04.083] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 10/25/2022]
Abstract
Two series of N-alkyl, N-alkoxy, and N-hydroxy bisguanidines derived from the N-phenylbenzamide and 1,3-diphenylurea scaffolds were synthesised in three steps from the corresponding 4-amino-N-(4-aminophenyl)benzamide and 1,3-bis(4-aminophenyl)urea, respectively. All of the new compounds were evaluated in vitro against T. b. rhodesiense (STIB900) trypomastigotes and Plasmodium falciparum NF54 parasites (erythrocytic stage). N-alkoxy and N-hydroxy derivatives showed weak micromolar range IC50 values against T. b. rhodesiense and P. falciparum whereas the N-alkyl analogues displayed submicromolar and low nanomolar IC50 values against P. falciparum and Trypanosoma brucei, respectively. Two compounds, 4-(2-ethylguanidino)-N-(4-(2-ethylguanidino)phenyl)benzamide dihydrochloride (7b) and 4-(2-isopropylguanidino)-N-(4-(2-isopropylguanidino)phenyl)benzamide dihydrochloride (7c), which showed favourable drug-like properties and in vivo efficacy (100% cures) in the STIB900 mouse model of acute human African trypanosomiasis represent interesting leads for further in vivo studies. The binding of these compounds to AT-rich DNA was confirmed by surface plasmon resonance (SPR) biosensor experiments.
Collapse
Affiliation(s)
| | - Laura Lagartera
- Instituto de Química Médica, IQM-CSIC, Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland; University of Basel, Basel, Switzerland
| | | |
Collapse
|
10
|
Ju W, Yang S, Ansede JH, Stephens CE, Bridges AS, Voyksner RD, Ismail MA, Boykin DW, Tidwell RR, Hall JE, Wang MZ. CYP1A1 and CYP1B1-mediated biotransformation of the antitrypanosomal methamidoxime prodrug DB844 forms novel metabolites through intramolecular rearrangement. J Pharm Sci 2013; 103:337-49. [PMID: 24186380 DOI: 10.1002/jps.23765] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 09/03/2013] [Accepted: 10/10/2013] [Indexed: 12/18/2022]
Abstract
DB844 (CPD-594-12), N-methoxy-6-{5-[4-(N-methoxyamidino)phenyl]-furan-2-yl}-nicotinamidine, is an oral prodrug that has shown promising efficacy in both mouse and monkey models of second stage human African trypanosomiasis. However, gastrointestinal (GI) toxicity was observed with high doses in a vervet monkey safety study. In the current study, we compared the metabolism of DB844 by hepatic and extrahepatic cytochrome P450s to determine whether differences in metabolite formation underlie the observed GI toxicity. DB844 undergoes sequential O-demethylation and N-dehydroxylation in the liver to form the active compound DB820 (CPD-593-12). However, extrahepatic CYP1A1 and CYP1B1 produced two new metabolites, MX and MY. Accurate mass and collision-induced dissociation mass spectrometry analyses of the metabolites supported proposed structures of MX and MY. In addition, MY was confirmed with a synthetic standard and detection of nitric oxide (NO) release when DB844 was incubated with CYP1A1. Taken altogether, we propose that MX is formed by insertion of oxygen into the amidine CN to form an oxaziridine, which is followed by intramolecular rearrangement of the adjacent O-methyl group and subsequent release of NO. The resulting imine ester, MX, is further hydrolyzed to form MY. These findings may contribute to furthering the understanding of toxicities associated with benzamidoxime- and benzmethamidoxime-containing molecules.
Collapse
Affiliation(s)
- Wujian Ju
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, Kansas
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Thuita JK, Wolf KK, Murilla GA, Liu Q, Mutuku JN, Chen Y, Bridges AS, Mdachi RE, Ismail MA, Ching S, Boykin DW, Hall JE, Tidwell RR, Paine MF, Brun R, Wang MZ. Safety, pharmacokinetic, and efficacy studies of oral DB868 in a first stage vervet monkey model of human African trypanosomiasis. PLoS Negl Trop Dis 2013; 7:e2230. [PMID: 23755309 PMCID: PMC3674995 DOI: 10.1371/journal.pntd.0002230] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/10/2013] [Indexed: 11/19/2022] Open
Abstract
There are no oral drugs for human African trypanosomiasis (HAT, sleeping sickness). A successful oral drug would have the potential to reduce or eliminate the need for patient hospitalization, thus reducing healthcare costs of HAT. The development of oral medications is a key objective of the Consortium for Parasitic Drug Development (CPDD). In this study, we investigated the safety, pharmacokinetics, and efficacy of a new orally administered CPDD diamidine prodrug, 2,5-bis[5-(N-methoxyamidino)-2-pyridyl]furan (DB868; CPD-007-10), in the vervet monkey model of first stage HAT. DB868 was well tolerated at a dose up to 30 mg/kg/day for 10 days, a cumulative dose of 300 mg/kg. Mean plasma levels of biomarkers indicative of liver injury (alanine aminotransferase, aspartate aminotransferase) were not significantly altered by drug administration. In addition, no kidney-mediated alterations in creatinine and urea concentrations were detected. Pharmacokinetic analysis of plasma confirmed that DB868 was orally available and was converted to the active compound DB829 in both uninfected and infected monkeys. Treatment of infected monkeys with DB868 began 7 days post-infection. In the infected monkeys, DB829 attained a median Cmax (dosing regimen) that was 12-fold (3 mg/kg/day for 7 days), 15-fold (10 mg/kg/day for 7 days), and 31-fold (20 mg/kg/day for 5 days) greater than the IC50 (14 nmol/L) against T. b. rhodesiense STIB900. DB868 cured all infected monkeys, even at the lowest dose tested. In conclusion, oral DB868 cured monkeys with first stage HAT at a cumulative dose 14-fold lower than the maximum tolerated dose and should be considered a lead preclinical candidate in efforts to develop a safe, short course (5–7 days), oral regimen for first stage HAT. Development of orally administered medicines for human African trypanosomiasis (HAT) would potentially reduce the need for patient hospitalization, thus lowering healthcare costs. In this study, we investigated the potential of a novel diamidine prodrug, DB868 (CPD-007-10), as an oral treatment for first stage HAT. When administered to uninfected monkeys by oral gavage, DB868 was well tolerated up to a maximum dose of 30 mg/kg/day for 10 days (cumulative dose [CD] = 300 mg/kg). DB868 was absorbed into the systemic circulation and was converted to the active compound DB829 in concentrations that were potentially therapeutic for blood trypanosomes. Subsequently, DB868 was evaluated for efficacy in the first stage vervet monkey model of HAT in which treatment was initiated at 7 days post-infection with T. b. rhodesiense KETRI 2537. All infected monkeys were cured, even at the lowest of the three dose regimens tested: 3 mg/kg/day for 7 days (CD = 21 mg/kg), 10 mg/kg/day for 7 days (CD = 70 mg/kg) and 20 mg/kg/day for 5 days (CD = 100 mg/kg). DB868 conversion to DB829 was comparable between uninfected and infected monkeys. In view of its favourable safety and oral efficacy profile, we conclude that DB868 is a suitable candidate for development as a new treatment for first stage HAT.
Collapse
Affiliation(s)
- John K. Thuita
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (KARI-TRC), Kikuyu, Kenya
- University of Basel, Basel, Switzerland
| | - Kristina K. Wolf
- UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Grace A. Murilla
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (KARI-TRC), Kikuyu, Kenya
| | - Qiang Liu
- UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - James N. Mutuku
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (KARI-TRC), Kikuyu, Kenya
| | - Yao Chen
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas, United States of America
| | - Arlene S. Bridges
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Raymond E. Mdachi
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (KARI-TRC), Kikuyu, Kenya
| | - Mohamed A. Ismail
- Department of Chemistry, Georgia State University, Atlanta, Georgia, United States of America
| | - Shelley Ching
- SVC Associates, Inc., Apex, North Carolina, United States of America
| | - David W. Boykin
- Department of Chemistry, Georgia State University, Atlanta, Georgia, United States of America
| | - James Edwin Hall
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard R. Tidwell
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mary F. Paine
- UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Reto Brun
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Michael Zhuo Wang
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
12
|
Nanjunda R, Wilson WD. Binding to the DNA minor groove by heterocyclic dications: from AT-specific monomers to GC recognition with dimers. ACTA ACUST UNITED AC 2013; Chapter 8:Unit8.8. [PMID: 23255206 DOI: 10.1002/0471142700.nc0808s51] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Compounds that bind in the DNA minor groove have provided critical information on DNA molecular recognition, have found extensive uses in biotechnology, and are providing clinically useful drugs against diseases as diverse as cancer and sleeping sickness. This review focuses on the development of clinically useful heterocyclic diamidine minor groove binders. These compounds have shown us that the classical model for minor groove binding in AT DNA sequences must be expanded in several ways: compounds with nonstandard shapes can bind strongly to the groove, water can be directly incorporated into the minor groove complex in an interfacial interaction, and the compounds can form cooperative stacked dimers to recognize GC and mixed AT/GC base pair sequences.
Collapse
Affiliation(s)
- Rupesh Nanjunda
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | | |
Collapse
|
13
|
Ettari R, Tamborini L, Angelo IC, Micale N, Pinto A, De Micheli C, Conti P. Inhibition of Rhodesain as a Novel Therapeutic Modality for Human African Trypanosomiasis. J Med Chem 2013; 56:5637-58. [DOI: 10.1021/jm301424d] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Roberta Ettari
- Dipartimento
di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli
25, 20133 Milano, Italy
| | - Lucia Tamborini
- Dipartimento
di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli
25, 20133 Milano, Italy
| | - Ilenia C. Angelo
- Dipartimento di Scienze del
Farmaco e Prodotti per la Salute, Università degli Studi di Messina, Viale Annunziata, 98168 Messina, Italy
| | - Nicola Micale
- Dipartimento di Scienze del
Farmaco e Prodotti per la Salute, Università degli Studi di Messina, Viale Annunziata, 98168 Messina, Italy
| | - Andrea Pinto
- Dipartimento
di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli
25, 20133 Milano, Italy
| | - Carlo De Micheli
- Dipartimento
di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli
25, 20133 Milano, Italy
| | - Paola Conti
- Dipartimento
di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli
25, 20133 Milano, Italy
| |
Collapse
|
14
|
Novel amidines and analogues as promising agents against intracellular parasites: a systematic review. Parasitology 2013; 140:929-51. [PMID: 23561006 DOI: 10.1017/s0031182013000292] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Parasitic protozoa comprise diverse aetiological agents responsible for important diseases in humans and animals including sleeping sickness, Chagas disease, leishmaniasis, malaria, toxoplasmosis and others. They are major causes of mortality and morbidity in tropical and subtropical countries, and are also responsible for important economic losses. However, up to now, for most of these parasitic diseases, effective vaccines are lacking and the approved chemotherapeutic compounds present high toxicity, increasing resistance, limited efficacy and require long periods of treatment. Many of these parasitic illnesses predominantly affect low-income populations of developing countries for which new pharmaceutical alternatives are urgently needed. Thus, very low research funding is available. Amidine-containing compounds such as pentamidine are DNA minor groove binders with a broad spectrum of activities against human and veterinary pathogens. Due to their promising microbicidal activity but their rather poor bioavailability and high toxicity, many analogues and derivatives, including pro-drugs, have been synthesized and screened in vitro and in vivo in order to improve their selectivity and pharmacological properties. This review summarizes the knowledge on amidines and analogues with respect to their synthesis, pharmacological profile, mechanistic and biological effects upon a range of intracellular protozoan parasites. The bulk of these data may contribute to the future design and structure optimization of new aromatic dicationic compounds as novel antiparasitic drug candidates.
Collapse
|
15
|
Mäser P, Wittlin S, Rottmann M, Wenzler T, Kaiser M, Brun R. Antiparasitic agents: new drugs on the horizon. Curr Opin Pharmacol 2012; 12:562-6. [DOI: 10.1016/j.coph.2012.05.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/03/2012] [Indexed: 11/29/2022]
|
16
|
Thuita JK, Wang MZ, Kagira JM, Denton CL, Paine MF, Mdachi RE, Murilla GA, Ching S, Boykin DW, Tidwell RR, Hall JE, Brun R. Pharmacology of DB844, an orally active aza analogue of pafuramidine, in a monkey model of second stage human African trypanosomiasis. PLoS Negl Trop Dis 2012; 6:e1734. [PMID: 22848769 PMCID: PMC3404106 DOI: 10.1371/journal.pntd.0001734] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/04/2012] [Indexed: 11/18/2022] Open
Abstract
Novel drugs to treat human African trypanosomiasis (HAT) are still urgently needed despite the recent addition of nifurtimox-eflornithine combination therapy (NECT) to WHO Model Lists of Essential Medicines against second stage HAT, where parasites have invaded the central nervous system (CNS). The pharmacology of a potential orally available lead compound, N-methoxy-6-{5-[4-(N-methoxyamidino) phenyl]-furan-2-yl}-nicotinamidine (DB844), was evaluated in a vervet monkey model of second stage HAT, following promising results in mice. DB844 was administered orally to vervet monkeys, beginning 28 days post infection (DPI) with Trypanosoma brucei rhodesiense KETRI 2537. DB844 was absorbed and converted to the active metabolite 6-[5-(4-phenylamidinophenyl)-furanyl-2-yl]-nicotinamide (DB820), exhibiting plasma C(max) values of 430 and 190 nM for DB844 and DB820, respectively, after the 14th dose at 6 mg/kg qd. A 100-fold reduction in blood trypanosome counts was observed within 24 h of the third dose and, at the end of treatment evaluation performed four days post the last drug dose, trypanosomes were not detected in the blood or cerebrospinal fluid of any monkey. However, some animals relapsed during the 300 days of post treatment monitoring, resulting in a cure rate of 3/8 (37.5%) and 3/7 (42.9%) for the 5 mg/kg×10 days and the 6 mg/kg×14 days dose regimens respectively. These DB844 efficacy data were an improvement compared with pentamidine and pafuramidine both of which were previously shown to be non-curative in this model of CNS stage HAT. These data show that synthesis of novel diamidines with improved activity against CNS-stage HAT was possible.
Collapse
Affiliation(s)
- John K. Thuita
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Michael Z. Wang
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - John M. Kagira
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Cathrine L. Denton
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mary F. Paine
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Raymond E. Mdachi
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Grace A. Murilla
- Trypanosomiasis Research Centre, Kenya Agricultural Research Institute (TRC-KARI), Kikuyu, Kenya
| | - Shelley Ching
- SVC Associates, Inc., Apex, North Carolina, United States of America
| | - David W. Boykin
- Chemistry Department, Georgia State University, Atlanta, Georgia, United States of America
| | - Richard R. Tidwell
- Pathology Department, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, United States of America
| | - James E. Hall
- Pathology Department, School of Medicine, University of North Carolina at Chapel Hill, North Carolina, United States of America
| | - Reto Brun
- Swiss Tropical and Public Health Institute and University of Basel, Basel, Switzerland
| |
Collapse
|
17
|
Yan GZ, Generaux CN, Yoon M, Goldsmith RB, Tidwell RR, Hall JE, Olson CA, Clewell HJ, Brouwer KLR, Paine MF. A semiphysiologically based pharmacokinetic modeling approach to predict the dose-exposure relationship of an antiparasitic prodrug/active metabolite pair. Drug Metab Dispos 2011; 40:6-17. [PMID: 21953913 DOI: 10.1124/dmd.111.040063] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Dose selection during antiparasitic drug development in animal models and humans traditionally has relied on correlations between plasma concentrations obtained at or below maximally tolerated doses that are efficacious. The objective of this study was to improve the understanding of the relationship between dose and plasma/tissue exposure of the model antiparasitic agent, pafuramidine, using a semiphysiologically based pharmacokinetic (semi-PBPK) modeling approach. Preclinical and clinical data generated during the development of pafuramidine, a prodrug of the active metabolite, furamidine, were used. A whole-body semi-PBPK model for rats was developed based on a whole-liver PBPK model using rat isolated perfused liver data. A whole-body semi-PBPK model for humans was developed on the basis of the whole-body rat model. Scaling factors were calculated using metabolic and transport clearance data generated from rat and human sandwich-cultured hepatocytes. Both whole-body models described pafuramidine and furamidine disposition in plasma and predicted furamidine tissue (liver and kidney) exposure and excretion profiles (biliary and renal). The whole-body models predicted that the intestine contributes significantly (30-40%) to presystemic furamidine formation in both rats and humans. The predicted terminal elimination half-life of furamidine in plasma was 3- to 4-fold longer than that of pafuramidine in rats (170 versus 47 h) and humans (64 versus 19 h). The dose-plasma/tissue exposure relationship for the prodrug/active metabolite pair was determined using the whole-body models. The human model proposed a dose regimen of pafuramidine (40 mg once daily) based on a predefined efficacy-safety index. A similar approach could be used to guide dose-ranging studies in humans for next-in-class compounds.
Collapse
Affiliation(s)
- Grace Zhixia Yan
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7569, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
The trypanocidal activity of amidine compounds does not correlate with their binding affinity to Trypanosoma cruzi kinetoplast DNA. Antimicrob Agents Chemother 2011; 55:4765-73. [PMID: 21807972 DOI: 10.1128/aac.00229-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Due to limited efficacy and considerable toxicity, the therapy for Chagas' disease is far from being ideal, and thus new compounds are desirable. Diamidines and related compounds such as arylimidamides have promising trypanocidal activity against Trypanosoma cruzi. To better understand the mechanism of action of these heterocyclic cations, we investigated the kinetoplast DNA (kDNA) binding properties and trypanocidal efficacy against T. cruzi of 13 compounds. Four diamidines (DB75, DB569, DB1345, and DB829), eight arylimidamides (DB766, DB749, DB889, DB709, DB613, DB1831, DB1852, and DB2002), and one guanylhydrazone (DB1080) were assayed in thermal denaturation (T(m)) and circular dichroism (CD) studies using whole purified T. cruzi kDNA and a conserved synthetic parasite sequence. The overall CD spectra using the whole kDNA were similar to those found for the conserved sequence and were indicative of minor groove binding. Our findings showed that some of the compounds that exhibited the highest trypanocidal activities (e.g., DB766) caused low or no change in the T(m) measurements. However, while some active compounds, such as DB766, induced profound alterations of kDNA topology, others, like DB1831, although effective, did not result in altered T(m) and CD measurements. Our data suggest that the strong affinity of amidines with kDNA per se is not sufficient to generate and trigger their trypanocidal activity. Cell uptake differences and possibly distinct cellular targets need to be considered in the final evaluation of the mechanisms of action of these compounds.
Collapse
|
19
|
Brun R, Don R, Jacobs RT, Wang MZ, Barrett MP. Development of novel drugs for human African trypanosomiasis. Future Microbiol 2011; 6:677-91. [DOI: 10.2217/fmb.11.44] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Human African trypanosomiasis (HAT) or ‘sleeping sickness’ is a neglected tropical disease caused by the parasite Trypanosoma brucei. Novel models for funding pharmaceutical development against HAT are beginning to yield results. The Drugs for Neglected Diseases initiative (DNDi) rediscovered a nitroimidazole, fexinidazole, which is currently in Phase I clinical trials. Novel benzoxaboroles, discovered by Anacor, Scynexis and DNDi, have good pharmacokinetic properties in plasma and in the brain and are curative in a murine model of stage two HAT with brain infection. The Consortium for Parasitic Drug Development (CPDD) has identified a series of dicationic compounds that can cure a monkey model of stage two HAT. With other screening programs yielding hits, the pipeline for new HAT drugs might finally begin to fill.
Collapse
Affiliation(s)
- Reto Brun
- Department Medical Parasitology & Infection Biology, Swiss Tropical & Public Health Institute, and, University of Basel, CH-4002 Basel, Switzerland
| | - Robert Don
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Robert T Jacobs
- Department of Chemistry, SCYNEXIS, Inc., PO Box 12878, Research Triangle Park, NC, 27709-2878, USA
| | - Michael Zhuo Wang
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Michael P Barrett
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow, Scotland
| |
Collapse
|
20
|
Kotthaus J, Hungeling H, Reeh C, Kotthaus J, Schade D, Wein S, Wolffram S, Clement B. Synthesis and biological evaluation of l-valine-amidoximeesters as double prodrugs of amidines. Bioorg Med Chem 2011; 19:1907-14. [DOI: 10.1016/j.bmc.2011.01.066] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2010] [Revised: 01/28/2011] [Accepted: 01/31/2011] [Indexed: 11/26/2022]
|
21
|
|
22
|
Jacobs RT, Nare B, Phillips MA. State of the art in African trypanosome drug discovery. Curr Top Med Chem 2011; 11:1255-74. [PMID: 21401507 PMCID: PMC3101707 DOI: 10.2174/156802611795429167] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 11/25/2010] [Indexed: 11/22/2022]
Abstract
African sleeping sickness is endemic in sub-Saharan Africa where the WHO estimates that 60 million people are at risk for the disease. Human African trypanosomiasis (HAT) is 100% fatal if untreated and the current drug therapies have significant limitations due to toxicity and difficult treatment regimes. No new chemical agents have been approved since eflornithine in 1990. The pentamidine analog DB289, which was in late stage clinical trials for the treatment of early stage HAT recently failed due to toxicity issues. A new protocol for the treatment of late-stage T. brucei gambiense that uses combination nifurtomox/eflornithine (NECT) was recently shown to have better safety and efficacy than eflornithine alone, while being easier to administer. This breakthrough represents the only new therapy for HAT since the approval of eflornithine. A number of research programs are on going to exploit the unusual biochemical pathways in the parasite to identify new targets for target based drug discovery programs. HTS efforts are also underway to discover new chemical entities through whole organism screening approaches. A number of inhibitors with anti-trypanosomal activity have been identified by both approaches, but none of the programs are yet at the stage of identifying a preclinical candidate. This dire situation underscores the need for continued effort to identify new chemical agents for the treatment of HAT.
Collapse
Affiliation(s)
- Robert T. Jacobs
- SCYNEXIS, Inc., Research Triangle Park, North Carolina 27709-2878
| | - Bakela Nare
- SCYNEXIS, Inc., Research Triangle Park, North Carolina 27709-2878
| | - Margaret A. Phillips
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park Rd, Dallas, Texas 75390-9041
| |
Collapse
|
23
|
Nieto L, Mascaraque A, Miller F, Glacial F, Ríos Martínez C, Kaiser M, Brun R, Dardonville C. Synthesis and antiprotozoal activity of N-alkoxy analogues of the trypanocidal lead compound 4,4'-bis(imidazolinylamino)diphenylamine with improved human blood-brain barrier permeability. J Med Chem 2010; 54:485-94. [PMID: 21175162 DOI: 10.1021/jm101335q] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To improve the blood-brain barrier permeability of the trypanocidal lead compound 4,4'-bis(imidazolinylamino)diphenylamine (1), five N-alkoxy analogues were synthesized from bis(4-isothiocyanatophenyl)amine and N-alkoxy-N-(2-aminoethyl)-2-nitrobenzenesulfonamides following successive chemical reactions in just one reactor ("one-pot procedure"). This involved: (a) formation of a thiourea intermediate, (b) removal of the amine protecting groups, and (c) intramolecular cyclization. The blood-brain barrier permeability of the compounds determined in vitro by transport assays through the hCMEC/D3 human cell line, a well-known and characterized human cellular blood-brain barrier model, showed that the N-hydroxy analogue 16 had enhanced blood-brain barrier permeability compared with the unsubstituted lead compound. Moreover, this compound displayed low micromolar IC(50) against Trypanosoma brucei rhodesiense and Plasmodium falciparum and moderate activity by intraperitoneal administration in the STIB900 murine model of acute sleeping sickness.
Collapse
Affiliation(s)
- Lidia Nieto
- Instituto de Química Médica, CSIC, Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Mallari JP, Zhu F, Lemoff A, Kaiser M, Lu M, Brun R, Guy RK. Optimization of purine-nitrile TbcatB inhibitors for use in vivo and evaluation of efficacy in murine models. Bioorg Med Chem 2010; 18:8302-9. [DOI: 10.1016/j.bmc.2010.09.073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/24/2010] [Accepted: 09/30/2010] [Indexed: 11/25/2022]
|
25
|
Goldsmith RB, Gray DR, Yan Z, Generaux CN, Tidwell RR, Reisner HM. Application of monoclonal antibodies to measure metabolism of an anti-trypanosomal compound in vitro and in vivo. J Clin Lab Anal 2010; 24:187-94. [PMID: 20486201 PMCID: PMC6647651 DOI: 10.1002/jcla.20380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human African trypanosomiasis (HAT), also called African sleeping sickness, is a neglected tropical parasitic disease indigenous to sub-Saharan Africa. Diamidine compounds, including pentamidine and CPD-0801, are potent anti-trypanosomal molecules. The latter is a potential drug in the development at the UNC based Consortium for Parasitic Drug Development. An orally bioavailable prodrug of CPD-0801, DB868, is metabolized primarily in the liver to the active form. A monoclonal antibody developed against a pentamidine derivative has shown significant reactivity with CPD-0801 (EC(50) 65.1 nM), but not with the prodrug (EC(50)>18,000 nM). An inhibitory enzyme-linked immunosorbent assay (IELISA) has been used to quantitatively monitor prodrug metabolism by detecting the production of the active compound over time in a sandwich culture rat hepatocyte system and in rats. These results were compared with the results of the standard LC/MS/MS assay. Spearman coefficients of 0.96 and 0.933 (in vitro and in vivo, respectively) indicate a high correlation between these two measurement methods. This novel IELISA provides a facile, inexpensive, and accurate method for drug detection that may aide in elucidating the mechanisms of action and toxicity of existing and future diamidine compounds.
Collapse
Affiliation(s)
- Rachel Beaulieu Goldsmith
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Danny R. Gray
- Center for Delivery of Molecules and Cells, Department of Biomedical Engineering, Case Western Reserve, Cleveland, Ohio
| | - Zhixia Yan
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Claudia N. Generaux
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Richard R. Tidwell
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Howard M. Reisner
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
26
|
Jacobs RT, Ding C. Recent Advances in Drug Discovery for Neglected Tropical Diseases Caused by Infective Kinetoplastid Parasites. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2010. [DOI: 10.1016/s0065-7743(10)45017-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
27
|
New treatment option for second-stage African sleeping sickness: in vitro and in vivo efficacy of aza analogs of DB289. Antimicrob Agents Chemother 2009; 53:4185-92. [PMID: 19620327 DOI: 10.1128/aac.00225-09] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
African sleeping sickness is a fatal parasitic disease, and all drugs currently in use for treatment have strong liabilities. It is essential to find new, effective, and less toxic drugs, ideally with oral application, to control the disease. In this study, the aromatic diamidine DB75 (furamidine) and two aza analogs, DB820 and DB829 (CPD-0801), as well as their methoxyamidine prodrugs and amidoxime metabolites, were evaluated against African trypanosomes. The active parent diamidines showed similar in vitro profiles against different Trypanosoma brucei strains, melarsoprol- and pentamidine-resistant lines, and a P2 transporter knockout strain (AT1KO), with DB75 as the most trypanocidal molecule. In the T. b. rhodesiense strain STIB900 acute mouse model, the aza analogs DB820 and DB829 demonstrated activities superior to that of DB75. The aza prodrugs DB844 and DB868, as well as two metabolites of DB844, were orally more potent in the T. b. brucei strain GVR35 mouse central nervous system (CNS) model than DB289 (pafuramidine maleate). Unexpectedly, the parent diamidine DB829 showed high activity in the mouse CNS model by the intraperitoneal route. In conclusion, DB868 with oral and DB829 with parenteral application are potential candidates for further development of a second-stage African sleeping sickness drug.
Collapse
|
28
|
Cavalli A, Bolognesi ML. Neglected Tropical Diseases: Multi-Target-Directed Ligands in the Search for Novel Lead Candidates against Trypanosoma and Leishmania. J Med Chem 2009; 52:7339-59. [DOI: 10.1021/jm9004835] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Andrea Cavalli
- Department of Pharmaceutical Sciences, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
- Department of Drug Discovery and Development, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| | - Maria Laura Bolognesi
- Department of Pharmaceutical Sciences, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|