Mechanistic Studies of Anti-Leishmanial Arylimidamides

Total Page:16

File Type:pdf, Size:1020Kb

Mechanistic Studies of Anti-Leishmanial Arylimidamides MECHANISTIC STUDIES OF ANTI-LEISHMANIAL ARYLIMIDAMIDES DISSERTATION Presented in Partial Fulfillments of the Requirements for the Degree Doctor of Philosophy from the Graduate School of The Ohio State University By Trupti Pandharkar, M.S. ****** Graduate Program in Medicinal and Pharmaceutical Chemistry The Ohio State University 2012 Dissertation Committee: Karl A. Werbovetz, Ph.D., Advisor Esperanza Carcache de Blanco, Ph.D. Mark E. Drew, Ph.D. Juan D. Alfonzo, PhD. Copyright by Trupti Pandharkar 2012 ABSTRACT Leishmaniasis is a neglected tropical disease caused by protozoan parasites of the genus Leishmania . With the estimated global incidences of 0.7 to 1.2 million for CL cases and 0.2 to 0.4 million VL cases cases per year, leishmaniasis is estimated to cause the ninth largest burden of disease. In the absence of effective treatment, visceral leishmaniasis is most often fatal and cutaneous and other forms of this disease often result in severe disfigurement and can be debilitating. Given the issues with existing treatment options, like toxicity, prohibitive cost and loss of effectiveness due to emergence of drug resistant strains, the need for the new drugs is urgent. With the mission to develop cheaper, safer and more efficacious drugs, the Consortium for Parasitic Drug Development (CPDD) has synthesized and evaluated series of diamidine analogs for their anti-parasitic activity against several pathogens including Leishmania. Medicinal chemistry efforts to improve the efficacy of diamidines resulted in discovery of new class of molecules called ‘arylimidamides (AIAs)’ with extraordinary activity, especially against intracellular pathogens like T. cruzi and Leishmania . The anti- leishmanial efficacy of an AIA, DB766 (2,5-bis[2-(2-propoxy)-4-(2- ii pyridylimino)aminophenyl]furan hydrochloride) that displayed outstanding activity against the intracellular form of Leishmania donovani [IC 50 = 0.036 µM] and oral efficacy in murine and hamster models of visceral leishmaniasis [71% and 89% reduction in liver parasitemia at 100 mg/kg/day × 5, respectively] was recently reported. Despite intensive lead optimization efforts that permitted exhaustive analysis of the AIA structure activity relationship, attempts to improve the efficacy of AIAs have met with limited success. The lack of knowledge about the parasite drug target and the host toxicity mechanism has precluded further pre-clinical development of AIAs. In the present study, we employed three different approaches, 1) a 2 dimensional difference in gel electrophoresis-mass spectrometry (2D-DiGE-MS) assisted comparative proteomics analysis to study changes in the Leishmania proteome post-treatment with the lead AIA- DB766, 2) transmission electron microscopy to study the ultrastructural alterations caused by DB766 treatment in Leishmania and 3) generation and characterization of a Leishmania cell line that is over 10-fold resistant to DB766 through stepwise increases in the concentration of the compound to identify the target and understand the anti- leishmanial mechanism of action of AIAs to facilitate future anti-leishmanial drug discovery efforts. The DiGE approach used in this study led to the identification of 19 proteins that were differentially modulated in DB766 treated Leishmania. Most of the downregulated proteins like RNA helicase, mitochondrial tryparedoxin peroxidase (mTXNPx), HSP60, ATP synthase and ATPase were nuclear encoded mitochondrial proteins and four iii upregulated proteins corresponding to HSP60 were mitochondrial precursors, leading to the evaluation and confirmation of mitochondrial involvement in response to DB766 treatment in Leishmania . However, in the absence of the specific target information and considering the fact that mitochondrial involvement could be a secondary event downstream of a primary target, more detailed investigation employing other approaches was undertaken. Comparison of the ultrastructural effects of AIAs with the diamidine DB1111 in Leishmania donovani axenic amastigotes indicated that, unlike DB1111, DB766 treatment did not result in any change in the mitochondrial morphology of these parasites. However, dramatic ultrastructural alterations were noted in other organelles. The similarities in the ultrastructural profile induced by DB766 to those produced by sterol biosynthesis inhibitors and protease inhibitors in Leishmania led to evaluation of hypotheses that AIAs act via protease inhibition and/or disturbances in sterol metabolism in Leishmania . In fluoregenic serine protease substrate based assays, DB766 failed to show marked inhibition of oligopeptidase B like enzyme activity in Leishmania lysates, leading to cessation of its pursuit as a target of the AIAs. However, GC-MS analysis of Leishmania sterols indicated important changes in the sterol profile of DB766 treated Leishmania, consistent with the ‘disturbances in sterol metabolism’ hypothesis . In an attempt to identify mutations that cause resistance and in the hope of obtaining additional mechanistic information, a L. donovani axenic amastigotes cell line over 10- iv fold resistant to DB766 was developed and characterized. In vitro susceptibility assays revealed that these DB766 resistant parasites (766R) are hypersensitive to miltefosine (over 2-fold more sensitive) and anti-fungal azoles (over 1000-fold more sensitive). Systematic studies to test the hypothesis that resistance to DB766 is associated with perturbations in Leishmania sterol metabolism and enzymes in the sterol biosynthetic pathway were undertaken. Western blot analysis of 766R parasites indicated dramatically reduced expression of CYP5122A1, a recently identified cytochrome P450 associated with ergosterol metabolism in Leishmania , in the DB766 resistant parasites. GC-MS analysis of sterols extracted from DB766 sensitive and resistant parasites indicated that the reduced expression of CYP5122A1 was associated with changes in the levels of sterol intermediates without affecting the ergosterol content. Susceptibility assays demonstrated that CYP5122A1 single knockout (HKO) Leishmania donovani promastigotes are significantly less susceptible to DB766 and more susceptible to ketoconazole than their wild type counterparts, consistent with the observations in DB766 resistant parasites. Our studies demonstrate that 1) DB766 disrupts sterol metabolism in Leishmania and synergizes the anti-leishmanial activity of posaconazole 2) CYP5122A1 plays an important role in governing susceptibility and resistance to DB766 in this parasite, and 3) CYP5122A1 also modulates the susceptibility of Leishmania to antifungal azoles. These results support our hypothesis that DB766 and other AIAs disrupt sterol metabolism by targeting CYP5122A1, in Leishmania. v DEDICATED TO MY DAUGHTER AMBAR, HUSBAND ROHIT TIWARI AND MY PARENTS, DILIP AND SANDHYA PANDHARKAR vi ACKNOWLEDGMENTS The pursuit of my PhD has been a long journey. It wouldn’t have been possible for me to complete my studies without invaluable help and support from numerous people during this journey. Beginning with my advisor, I would like to give my special thanks to Karl Werbovetz for providing me with intellectual insights, guidance and encouragement throughout my graduate career. If not for his patience with me, faith in me, flexibility, genuine care and concern, I would not have been able to attend to demands of life outside the scientific career. He has never judged me and has always gently encouraged me, knowing that I need to juggle priorities. I am also grateful to Richard Burchmore, University of Glasgow, UK, for his help and discussion with the initial proteomics experiment. I also sincerely appreciate Frederick Buckner and members of his group at University of Washington, Seattle, for their help with the GC-MS analysis of Leishmania sterol samples and for providing me anti-CYP51 antibody for Western blot analysis. I thank Chandrima Shaha and members of her group at National Institute of Immunology, India for their help with drug susceptibility studies with genetically modified Leishmania cell line and also for providing me with anti- CYP5122A1 antibody for Western blot analysis and CYP5122A1 expression plasmid for future studies. I am also grateful to Mark Drew and members of his group for helping me with flow cytometry, Robert Curley for helping with GC instrumentation, Werner Tjarks and members of his group for helping me with HPLC instrumentation and Thomas Schmittgen and members of his group for helping me with Real Time PCR assays. vii I am incredibly grateful for the financial support from the Bill and Melinda Gates Foundation. I am also grateful for scientific feedbacks and invaluable insights from members of my academic committee: Pui-Kai (Tom) Li, Esperanza Carcache de Blanco, Mark Drew and Juan Alfonzo. I also extend my heartfelt gratitude to my fellow graduate students and post docs in both Werbovetz and Li labs, especially Xiaohua, Carolyn, Sihui, Molla, Julian, Shanshan, Jason, Bulbul, Deepak, Nick, Som, Jonathan and Justin. Their presence helped to lighten my stress and keep the lab environment fun and sane. I have a learned a lot from all of them during the humorous marathon discussions that we had on topics like science, politics, culture, spirituality and many more. I also give my special thanks to Swati Dhar and Dakshayini Rao for playing the part of a friend and confidante and providing support and care. In addition, I give my sincere thanks to people for their assistance with some of the experiments described in this dissertation:
Recommended publications
  • (12) United States Patent (10) Patent No.: US 6,395,889 B1 Robison (45) Date of Patent: May 28, 2002
    USOO6395889B1 (12) United States Patent (10) Patent No.: US 6,395,889 B1 Robison (45) Date of Patent: May 28, 2002 (54) NUCLEIC ACID MOLECULES ENCODING WO WO-98/56804 A1 * 12/1998 ........... CO7H/21/02 HUMAN PROTEASE HOMOLOGS WO WO-99/0785.0 A1 * 2/1999 ... C12N/15/12 WO WO-99/37660 A1 * 7/1999 ........... CO7H/21/04 (75) Inventor: fish E. Robison, Wilmington, MA OTHER PUBLICATIONS Vazquez, F., et al., 1999, “METH-1, a human ortholog of (73) Assignee: Millennium Pharmaceuticals, Inc., ADAMTS-1, and METH-2 are members of a new family of Cambridge, MA (US) proteins with angio-inhibitory activity', The Journal of c: - 0 Biological Chemistry, vol. 274, No. 33, pp. 23349–23357.* (*) Notice: Subject to any disclaimer, the term of this Descriptors of Protease Classes in Prosite and Pfam Data patent is extended or adjusted under 35 bases. U.S.C. 154(b) by 0 days. * cited by examiner (21) Appl. No.: 09/392, 184 Primary Examiner Ponnathapu Achutamurthy (22) Filed: Sep. 9, 1999 ASSistant Examiner William W. Moore (51) Int. Cl." C12N 15/57; C12N 15/12; (74) Attorney, Agent, or Firm-Alston & Bird LLP C12N 9/64; C12N 15/79 (57) ABSTRACT (52) U.S. Cl. .................... 536/23.2; 536/23.5; 435/69.1; 435/252.3; 435/320.1 The invention relates to polynucleotides encoding newly (58) Field of Search ............................... 536,232,235. identified protease homologs. The invention also relates to 435/6, 226, 69.1, 252.3 the proteases. The invention further relates to methods using s s s/ - - -us the protease polypeptides and polynucleotides as a target for (56) References Cited diagnosis and treatment in protease-mediated disorders.
    [Show full text]
  • A Multifaceted Approach to Combating Leishmaniasis, a Neglected Tropical Disease
    OLD TARGETS AND NEW BEGINNINGS: A MULTIFACETED APPROACH TO COMBATING LEISHMANIASIS, A NEGLECTED TROPICAL DISEASE DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy from the Graduate School of The Ohio State University By Adam Joseph Yakovich, B.S. ***** The Ohio State University 2007 Dissertation Committee: Karl A Werbovetz, Ph.D., Advisor Approved by Pui-Kai Li, Ph.D. Werner Tjarks, Ph.D. ___________________ Ching-Shih Chen, Ph.D Advisor Graduate Program In Pharmacy ABSTRACT Leishmaniasis, a broad spectrum of disease which is caused by the protozoan parasite Leishmania , currently affects 12 million people in 88 countries worldwide. There are over 2 million of new cases of leishmaniasis occurring annually. Clinical manifestations of leishmaniasis range from potentially disfiguring cutaneous leishmaniasis to the most severe manifestation, visceral leishmaniasis, which attacks the reticuloendothelial system and has a fatality rate near 100% if left untreated. All currently available therapies all suffer from drawbacks including expense, route of administration and developing resistance. In the laboratory of Dr. Karl Werbovetz our primary goal is the identification and development of an inexpensive, orally available antileishmanial chemotherapeutic agent. Previous efforts in the lab have identified a series of dinitroaniline compounds which have promising in vitro activity in inhibiting the growth of Leishmania parasites. It has since been discovered that these compounds exert their antileishmanial effects by binding to tubulin and inhibiting polymerization. Remarkably, although mammalian and Leishmania tubulins are ~84 % identical, the dinitroaniline compounds show no effect on mammalian tubulin at concentrations greater than 10-fold the IC 50 value determined for inhibiting Leishmania tubulin ii polymerization.
    [Show full text]
  • Supplementary Information
    Supplementary Information Network-based Drug Repurposing for Novel Coronavirus 2019-nCoV Yadi Zhou1,#, Yuan Hou1,#, Jiayu Shen1, Yin Huang1, William Martin1, Feixiong Cheng1-3,* 1Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA 2Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA 3Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA #Equal contribution *Correspondence to: Feixiong Cheng, PhD Lerner Research Institute Cleveland Clinic Tel: +1-216-444-7654; Fax: +1-216-636-0009 Email: [email protected] Supplementary Table S1. Genome information of 15 coronaviruses used for phylogenetic analyses. Supplementary Table S2. Protein sequence identities across 5 protein regions in 15 coronaviruses. Supplementary Table S3. HCoV-associated host proteins with references. Supplementary Table S4. Repurposable drugs predicted by network-based approaches. Supplementary Table S5. Network proximity results for 2,938 drugs against pan-human coronavirus (CoV) and individual CoVs. Supplementary Table S6. Network-predicted drug combinations for all the drug pairs from the top 16 high-confidence repurposable drugs. 1 Supplementary Table S1. Genome information of 15 coronaviruses used for phylogenetic analyses. GenBank ID Coronavirus Identity % Host Location discovered MN908947 2019-nCoV[Wuhan-Hu-1] 100 Human China MN938384 2019-nCoV[HKU-SZ-002a] 99.99 Human China MN975262
    [Show full text]
  • Rediscovery of Fexinidazole
    New Drugs against Trypanosomatid Parasites: Rediscovery of Fexinidazole INAUGURALDISSERTATION zur Erlangung der Würde eines Doktors der Philosophie vorgelegt der Philosophisch-Naturwissenschaftlichen Fakultät der Universität Basel von Marcel Kaiser aus Obermumpf, Aargau Basel, 2014 Originaldokument gespeichert auf dem Dokumentenserver der Universität Basel edoc.unibas.ch Dieses Werk ist unter dem Vertrag „Creative Commons Namensnennung-Keine kommerzielle Nutzung-Keine Bearbeitung 3.0 Schweiz“ (CC BY-NC-ND 3.0 CH) lizenziert. Die vollständige Lizenz kann unter creativecommons.org/licenses/by-nc-nd/3.0/ch/ eingesehen werden. 1 Genehmigt von der Philosophisch-Naturwissenschaftlichen Fakultät der Universität Basel auf Antrag von Prof. Reto Brun, Prof. Simon Croft Basel, den 10. Dezember 2013 Prof. Dr. Jörg Schibler, Dekan 2 3 Table of Contents Acknowledgement .............................................................................................. 5 Summary ............................................................................................................ 6 Zusammenfassung .............................................................................................. 8 CHAPTER 1: General introduction ................................................................. 10 CHAPTER 2: Fexinidazole - A New Oral Nitroimidazole Drug Candidate Entering Clinical Development for the Treatment of Sleeping Sickness ........ 26 CHAPTER 3: Anti-trypanosomal activity of Fexinidazole – A New Oral Nitroimidazole Drug Candidate for the Treatment
    [Show full text]
  • Australian Public Assessment Report for Tafenoquine (As Succinate)
    Australian Public Assessment Report for Tafenoquine (as succinate) Proprietary Product Name: Kozenis Sponsor: GlaxoSmithKline Australia Pty Ltd November 2018 Therapeutic Goods Administration About the Therapeutic Goods Administration (TGA) • The Therapeutic Goods Administration (TGA) is part of the Australian Government Department of Health and is responsible for regulating medicines and medical devices. • The TGA administers the Therapeutic Goods Act 1989 (the Act), applying a risk management approach designed to ensure therapeutic goods supplied in Australia meet acceptable standards of quality, safety and efficacy (performance) when necessary. • The work of the TGA is based on applying scientific and clinical expertise to decision- making, to ensure that the benefits to consumers outweigh any risks associated with the use of medicines and medical devices. • The TGA relies on the public, healthcare professionals and industry to report problems with medicines or medical devices. TGA investigates reports received by it to determine any necessary regulatory action. • To report a problem with a medicine or medical device, please see the information on the TGA website <https://www.tga.gov.au>. About AusPARs • An Australian Public Assessment Report (AusPAR) provides information about the evaluation of a prescription medicine and the considerations that led the TGA to approve or not approve a prescription medicine submission. • AusPARs are prepared and published by the TGA. • An AusPAR is prepared for submissions that relate to new chemical entities, generic medicines, major variations and extensions of indications. • An AusPAR is a static document; it provides information that relates to a submission at a particular point in time. • A new AusPAR will be developed to reflect changes to indications and/or major variations to a prescription medicine subject to evaluation by the TGA.
    [Show full text]
  • Serine Proteases with Altered Sensitivity to Activity-Modulating
    (19) & (11) EP 2 045 321 A2 (12) EUROPEAN PATENT APPLICATION (43) Date of publication: (51) Int Cl.: 08.04.2009 Bulletin 2009/15 C12N 9/00 (2006.01) C12N 15/00 (2006.01) C12Q 1/37 (2006.01) (21) Application number: 09150549.5 (22) Date of filing: 26.05.2006 (84) Designated Contracting States: • Haupts, Ulrich AT BE BG CH CY CZ DE DK EE ES FI FR GB GR 51519 Odenthal (DE) HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI • Coco, Wayne SK TR 50737 Köln (DE) •Tebbe, Jan (30) Priority: 27.05.2005 EP 05104543 50733 Köln (DE) • Votsmeier, Christian (62) Document number(s) of the earlier application(s) in 50259 Pulheim (DE) accordance with Art. 76 EPC: • Scheidig, Andreas 06763303.2 / 1 883 696 50823 Köln (DE) (71) Applicant: Direvo Biotech AG (74) Representative: von Kreisler Selting Werner 50829 Köln (DE) Patentanwälte P.O. Box 10 22 41 (72) Inventors: 50462 Köln (DE) • Koltermann, André 82057 Icking (DE) Remarks: • Kettling, Ulrich This application was filed on 14-01-2009 as a 81477 München (DE) divisional application to the application mentioned under INID code 62. (54) Serine proteases with altered sensitivity to activity-modulating substances (57) The present invention provides variants of ser- screening of the library in the presence of one or several ine proteases of the S1 class with altered sensitivity to activity-modulating substances, selection of variants with one or more activity-modulating substances. A method altered sensitivity to one or several activity-modulating for the generation of such proteases is disclosed, com- substances and isolation of those polynucleotide se- prising the provision of a protease library encoding poly- quences that encode for the selected variants.
    [Show full text]
  • Injectable Anti-Malarials Revisited: Discovery and Development of New Agents to Protect Against Malaria
    Macintyre et al. Malar J (2018) 17:402 https://doi.org/10.1186/s12936-018-2549-1 Malaria Journal REVIEW Open Access Injectable anti‑malarials revisited: discovery and development of new agents to protect against malaria Fiona Macintyre1, Hanu Ramachandruni1, Jeremy N. Burrows1, René Holm2,3, Anna Thomas1, Jörg J. Möhrle1, Stephan Duparc1, Rob Hooft van Huijsduijnen1 , Brian Greenwood4, Winston E. Gutteridge1, Timothy N. C. Wells1* and Wiweka Kaszubska1 Abstract Over the last 15 years, the majority of malaria drug discovery and development eforts have focused on new mol- ecules and regimens to treat patients with uncomplicated or severe disease. In addition, a number of new molecular scafolds have been discovered which block the replication of the parasite in the liver, ofering the possibility of new tools for oral prophylaxis or chemoprotection, potentially with once-weekly dosing. However, an intervention which requires less frequent administration than this would be a key tool for the control and elimination of malaria. Recent progress in HIV drug discovery has shown that small molecules can be formulated for injections as native molecules or pro-drugs which provide protection for at least 2 months. Advances in antibody engineering ofer an alternative approach whereby a single injection could potentially provide protection for several months. Building on earlier profles for uncomplicated and severe malaria, a target product profle is proposed here for an injectable medicine providing long-term protection from this disease. As with all of such profles, factors such as efcacy, cost, safety and tolerability are key, but with the changing disease landscape in Africa, new clinical and regulatory approaches are required to develop prophylactic/chemoprotective medicines.
    [Show full text]
  • Data Sheet of Clinical Trial C.T
    DATA SHEET OF CLINICAL TRIAL C.T. No 048-14 CLINICAL TRIAL REGISTRATION (EC) I. SPONSOR INFORMATION Foreign National 2. LEGAL PERSON 2.1 FOREIGN SPONSOR Registered Name: GlaxoSmithKline Registered Tradename: GlaxoSmithKline FOREIGN SPONSOR REPRESENTATIVE IN PERU SUBSIDIARY: BRANCH: CRO: OTHER: _________ Tradename: N/A TYPE OF INSTITUTION Laboratorio (Industria Farmacéutica) II. CLINICAL TRIAL GENERAL INFORMATION 1. CLINICAL TRIAL IDENTIFICATION Scientific Title: A RANDOMIZED, DOUBLE-BLIND, DOUBLE DUMMY, COMPARATIVE, MULTICENTER STUDY TO ASSESS THE INCIDENCE OF HEMOLYSIS, SAFETY, AND EFFICACY OF TAFENOQUINE (SB-252263, WR238605) VERSUS PRIMAQUINE IN THE TREATMENT OF SUBJECTS WITH PLASMODIUM VIVAX MALARIA Public Title: A RANDOMIZED, DOUBLE-BLIND, DOUBLE DUMMY, COMPARATIVE, MULTICENTER STUDY TO ASSESS THE INCIDENCE OF HEMOLYSIS, SAFETY, AND EFFICACY OF TAFENOQUINE (SB-252263, WR238605) VERSUS PRIMAQUINE IN THE TREATMENT OF SUBJECTS WITH PLASMODIUM VIVAX MALARIA Secundary ID(s): WHO UTN: PER-048-14 Protocol Code: TAF116564 Clinicaltrials.gov: NA EUDRACT N°: NA 1 1-2 2 2-3 Study clinical phase: 3 4 Clinical Trial Total Duration: 24 months No Aplica 0(exploratory trials) Enrolment start date in Peru (Initial) 30/12/2014 Worldwide enrolment start date (dd/ (dd/mm/aaaa): 18/09/2014 mm/aaaa): Enrolment start date in Peru (Posterior) (dd/mm/aaaa): Without starting enrollment In enrollment Peru enrolment status : Enrollment stopped Enrollment closed Other 2. CLINICAL TRIAL GOALS AND DESIGN Randomnized Simple Non randomnized Double Assignation method Type of blinding No aplica Triple Open Single arm Parallel Crossed Factorial Assignation Others: ____________________ Study Design In this prospective, double-blind, double-dummy design, a total of 300 subjects will be randomized to treatment on Day 1, of which a minimum of 50 female subjects must be enrolled that display moderate G6PD deficiency (&#8805;40% - <70% of the site median G6PD value).
    [Show full text]
  • 2-Amino-1,3,4-Thiadiazoles in Leishmaniasis
    Review Future Prospects in the Treatment of Parasitic Diseases: 2‐Amino‐1,3,4‐Thiadiazoles in Leishmaniasis Georgeta Serban Pharmaceutical Chemistry Department, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga, 410028 Oradea, Romania; [email protected]; Tel: +4‐0756‐276‐377 Received: 22 March 2019; Accepted: 17 April 2019; Published: 19 April 2019 Abstract: Neglected tropical diseases affect the lives of a billion people worldwide. Among them, the parasitic infections caused by protozoan parasites of the Trypanosomatidae family have a huge impact on human health. Leishmaniasis, caused by Leishmania spp., is an endemic parasitic disease in over 88 countries and is closely associated with poverty. Although significant advances have been made in the treatment of leishmaniasis over the last decade, currently available chemotherapy is far from satisfactory. The lack of an approved vaccine, effective medication and significant drug resistance worldwide had led to considerable interest in discovering new, inexpensive, efficient and safe antileishmanial agents. 1,3,4‐Thiadiazole rings are found in biologically active natural products and medicinally important synthetic compounds. The thiadiazole ring exhibits several specific properties: it is a bioisostere of pyrimidine or benzene rings with prevalence in biologically active compounds; the sulfur atom increases lipophilicity and combined with the mesoionic character of thiadiazoles imparts good oral absorption and good cell permeability, resulting in good bioavailability. This review presents synthetic 2‐amino‐1,3,4‐thiadiazole derivatives with antileishmanial activity. Many reported derivatives can be considered as lead compounds for the synthesis of future agents as an alternative to the treatment of leishmaniasis. Keywords: 2‐amino‐1,3,4‐thiadiazole; neglected tropical diseases; protozoan parasites; Leishmania spp.; antileishmanial activity; inhibitory concentration 1.
    [Show full text]
  • Viewed in Detail
    NEGLECTED TROPICAL DISEASE CHEMOTHERAPY: MECHANISTIC CHARACTERIZATION OF ANTITRYPANOSOMAL DIHYDROQUINOLINES AND DEVELOPMENT OF A HIGH THROUGHPUT ANTILEISHMANIAL SCREENING ASSAY DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy from the Graduate School of The Ohio State University By Shanshan He, M.S. ****** Graduate Program in Pharmaceutical Sciences The Ohio State University 2012 Dissertation Committee: Karl A Werbovetz, Ph.D., Advisor Mark E Drew, Ph.D. Co-advisor Werner Tjarks, Ph.D. Juan D D Alfonzo, Ph.D Copyright by Shanshan He 2012 ABSTRACT Human African trypanosomiasis (HAT) and leishmaniasis are identified by the World Health Organization (WHO) as neglected tropical diseases (NTDs), together with Chagas disease and Buruli ulcer. These NTDs mostly affect people in remote or rural area, and there are very limited control and therapeutic options. The investment on research and development against NTDs is insufficient. Human African trypanosomiasis (HAT) is a vector-borne parasitic disease caused by Trypanosoma brucei subspecies. Transmitted by the tsetse fly, the disease mainly affects rural populations in sub-Saharan Africa and is fatal if untreated. New drugs are needed against HAT that are safe, affordable, easy to administer, active against first and second stage disease, and effective against both subspecies of T. brucei (11, 139). From medicinal chemistry investigation in Karl Werbovetz group, several N1-substituted 1,2-dihydroquinoline-6-ols were discovered displaying nanomolar IC50 values in vitro against T. b. rhodesiense and selectivity indexes (SI) up to >18,000 (39). OSU-40 (1- benzyl-1,2-dihydro-2,2,4–trimethylquinolin-6-yl acetate) is selectively potent against T.
    [Show full text]
  • Press Release Tafenoquine Approved in Peru
    PRESS RELEASE TAFENOQUINE APPROVED IN PERU Perú becomes second malaria-endemic country in Latin America to approve single- dose tafenoquine for radical cure of P. vivax malaria • Tafenoquine is the first drug approved for the radical cure (relapse prevention) of P. vivax malaria in more than 60 years. • As a single dose, tafenoquine offers a much shorter treatment regimen compared to current standard of care, with the benefit of increasing patient compliance and helping to advance malaria elimination efforts. Lima, January 2021. GSK and Medicines for Malaria Venture (MMV) announced that the General Directorate of Medicines, Supplies and Drugs (DIGEMID) has issued marketing authorization for single-dose tafenoquine for radical cure (prevention relapse) of Plasmodium vivax (P. vivax) malaria in patients 16 years of age or older receiving chloroquine for acute P. vivax infection (blood-stage). Peru becomes the second malaria endemic country in Latin America after Brazil to approve single dose tafenoquine for the prevention of relapse of P. vivax malaria. As a single-dose treatment, tafenoquine facilitates compliance and thus overcomes one of the major limitations of the only other drug approved for the prevention of relapse of P. vivax malaria, primaquine, which needs to be taken for 7 days. “The history of Peru and malaria are deeply entwined. In the nineteenth century, the famous writer Ricardo Palma, made malaria part of his narratives in the book Tradiciones Peruanas. In the same way, the cinchona tree, formerly known to be effective against malaria and from which quinine is derived, is part of the National Emblem”, said Dr José Sandoval, GSK Peru’s Medical Director.
    [Show full text]
  • ARAKODA (Tafenoquine)
    HIGHLIGHTS OF PRESCRIBING INFORMATION • G6PD Deficiency in Pregnancy or Lactation: ARAKODA may cause These highlights do not include all the information needed to use fetal harm when administered to a pregnant woman with a G6PD- ARAKODA™ safely and effectively. See full prescribing information for deficient fetus. ARAKODA is not recommended during pregnancy. A ARAKODA™. G6PD-deficient infant may be at risk for hemolytic anemia from exposure to ARAKODA through breast milk. Check infant’s G6PD ARAKODA™ (tafenoquine) tablets, for oral use status before breastfeeding begins. (5.2, 8.1, 8.2) Initial U.S. Approval: 2018 • Methemoglobinemia: Asymptomatic elevations in blood methemoglobin have been observed. Initiate appropriate therapy if signs or symptoms of ----------------------------INDICATIONS AND USAGE--------------------------- methemoglobinemia occur. (5. 3) ARAKODA is an antimalarial indicated for the prophylaxis of malaria in • Psychiatric Effects: Serious psychotic adverse reactions have been patients aged 18 years and older. (1) observed in patients with a history of psychosis or schizophrenia, at doses different from the approved dose. If psychotic symptoms ----------------------DOSAGE AND ADMINISTRATION----------------------- (hallucinations, delusions, or grossly disorganized thinking or behavior) • All patients must be tested for glucose-6-phosphate dehydrogenase (G6PD) occur, consider discontinuation of ARAKODA therapy and, evaluation deficiency prior to prescribing ARAKODA. (2.1) by a mental health professional as soon as possible.
    [Show full text]