113
Views
43
CrossRef citations to date
0
Altmetric
Review

Developments in diagnosis and treatment of visceral leishmaniasis during the last decade and future prospects

&
Pages 79-98 | Published online: 10 Jan 2014

References

  • Chappuis F, Sundar S, Hailu A et al. Visceral leishmaniasis: what are the needs for diagnosis, treatment and control? Nat. Rev. Microbiol. 5(11), 873–882 (2007).
  • Mondal S, Bhattacharya P, Ali N. Current diagnosis and treatment of visceral leishmaniasis. Expert Rev. Anti. Infect. Ther. 8(8), 919–944 (2010).
  • Das VN, Siddiqui NA, Verma RB et al. Asymptomatic infection of visceral leishmaniasis in hyperendemic areas of Vaishali district, Bihar, India: a challenge to kala-azar elimination programmes. Trans. R. Soc. Trop. Med. Hyg. 105(11), 661–666 (2011).
  • Wang JY, Cui G, Chen HT, Zhou XN, Gao CH, Yang YT. Current epidemiological profile and features of visceral leishmaniasis in people’s republic of China. Parasit. Vectors 5, 31 (2012).
  • Srividya G, Kulshrestha A, Singh R, Salotra P. Diagnosis of visceral leishmaniasis: developments over the last decade. Parasitol. Res. 110(3), 1065–1078 (2012).
  • Alvar J, Vélez ID, Bern C et al.; WHO Leishmaniasis Control Team. Leishmaniasis worldwide and global estimates of its incidence. PLoS ONE 7(5), e35671 (2012).
  • Sharma MC, Gupta AK, Das VN et al. Leishmania donovani in blood smears of asymptomatic persons. Acta Trop. 76(2), 195–196 (2000).
  • Topno RK, Das VN, Ranjan A et al. Asymptomatic infection with visceral leishmaniasis in a disease-endemic area in bihar, India. Am. J. Trop. Med. Hyg. 83(3), 502–506 (2010).
  • Singh S, Chaudhry VP, Wali JP. Transfusion-transmitted kala-azar in India. Transfusion 36(9), 848–849 (1996).
  • Singh S. New developments in diagnosis of leishmaniasis. Indian J. Med. Res. 123(3), 311–330 (2006).
  • Silva LA, Romero HD, Nogueira Nascentes GA, Costa RT, Rodrigues V, Prata A. Anti-Leishmania immunological tests for asymptomatic subjects living in a visceral leishmaniasis-endemic area in Brazil. Am. J. Trop. Med. Hyg. 84(2), 261–266 (2011).
  • Mondal D, Nasrin KN, Huda MM et al. Enhanced case detection and improved diagnosis of PKDL in a kala-azar-endemic area of Bangladesh. PLoS Negl. Trop. Dis. 4(10), 832 (2010).
  • Singh S, Sharma U, Mishra J. Post-kala-azar dermal leishmaniasis: recent developments. Int. J. Dermatol. 50(9), 1099–1108 (2011).
  • Salotra P, Singh R. Challenges in the diagnosis of post kala-azar dermal leishmaniasis. Indian J. Med. Res. 123(3), 295–310 (2006).
  • Katara GK, Ansari NA, Singh A, Ramesh V, Salotra P. Evidence for involvement of Th17 type responses in post kala azar dermal leishmaniasis (PKDL). PLoS Negl. Trop. Dis. 6(6), e1703 (2012).
  • Matlashewski G, Arana B, Kroeger A et al. Visceral leishmaniasis: elimination with existing interventions. Lancet Infect. Dis. 11(4), 322–325 (2011).
  • Islam S, Ashraful Alam Bhuiyan M, Bern C. Post-kala-azar dermal leishmaniasis in Mymensingh, Bangladesh. Am. J. Trop. Med. Hyg. 85(2), 193–194 (2011).
  • Uranw S, Ostyn B, Rijal A et al. Post-kala-azar dermal leishmaniasis in Nepal: a retrospective cohort study (2000-2010). PLoS Negl. Trop. Dis. 5(12), e1433 (2011).
  • Fisa R, Riera C, López-Chejade P et al. Leishmania infantum DNA detection in urine from patients with visceral leishmaniasis and after treatment control. Am. J. Trop. Med. Hyg. 78(5), 741–744 (2008).
  • Kumar D, Srividya G, Verma S et al. Presence of anti-Lepp12 antibody: a marker for diagnostic and prognostic evaluation of visceral leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 102(2), 167–171 (2008).
  • Mohapatra TM, Singh DP, Sen MR, Bharti K, Sundar S. Compararative evaluation of rK9, rK26 and rK39 antigens in the serodiagnosis of Indian visceral leishmaniasis. J. Infect. Dev. Ctries. 4(2), 114–117 (2010).
  • Sivakumar R, Sharma P, Chang KP, Singh S. Cloning, expression, and purification of a novel recombinant antigen from Leishmania donovani. Protein Expr. Purif. 46(1), 156–165 (2006).
  • Costa MM, Penido M, dos Santos MS et al. Improved canine and human visceral leishmaniasis immunodiagnosis using combinations of synthetic peptides in enzyme-linked immunosorbent assay. PLoS Negl. Trop. Dis. 6(5), e1622 (2012).
  • Boarino A, Scalone A, Gradoni L et al. Development of recombinant chimeric antigen expressing immunodominant B epitopes of Leishmania infantum for serodiagnosis of visceral leishmaniasis. Clin. Diagn. Lab. Immunol. 12(5), 647–653 (2005).
  • Antinori S, Schifanella L, Corbellino M. Leishmaniasis: new insights from an old and neglected disease. Eur. J. Clin. Microbiol. Infect. Dis. 31(2), 109–118 (2012).
  • Fernández-Guerrero ML, Aguado JM, Buzón L et al. Visceral leishmaniasis in immunocompromised hosts. Am. J. Med. 83(6), 1098–1102 (1987).
  • Srivastava P, Dayama A, Mehrotra S, Sundar S. Diagnosis of visceral leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 105(1), 1–6 (2011).
  • Guerin PJ, Olliaro P, Sundar S et al. Visceral leishmaniasis: current status of control, diagnosis, and treatment, and a proposed research and development agenda. Lancet Infect. Dis. 2(8), 494–501 (2002).
  • Chulay JD, Bryceson AD. Quantitation of amastigotes of Leishmania donovani in smears of splenic aspirates from patients with visceral leishmaniasis. Am. J. Trop. Med. Hyg. 32(3), 475–479 (1983).
  • Berman J. Visceral leishmaniasis in the New World & Africa. Indian J. Med. Res. 123(3), 289–294 (2006).
  • da Silva MR, Stewart JM, Costa CH. Sensitivity of bone marrow aspirates in the diagnosis of visceral leishmaniasis. Am. J. Trop. Med. Hyg. 72(6), 811–814 (2005).
  • Hide M, Singh R, Kumar B, Bañuls AL, Sundar S. A microculture technique for isolating live Leishmania parasites from peripheral blood of visceral leishmaniasis patients. Acta Trop. 102(3), 197–200 (2007).
  • Maurya R, Mehrotra S, Prajapati VK, Nylén S, Sacks D, Sundar S. Evaluation of blood agar microtiter plates for culturing leishmania parasites to titrate parasite burden in spleen and peripheral blood of patients with visceral leishmaniasis. J. Clin. Microbiol. 48(5), 1932–1934 (2010).
  • Allahverdiyev AM, Bagirova M, Uzun S et al. The value of a new microculture method for diagnosis of visceral leishmaniasis by using bone marrow and peripheral blood. Am. J. Trop. Med. Hyg. 73(2), 276–280 (2005).
  • Salam MA, Khan MG, Bhaskar KR, Afrad MH, Huda MM, Mondal D. Peripheral blood buffy coat smear: a promising tool for diagnosis of visceral leishmaniasis. J. Clin. Microbiol. 50(3), 837–840 (2012).
  • Zijlstra EE, Musa AM, Khalil EA, el-Hassan IM, el-Hassan AM. Post-kala-azar dermal leishmaniasis. Lancet Infect. Dis. 3(2), 87–98 (2003).
  • Pintado V, López-Vélez R. HIV-associated visceral leishmaniasis. Clin. Microbiol. Infect. 7(6), 291–300 (2001).
  • Galaï Y, Chabchoub N, Ben-Abid M et al. Diagnosis of mediterranean visceral leishmaniasis by detection of Leishmania antibodies and Leishmania DNA in oral fluid samples collected using an Oracol device. J. Clin. Microbiol. 49(9), 3150–3153 (2011).
  • Vaish M, Singh OP, Chakravarty J, Sundar S. rK39 antigen for the diagnosis of visceral leishmaniasis by using human saliva. Am. J. Trop. Med. Hyg. 86(4), 598–600 (2012).
  • Khan MG, Alam MS, Podder MP et al. Evaluation of rK39 strip test using urine for diagnosis of visceral leishmaniasis in an endemic area in Bangladesh. Parasit. Vectors 3, 114 (2010).
  • Singh D, Pandey K, Das VN et al. Novel noninvasive method for diagnosis of visceral leishmaniasis by rK39 testing of sputum samples. J. Clin. Microbiol. 47(8), 2684–2685 (2009).
  • Silva ES, Schoone GJ, Gontijo CM, Brazil RP, Pacheco RS, Schallig HD. Application of direct agglutination test (DAT) and fast agglutination screening test (FAST) for sero-diagnosis of visceral leishmaniasis in endemic area of Minas Gerais, Brazil. Kinetoplastid Biol. Dis. 4, 4 (2005).
  • Mandal J, Khurana S, Dubey ML, Bhatia P, Varma N, Malla N. Evaluation of direct agglutination test, rK39 test, and ELISA for the diagnosis of visceral leishmaniasis. Am. J. Trop. Med. Hyg. 79(1), 76–78 (2008).
  • Terán-Angel G, Schallig H, Zerpa O, Rodríguez V, Ulrich M, Cabrera M. The direct agglutination test as an alternative method for the diagnosis of canine and human visceral leishmaniasis. Biomedica. 27(3), 447–453 (2007).
  • Akhoundi B, Mohebali M, Edrissian GH et al. Preparation and evaluation of a glycerol-preserved direct agglutination antigen for long-term preservation: a comparative study of the detection of anti-Leishmania infantum antibodies in human and dog. Asian Pac. J. Trop. Med. 5(2), 117–120 (2012).
  • Oliveira E, Saliba SW, Andrade CF, Rabello A. Direct agglutination test (DAT): improvement of biosafety for laboratory diagnosis of visceral leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 105(7), 414–416 (2011).
  • Patil RR, Muliyil JP, Nandy A, Addy A, Maji AK, Chatterjee P. Dynamics of the antibodies in cohorts of cured cases of visceral leishmaniasis: its implication on the validity of serological test, value in prognosis and in post therapeutic assessment. Hum. Vaccin. Immunother. 8(6), 725–730 (2012).
  • Ostyn B, Gidwani K, Khanal B et al. Incidence of symptomatic and asymptomatic Leishmania donovani infections in high-endemic foci in India and Nepal: a prospective study. PLoS Negl. Trop. Dis. 5(10), e1284 (2011).
  • Khanal B, Rijal S, Ostyn B et al. Serological markers for Leishmania donovani infection in Nepal: Agreement between direct agglutination test and rK39 ELISA. Trop. Med. Int. Health 15(11), 1390–1394 (2010).
  • Schoone GJ, Hailu A, Kroon CC, Nieuwenhuys JL, Schallig HD, Oskam L. A fast agglutination screening test (FAST) for the detection of anti-Leishmania antibodies. Trans. R. Soc. Trop. Med. Hyg. 95(4), 400–401 (2001).
  • Akhoundi B, Mohebali M, Babakhan L et al. Rapid detection of human Leishmania infantum infection: a comparative field study using the fast agglutination screening test and the direct agglutination test. Travel Med. Infect. Dis. 8(5), 305–310 (2010).
  • Islam MZ, Itoh M, Mirza R et al. Direct agglutination test with urine samples for the diagnosis of visceral leishmaniasis. Am. J. Trop. Med. Hyg. 70(1), 78–82 (2004).
  • Singh R, Subba Raju BV, Jain RK, Salotra P. Potential of direct agglutination test based on promastigote and amastigote antigens for serodiagnosis of post-kala-azar dermal leishmaniasis. Clin. Diagn. Lab. Immunol. 12(10), 1191–1194 (2005).
  • Alam M, Shamsuzzaman A, Musa A et al. Antigen detection from urine of Kala-azar cases by latex agglutination test. Mymensingh Med. J. 17(1), 22–27 (2008).
  • ter Horst R, Tefera T, Assefa G, Ebrahim AZ, Davidson RN, Ritmeijer K. Field evaluation of rK39 test and direct agglutination test for diagnosis of visceral leishmaniasis in a population with high prevalence of human immunodeficiency virus in Ethiopia. Am. J. Trop. Med. Hyg. 80(6), 929–934 (2009).
  • de Almeida e Cavalcanti AT, Medeiros Z, Lopes F et al. Diagnosing visceral leishmaniasis and HIV/AIDS co-infection: a case series study in Pernambuco, Brazil. Rev. Inst. Med. Trop. Sao Paulo 54(1), 43–47 (2012).
  • Maia Z, Lírio M, Mistro S, Mendes CM, Mehta SR, Badaro R. Comparative study of rK39 Leishmania antigen for serodiagnosis of visceral leishmaniasis: systematic review with meta-analysis. PLoS Negl. Trop. Dis. 6(1), e1484 (2012).
  • Salotra P, Sreenivas G, Nasim AA, Subba Raju BV, Ramesh V. Evaluation of enzyme-linked immunosorbent assay for diagnosis of post-kala-azar dermal leishmaniasis with crude or recombinant k39 antigen. Clin. Diagn. Lab. Immunol. 9(2), 370–373 (2002).
  • Biyani N, Singh AK, Mandal S, Chawla B, Madhubala R. Differential expression of proteins in antimony-susceptible and -resistant isolates of Leishmania donovani. Mol. Biochem. Parasitol. 179(2), 91–99 (2011).
  • Vaish M, Bhatia A, Reed SG, Chakravarty J, Sundar S. Evaluation of rK28 antigen for serodiagnosis of visceral leishmaniasis in India. Clin. Microbiol. Infect. 18(1), 81–85 (2012).
  • Abass E, Mahamoud A, Mansour D, Mohebali M, El Harith A. Validation of a b-ME ELISA for detection of anti-Leishmania donovani antibodies in Eastern Sudan. Iran. J. Immunol. 8(3), 150–158 (2011).
  • Kumar S, Kumar D, Chakravarty J, Rai M, Sundar S. Identification and characterization of a novel Leishmania donovani antigen for serodiagnosis of visceral leishmaniasis. Am. J. Trop. Med. Hyg. 86(4), 601–605 (2012).
  • Kumar S, Kumar D, Chakravarty J, Sundar S. Identification and characterization of a novel, 37-kilodalton Leishmania donovani antigen for diagnosis of Indian visceral leishmaniasis. Clin. Vaccine Immunol. 18(5), 772–775 (2011).
  • Kumar D, Kumar S, Chakravarty J, Sundar S. A novel 12.6-kDa protein of Leishmania donovani for the diagnosis of Indian visceral leishmaniasis. Vector Borne Zoonotic Dis. 11(10), 1359–1364 (2011).
  • Gidwani K, Picado A, Ostyn B et al. Persistence of Leishmania donovani antibodies in past visceral leishmaniasis cases in India. Clin. Vaccine Immunol. 18(2), 346–348 (2011).
  • Abeijon C, Kashino SS, Silva FO et al. Identification and diagnostic utility of Leishmania infantum proteins found in urine samples from patients with visceral leishmaniasis. Clin. Vaccine Immunol. 19(6), 935–943 (2012).
  • Souza GF, Biscione F, Greco DB, Rabello A. Slow clinical improvement after treatment initiation in Leishmania/HIV co-infected patients. Rev. Soc. Bras. Med. Trop. 45(2), 147–150 (2012).
  • Sinha PK, Pandey K, Bhattacharya SK. Diagnosis & management of Leishmania/HIV co-infection. Indian J. Med. Res. 121(4), 407–414 (2005).
  • Mansour D, Abass EM, Mahamoud A, el Harith A. Qualitative and semi-quantitative comparison of an rK39 strip test and direct agglutination test for detection of anti-Leishmania donovani antibodies in the Sudan. Iran. J. Immunol. 6(4), 208–215 (2009).
  • Cañavate C, Herrero M, Nieto J et al. Evaluation of two rK39 dipstick tests, direct agglutination test, and indirect fluorescent antibody test for diagnosis of visceral leishmaniasis in a new epidemic site in highland Ethiopia. Am. J. Trop. Med. Hyg. 84(1), 102–106 (2011).
  • de Assis TS, Braga AS, Pedras MJ et al. Multi-centric prospective evaluation of rK39 rapid test and direct agglutination test for the diagnosis of visceral leishmaniasis in Brazil. Trans. R. Soc. Trop. Med. Hyg. 105(2), 81–85 (2011).
  • Machado de Assis TS, Rabello A, Werneck GL. Predictive models for the diagnostic of human visceral leishmaniasis in Brazil. PLoS Negl Trop Dis. 6(2), e1542 (2012).
  • Terán-Ángel G, Rodríguez V, Silva R et al. Non-invasive diagnostic tools for visceral leishmaniasis: a comparison of the immunoserological tests DAT, rK26 and rK39. Biomedica. 30(1), 39–45 (2010).
  • Das ML, Deb M, Karki BM et al. Use of rK39 for diagnosis of post-kala-azar dermal leishmaniasis in Nepal. Southeast Asian J. Trop. Med. Public Health 38(4), 619–625 (2007).
  • Das NK, Singh SK, Ghosh S et al. Case series of misdiagnosis with rK39-strip test in Indian leishmaniasis. Am. J. Trop. Med. Hyg. 84(5), 688–691 (2011).
  • Saha S, Goswami R, Pramanik N et al. Easy test for visceral leishmaniasis and post-kala-azar dermal leishmaniasis. Emerging Infect. Dis. 17(7), 1304–1306 (2011).
  • Pattabhi S, Whittle J, Mohamath R et al. Design, development and evaluation of rK28-based point-of-care tests for improving rapid diagnosis of visceral leishmaniasis. PLoS Negl. Trop. Dis. 4(9), (2010).
  • Chakravarty J, Kumar S, Kumar R, Gautam S, Rai M, Sundar S. Evaluation of rK39 immunochromatographic test with urine for diagnosis of visceral leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 105(9), 537–539 (2011).
  • Boelaert M, El-Safi S, Hailu A et al. Diagnostic tests for kala-azar: a multi-centre study of the freeze-dried DAT, rK39 strip test and KAtex in East Africa and the Indian subcontinent. Trans. R. Soc. Trop. Med. Hyg. 102(1), 32–40 (2008).
  • Ahsan MM, Islam MN, Mollah AH et al. Evaluation of latex agglutination test (KAtex) for early diagnosis of kala-azar. Mymensingh Med. J. 19(3), 335–339 (2010).
  • Salam MA, Khan MG, Mondal D. Urine antigen detection by latex agglutination test for diagnosis and assessment of initial cure of visceral leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 105(5), 269–272 (2011).
  • Pandey K, Pandey BD, Mallik AK et al. Diagnosis of visceral leishmaniasis by polymerase chain reaction of DNA extracted from Giemsa’s solution-stained slides. Parasitol. Res. 107(3), 727–730 (2010).
  • de Assis TS, Caligiorne RB, Romero GA, Rabello A. Detection of Leishmania kDNA in human serum samples for the diagnosis of visceral leishmaniasis. Trans. R. Soc. Trop. Med. Hyg. 103(12), 1269–1272 (2009).
  • Fraga TL, Brustoloni YM, Lima RB et al. Polymerase chain reaction of peripheral blood as a tool for the diagnosis of visceral leishmaniasis in children. Mem. Inst. Oswaldo Cruz 105(3), 310–313 (2010).
  • Bourgeois N, Bastien P, Reynes J, Makinson A, Rouanet I, Lachaud L. ‘Active chronic visceral leishmaniasis’ in HIV-1-infected patients demonstrated by biological and clinical long-term follow-up of 10 patients. HIV Med. 11(10), 670–673 (2010).
  • Motazedian M, Fakhar M, Motazedian MH, Hatam G, Mikaeili F. A urine-based polymerase chain reaction method for the diagnosis of visceral leishmaniasis in immunocompetent patients. Diagn. Microbiol. Infect. Dis. 60(2), 151–154 (2008).
  • Srivastava P, Mehrotra S, Tiwary P, Chakravarty J, Sundar S. Diagnosis of Indian visceral leishmaniasis by nucleic acid detection using PCR. PLoS ONE 6(4), e19304 (2011).
  • Vaish M, Mehrotra S, Chakravarty J, Sundar S. Noninvasive molecular diagnosis of human visceral leishmaniasis. J. Clin. Microbiol. 49(5), 2003–2005 (2011).
  • Roelfsema JH, Nozari N, Herremans T, Kortbeek LM, Pinelli E. Evaluation and improvement of two PCR targets in molecular typing of clinical samples of Leishmania patients. Exp. Parasitol. 127(1), 36–41 (2011).
  • Salam MA, Mondal D, Kabir M, Ekram AR, Haque R. PCR for diagnosis and assessment of cure in kala-azar patients in Bangladesh. Acta Trop. 113(1), 52–55 (2010).
  • Rassi Y, Sanei Dehkordi A, Oshaghi MA et al. First report on natural infection of the Phlebotomus tobbi by Leishmania infantum in northwestern Iran. Exp. Parasitol. 131(3), 344–349 (2012).
  • Chargui N, Haouas N, Jaouadi K et al. Usefulness of a PCR-based method in the detection and species identification of Leishmania from clinical samples. Pathol. Biol. (Paris) 60(6), e75–e79 (2012).
  • Oshaghi MA, Ravasan NM, Hide M et al. Development of species-specific PCR and PCR-restriction fragment length polymorphism assays for L.infantum/L.donovani discrimination. Exp. Parasitol. 122(1), 61–65 (2009).
  • Deborggraeve S, Laurent T, Espinosa D et al. A simplified and standardized polymerase chain reaction format for the diagnosis of leishmaniasis. J. Infect. Dis. 198(10), 1565–1572 (2008).
  • Mugasa CM, Laurent T, Schoone GJ et al. Simplified molecular detection of Leishmania parasites in various clinical samples from patients with leishmaniasis. Parasit. Vectors 3(1), 13 (2010).
  • Saad AA, Ahmed NG, Osman OS et al. Diagnostic accuracy of the Leishmania OligoC-TesT and NASBA-Oligochromatography for diagnosis of leishmaniasis in Sudan. PLoS Negl. Trop. Dis. 4(8), e776 (2010).
  • Basiye FL, Mbuchi M, Magiri C et al. Sensitivity and specificity of the Leishmania OligoC-TesT and NASBA-oligochromatography for diagnosis of visceral leishmaniasis in Kenya. Trop. Med. Int. Health 15(7), 806–810 (2010).
  • Alam MZ, Shamsuzzaman AK, Kuhls K, Schönian G. PCR diagnosis of visceral leishmaniasis in an endemic region, Mymensingh district, Bangladesh. Trop. Med. Int. Health 14(5), 499–503 (2009).
  • Singh N, Kumar M, Singh RK. Leishmaniasis: current status of available drugs and new potential drug targets. Asian Pac. J. Trop. Med. 5(6), 485–497 (2012).
  • Silva-Lopez RE, Morgado-Díaz JA, Chávez MA, Giovanni-De-Simone S. Effects of serine protease inhibitors on viability and morphology of Leishmania (Leishmania) amazonensis promastigotes. Parasitol. Res. 101(6), 1627–1635 (2007).
  • Xingi E, Smirlis D, Myrianthopoulos V et al. 6-Br-5methylindirubin-3’oxime (5-Me-6-BIO) targeting the leishmanial glycogen synthase kinase-3 (GSK-3) short form affects cell-cycle progression and induces apoptosis-like death: exploitation of GSK-3 for treating leishmaniasis. Int. J. Parasitol. 39(12), 1289–1303 (2009).
  • Lee N, Gannavaram S, Selvapandiyan A, Debrabant A. Characterization of metacaspases with trypsin-like activity and their putative role in programmed cell death in the protozoan parasite Leishmania. Eukaryotic Cell 6(10), 1745–1757 (2007).
  • Singh G, Jayanarayan KG, Dey CS. Novobiocin induces apoptosis-like cell death in topoisomerase II over-expressing arsenite resistant Leishmania donovani. Mol. Biochem. Parasitol. 141(1), 57–69 (2005).
  • Tiuman TS, Santos AO, Ueda-Nakamura T, Filho BP, Nakamura CV. Recent advances in leishmaniasis treatment. Int. J. Infect. Dis. 15(8), e525–e532 (2011).
  • Veeken H, Ritmeijer K, Seaman J, Davidson R. A randomized comparison of branded sodium stibogluconate and generic sodium stibogluconate for the treatment of visceral leishmaniasis under field conditions in Sudan. Trop. Med. Int. Health 5(5), 312–317 (2000).
  • Aït-Oudhia K, Gazanion E, Vergnes B, Oury B, Sereno D. Leishmania antimony resistance: what we know what we can learn from the field. Parasitol. Res. 109(5), 1225–1232 (2011).
  • Decuypere S, Vanaerschot M, Brunker K et al. Molecular mechanisms of drug resistance in natural Leishmania populations vary with genetic background. PLoS Negl. Trop. Dis. 6(2), e1514 (2012).
  • Mukhopadhyay R, Mukherjee S, Mukherjee B et al. Characterisation of antimony-resistant Leishmania donovani isolates: biochemical and biophysical studies and interaction with host cells. Int. J. Parasitol. 41(13–14), 1311–1321 (2011).
  • Downing T, Imamura H, Decuypere S et al. Whole genome sequencing of multiple Leishmania donovani clinical isolates provides insights into population structure and mechanisms of drug resistance. Genome Res. 21(12), 2143–2156 (2011).
  • Vanaerschot M, Decuypere S, Berg M, Roy S, Dujardin JC. Drug-resistant microorganisms with a higher fitness – can medicines boost pathogens? Crit. Rev. Microbiol. doi:10.3109/1040841X.2012.716818 (2012) (Epub ahead of print).
  • Kumar D, Singh R, Bhandari V, Kulshrestha A, Negi NS, Salotra P. Biomarkers of antimony resistance: need for expression analysis of multiple genes to distinguish resistance phenotype in clinical isolates of Leishmania donovani. Parasitol. Res. 111(1), 223–230 (2012).
  • Roychoudhury J, Sinha R, Ali N. Therapy with sodium stibogluconate in stearylamine-bearing liposomes confers cure against SSG-resistant Leishmania donovani in BALB/c mice. PLoS ONE 6(3), e17376 (2011).
  • Musa A, Khalil E, Hailu A et al. Sodium stibogluconate (SSG) & paromomycin combination compared to SSG for visceral leishmaniasis in East Africa: a randomised controlled trial. PLoS Negl. Trop. Dis. 6(6), e1674 (2012).
  • Haldar AK, Sen P, Roy S. Use of antimony in the treatment of leishmaniasis: current status and future directions. Mol. Biol. Int. 2011, 571242 (2011).
  • Hailu W, Weldegebreal T, Hurissa Z et al. Safety and effectiveness of meglumine antimoniate in the treatment of Ethiopian visceral leishmaniasis patients with and without HIV co-infection. Trans. R. Soc. Trop. Med. Hyg. 104(11), 706–712 (2010).
  • Ritmeijer K, Dejenie A, Assefa Y et al. A comparison of miltefosine and sodium stibogluconate for treatment of visceral leishmaniasis in an Ethiopian population with high prevalence of HIV infection. Clin. Infect. Dis. 43(3), 357–364 (2006).
  • Sundar S, Chakravarty J. Antimony toxicity. Int. J. Environ. Res. Public Health 7(12), 4267–4277 (2010).
  • Chattopadhyay A, Jafurulla M. A novel mechanism for an old drug: amphotericin B in the treatment of visceral leishmaniasis. Biochem. Biophys. Res. Commun. 416(1–2), 7–12 (2011).
  • Paila YD, Saha B, Chattopadhyay A. amphotericin B inhibits entry of Leishmania donovani into primary macrophages. Biochem. Biophys. Res. Commun. 399(3), 429–433 (2010).
  • Kumar N, Sinha PK, Pandey K et al. A rare case of visceral leishmaniasis with multiple relapse and multi-drug unresponsive: successfully treated with combination therapy. Int. J. Clin. Pharm. 33(5), 726–729 (2011).
  • Meheus F, Balasegaram M, Olliaro P et al. Cost–effectiveness analysis of combination therapies for visceral leishmaniasis in the Indian subcontinent. PLoS Negl. Trop. Dis. 4(9), e818 (2010).
  • Singh S, Sivakumar R. Challenges and new discoveries in the treatment of leishmaniasis. J. Infect. Chemother. 10(6), 307–315 (2004).
  • Thakur CP, Kumar A, Mitra G et al. Impact of amphotericin B in the treatment of kala-azar on the incidence of PKDL in Bihar, India. Indian J. Med. Res. 128(1), 38–44 (2008).
  • Saha S, Mondal S, Ravindran R et al. IL-10- and TGF-b-mediated susceptibility in kala-azar and post-kala-azar dermal leishmaniasis: the significance of amphotericin B in the control of Leishmania donovani infection in India. J. Immunol. 179(8), 5592–5603 (2007).
  • Olliaro PL, Guerin PJ, Gerstl S, Haaskjold AA, Rottingen JA, Sundar S. Treatment options for visceral leishmaniasis: a systematic review of clinical studies done in India, 1980–2004. Lancet Infect. Dis. 5(12), 763–774 (2005).
  • Thakur CP, Kumar A, Mitra DK, Roy A, Sinha AK, Ranjan A. Improving outcome of treatment of kala-azar by supplementation of amphotericin B with physiologic saline and potassium chloride. Am. J. Trop. Med. Hyg. 83(5), 1040–1043 (2010).
  • Sundar S, Sinha PK, Rai M et al. Comparison of short-course multidrug treatment with standard therapy for visceral leishmaniasis in India: an open-label, non-inferiority, randomised controlled trial. Lancet 377(9764), 477–486 (2011).
  • Thakur CP, Pandey AK, Sinha GP, Roy S, Behbehani K, Olliaro P. Comparison of three treatment regimens with liposomal amphotericin B (AmBisome) for visceral leishmaniasis in India: a randomized dose-finding study. Trans. R. Soc. Trop. Med. Hyg. 90(3), 319–322 (1996).
  • Sundar S, Mehta H, Suresh AV, Singh SP, Rai M, Murray HW. Amphotericin B treatment for Indian visceral leishmaniasis: conventional versus lipid formulations. Clin. Infect. Dis. 38(3), 377–383 (2004).
  • Sinha PK, Roddy P, Palma PP et al. Effectiveness and safety of liposomal amphotericin B for visceral leishmaniasis under routine program conditions in Bihar, India. Am. J. Trop. Med. Hyg. 83(2), 357–364 (2010).
  • Chappuis F, Alirol E, Worku DT, Mueller Y, Ritmeijer K. High mortality among older patients treated with pentavalent antimonials for visceral leishmaniasis in East Africa and rationale for switch to liposomal amphotericin B. Antimicrob. Agents Chemother. 55(1), 455–456 (2011).
  • Sundar S, Chakravarty J, Agarwal D, Rai M, Murray HW. Single-dose liposomal amphotericin B for visceral leishmaniasis in India. N. Engl. J. Med. 362(6), 504–512 (2010).
  • Edwards T, Omollo R, Khalil EA et al. Single-dose liposomal amphotericin B (AmBisome®) for the treatment of visceral leishmaniasis in East Africa: study protocol for a randomized controlled trial. Trials 12, 66 (2011).
  • Mondal S, Bhattacharya P, Rahaman M, Ali N, Goswami RP. A curative immune profile one week after treatment of Indian kala-azar patients predicts success with a short-course liposomal amphotericin B therapy. PLoS Negl. Trop. Dis. 4(7), e764 (2010).
  • Sinha PK, van Griensven J, Pandey K et al. Liposomal amphotericin B for visceral leishmaniasis in human immunodeficiency virus-co-infected patients: 2-year treatment outcomes in Bihar, India. Clin. Infect. Dis. 53(7), e91–e98 (2011).
  • Shakya N, Bajpai P, Gupta S. Therapeutic switching in Leishmania chemotherapy: a distinct approach towards unsatisfied treatment needs. J. Parasit. Dis. 35(2), 104–112 (2011).
  • Sundar S, Chakravarty J, Agarwal D, Shah A, Agrawal N, Rai M. Safety of a pre-formulated amphotericin B lipid emulsion for the treatment of Indian Kala-azar. Trop. Med. Int. Health 13(9), 1208–1212 (2008).
  • Croft SL, Neal RA, Pendergast W, Chan JH. The activity of alkyl phosphorylcholines and related derivatives against Leishmania donovani. Biochem. Pharmacol. 36(16), 2633–2636 (1987).
  • Paris C, Loiseau PM, Bories C, Bréard J. Miltefosine induces apoptosis-like death in Leishmania donovani promastigotes. Antimicrob. Agents Chemother. 48(3), 852–859 (2004).
  • Ponte CB, Alves EA, Sampaio RN et al. Miltefosine enhances phagocytosis but decreases nitric oxide production by peritoneal macrophages of C57BL/6 mice. Int. Immunopharmacol. 13(1), 114–119 (2012).
  • Gupta R, Kushawaha PK, Samant M, Jaiswal AK, Baharia RK, Dube A. Treatment of Leishmania donovani-infected hamsters with miltefosine: analysis of cytokine mRNA expression by real-time PCR, lymphoproliferation, nitrite production and antibody responses. J. Antimicrob. Chemother. 67(2), 440–443 (2012).
  • Mukhopadhyay D, Das NK, Roy S, Kundu S, Barbhuiya JN, Chatterjee M. Miltefosine effectively modulates the cytokine milieu in Indian post kala-azar dermal leishmaniasis. J. Infect. Dis. 204(9), 1427–1436 (2011).
  • Dorlo TP, Huitema AD, Beijnen JH, de Vries PJ. Optimal dosing of miltefosine in children and adults with visceral leishmaniasis. Antimicrob. Agents Chemother. 56(7), 3864–3872 (2012).
  • Rahman M, Ahmed BN, Faiz MA et al. Phase IV trial of miltefosine in adults and children for treatment of visceral leishmaniasis (kala-azar) in Bangladesh. Am. J. Trop. Med. Hyg. 85(1), 66–69 (2011).
  • Omollo R, Alexander N, Edwards T et al. Safety and efficacy of miltefosine alone and in combination with sodium stibogluconate and liposomal amphotericin B for the treatment of primary visceral leishmaniasis in East Africa: study protocol for a randomized controlled trial. Trials 12, 166 (2011).
  • Ramesh V, Katara GK, Verma S, Salotra P. Miltefosine as an effective choice in the treatment of post-kala-azar dermal leishmaniasis. Br. J. Dermatol. 165(2), 411–414 (2011).
  • Sundar S, Singh A, Rai M et al. Efficacy of miltefosine in the treatment of visceral leishmaniasis in India after a decade of use. Clin. Infect. Dis. 55(4), 543–550 (2012).
  • Bhandari V, Kulshrestha A, Deep DK et al. Drug susceptibility in Leishmania isolates following miltefosine treatment in cases of visceral leishmaniasis and post kala-azar dermal leishmaniasis. PLoS Negl. Trop. Dis. 6(5), e1657 (2012).
  • Maltezou HC. Visceral leishmaniasis: advances in treatment. Recent Pat. Antiinfect. Drug Discov. 3(3), 192–198 (2008).
  • Bhattacharya SK, Jha TK, Sundar S et al. Efficacy and tolerability of miltefosine for childhood visceral leishmaniasis in India. Clin. Infect. Dis. 38(2), 217–221 (2004).
  • Sundar S, Chakravarty J. Recent advances in the diagnosis and treatment of kala-azar. Natl. Med. J. India 25(2), 85–89 (2012).
  • Luque-Ortega JR, de la Torre BG, Hornillos V et al. Defeating Leishmania resistance to miltefosine (hexadecylphosphocholine) by peptide-mediated drug smuggling: a proof of mechanism for trypanosomatid chemotherapy. J. Control. Release 161(3), 835–842 (2012).
  • Shakya N, Sane SA, Gupta S. Antileishmanial efficacy of fluconazole and miltefosine in combination with an immunomodulator–picroliv. Parasitol. Res. 108(4), 793–800 (2011).
  • Shakya N, Sane SA, Vishwakarma P, Bajpai P, Gupta S. Improved treatment of visceral leishmaniasis (kala-azar) by using combination of ketoconazole, miltefosine with an immunomodulator-Picroliv. Acta Trop. 119(2–3), 188–193 (2011).
  • Shakya N, Sane SA, Vishwakarma P, Gupta S. Enhancement in therapeutic efficacy of miltefosine in combination with synthetic bacterial lipopeptide, Pam3Cys against experimental visceral leishmaniasis. Exp. Parasitol. 131(3), 377–382 (2012).
  • Gupta S, Sane SA, Shakya N, Vishwakarma P, Haq W. CpG oligodeoxynucleotide 2006 and miltefosine, a potential combination for treatment of experimental visceral leishmaniasis. Antimicrob. Agents Chemother. 55(7), 3461–3464 (2011).
  • Dorlo TP, Balasegaram M, Lima MA, de Vries PJ, Beijnen JH, Huitema AD. Translational pharmacokinetic modelling and simulation for the assessment of duration of contraceptive use after treatment with miltefosine. J. Antimicrob. Chemother. 67(8), 1996–2004 (2012).
  • Coimbra ES, Libong D, Cojean S et al. Mechanism of interaction of sitamaquine with Leishmania donovani. J. Antimicrob. Chemother. 65(12), 2548–2555 (2010).
  • Dietze R, Carvalho SF, Valli LC et al. Phase 2 trial of WR6026, an orally administered 8-aminoquinoline, in the treatment of visceral leishmaniasis caused by Leishmania chagasi. Am. J. Trop. Med. Hyg. 65(6), 685–689 (2001).
  • Sherwood JA, Gachihi GS, Muigai RK et al. Phase 2 efficacy trial of an oral 8-aminoquinoline (WR6026) for treatment of visceral leishmaniasis. Clin. Infect. Dis. 19(6), 1034–1039 (1994).
  • Wasunna MK, Rashid JR, Mbui J et al. A Phase II dose-increasing study of sitamaquine for the treatment of visceral leishmaniasis in Kenya. Am. J. Trop. Med. Hyg. 73(5), 871–876 (2005).
  • Jha TK, Sundar S, Thakur CP, Felton JM, Sabin AJ, Horton J. A Phase II dose-ranging study of sitamaquine for the treatment of visceral leishmaniasis in India. Am. J. Trop. Med. Hyg. 73(6), 1005–1011 (2005).
  • Sundar S, Sinha PK, Dixon SA et al. Pharmacokinetics of oral sitamaquine taken with or without food and safety and efficacy for treatment of visceral leishmaniais: a randomized study in Bihar, India. Am. J. Trop. Med. Hyg. 84(6), 892–900 (2011).
  • Bories C, Cojean S, Huteau F, Loiseau PM. Selection and phenotype characterisation of sitamaquine-resistant promastigotes of Leishmania donovani. Biomed. Pharmacother. 62(3), 164–167 (2008).
  • Kulshrestha A, Singh R, Kumar D, Negi NS, Salotra P. Antimony-resistant clinical isolates of Leishmania donovani are susceptible to paromomycin and sitamaquine. Antimicrob. Agents Chemother. 55(6), 2916–2921 (2011).
  • Pérez-Victoria JM, Bavchvarov BI, Torrecillas IR et al. Sitamaquine overcomes ABC-mediated resistance to miltefosine and antimony in Leishmania. Antimicrob. Agents Chemother. 55(8), 3838–3844 (2011).
  • Loiseau PM, Cojean S, Schrével J. Sitamaquine as a putative antileishmanial drug candidate: from the mechanism of action to the risk of drug resistance. Parasite 18(2), 115–119 (2011).
  • Alvar J, Croft S, Olliaro P. Chemotherapy in the treatment and control of leishmaniasis. Adv. Parasitol. 61, 223–274 (2006).
  • el-On J, Halevy S, Grunwald MH, Weinrauch L. Topical treatment of Old World cutaneous leishmaniasis caused by Leishmania major: a double-blind control study. J. Am. Acad. Dermatol. 27(2 Pt 1), 227–231 (1992).
  • Melaku Y, Collin SM, Keus K, Gatluak F, Ritmeijer K, Davidson RN. Treatment of kala-azar in southern Sudan using a 17-day regimen of sodium stibogluconate combined with paromomycin: a retrospective comparison with 30-day sodium stibogluconate monotherapy. Am. J. Trop. Med. Hyg. 77(1), 89–94 (2007).
  • Musa AM, Younis B, Fadlalla A et al. Paromomycin for the treatment of visceral leishmaniasis in Sudan: a randomized, open-label, dose-finding study. PLoS Negl. Trop. Dis. 4(10), e855 (2010).
  • Hailu A, Musa A, Wasunna M et al.; Leishmaniasis East Africa Platform (LEAP) group. Geographical variation in the response of visceral leishmaniasis to paromomycin in East Africa: a multicentre, open-label, randomized trial. PLoS Negl. Trop. Dis. 4(10), e709 (2010).
  • Sundar S, Jha TK, Thakur CP, Sinha PK, Bhattacharya SK. Injectable paromomycin for Visceral leishmaniasis in India. N. Engl. J. Med. 356(25), 2571–2581 (2007).
  • Sinha PK, Jha TK, Thakur CP et al. Phase 4 pharmacovigilance trial of paromomycin injection for the treatment of visceral leishmaniasis in India. J. Trop. Med. 2011, 645203 (2011).
  • Banerjee A, De M, Ali N. Combination therapy with paromomycin-associated stearylamine-bearing liposomes cures experimental visceral leishmaniasis through Th1-biased immunomodulation. Antimicrob. Agents Chemother. 55(4), 1661–1670 (2011).
  • Pandey K, Das VN, Singh D et al. Post-kala-azar dermal leishmaniasis in a patient treated with injectable paromomycin for visceral leishmaniasis in India. J. Clin. Microbiol. 50(4), 1478–1479 (2012).
  • Hendrickx S, Inocêncio da Luz RA, Bhandari V et al. Experimental induction of paromomycin resistance in antimony-resistant strains of L. donovani: outcome dependent on in vitro selection protocol. PLoS Negl. Trop. Dis. 6(5), e1664 (2012).
  • Jhingran A, Chawla B, Saxena S, Barrett MP, Madhubala R. Paromomycin: uptake and resistance in Leishmania donovani. Mol. Biochem. Parasitol. 164(2), 111–117 (2009).
  • Chawla B, Jhingran A, Panigrahi A, Stuart KD, Madhubala R. Paromomycin affects translation and vesicle-mediated trafficking as revealed by proteomics of paromomycin -susceptible -resistant Leishmania donovani. PLoS ONE 6(10), e26660 (2011).
  • Das VN, Ranjan A, Sinha AN et al. A randomized clinical trial of low dosage combination of pentamidine and allopurinol in the treatment of antimony unresponsive cases of visceral leishmaniasis. J. Assoc. Physicians India 49, 609–613 (2001).
  • Bray PG, Barrett MP, Ward SA, de Koning HP. Pentamidine uptake and resistance in pathogenic protozoa: past, present and future. Trends Parasitol. 19(5), 232–239 (2003).
  • Basselin M, Coombs GH, Barrett MP. Putrescine and spermidine transport in Leishmania. Mol. Biochem. Parasitol. 109(1), 37–46 (2000).
  • Basselin M, Lawrence F, Robert-Gero M. Pentamidine uptake in Leishmania donovani and Leishmania amazonensis promastigotes and axenic amastigotes. Biochem. J. 315 (Pt 2), 631–634 (1996).
  • Mukherjee A, Padmanabhan PK, Sahani MH, Barrett MP, Madhubala R. Roles for mitochondria in pentamidine susceptibility and resistance in Leishmania donovani. Mol. Biochem. Parasitol. 145(1), 1–10 (2006).
  • Buffet PA, Rosenthal É, Gangneux JP et al.; Société de Pathologie Exotique. Therapy of leishmaniasis in France: consensus on proposed guidelines. Presse Med. 40(2), 173–184 (2011).
  • Jha TK. Drug unresponsiveness & combination therapy for kala-azar. Indian J. Med. Res. 123(3), 389–398 (2006).
  • Sundar S, Rai M. Treatment of visceral leishmaniasis. Expert Opin. Pharmacother. 6(16), 2821–2829 (2005).
  • Das VN, Siddiqui NA, Pandey K et al. A controlled, randomized nonblinded clinical trial to assess the efficacy of amphotericin B deoxycholate as compared to pentamidine for the treatment of antimony unresponsive visceral leishmaniasis cases in Bihar, India. Ther. Clin. Risk Manag. 5(1), 117–124 (2009).
  • Patel TA, Lockwood DN. Pentamidine as secondary prophylaxis for visceral leishmaniasis in the immunocompromised host: report of four cases. Trop. Med. Int. Health 14(9), 1064–1070 (2009).
  • Rybniker J, Goede V, Mertens J et al. Treatment of visceral leishmaniasis with intravenous pentamidine and oral fluconazole in an HIV-positive patient with chronic renal failure – a case report and brief review of the literature. Int. J. Infect. Dis. 14(6), e522–e525 (2010).
  • Sundar S, Sinha PK, Verma DK et al. Ambisome plus miltefosine for Indian patients with kala-azar. Trans. R. Soc. Trop. Med. Hyg. 105(2), 115–117 (2011).
  • Saha P, Mukhopadhyay D, Chatterjee M. Immunomodulation by chemotherapeutic agents against Leishmaniasis. Int. Immunopharmacol. 11(11), 1668–1679 (2011).
  • Seifert K. Structures, targets and recent approaches in anti-leishmanial drug discovery and development. Open Med. Chem. J. 5, 31–39 (2011).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.