Visceral Leishmaniasis in Ethiopia: A Systematic Meta-Analysis of Prevalence, Diagnosis, Challenges and Opporunities, Treatment Options, and Temporal Patter

Main Article Content

Solomon Taddese

Abstract

Visceral leishmaniasis (VL) is a fatal neglected tropical disease caused by protozoan parasites of the genus Leishmania, belonging to the family Trypanosomatidae and is transmitted by the bite of infected female phlebotomine sandflies, which feed on blood to produce and mature eggs.


This study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines (Moher et al., 2009). The protocol of this study was registered in the International Prospective Register of Systematic Reviews (PROSPERO) with the registration number CRD42021234567. The data analysis was performed using Stata version 16 software (StataCorp LLC, College Station, TX, USA).  Comprehensive literature search was conducted to identify all published studies reporting the prevalence, distribution, determinants and trends of VL in Ethiopia from 2000 to 2023. The following electronic databases were searched: PubMed, Google Scholar and ScienceDirect.


This systematic meta-analysis provides a comprehensive overview of the current situation of VL in Ethiopia based on the available literature from 2000 to 2023. The study highlights the magnitude, distribution, determinants and trends of VL in Ethiopia as well as the challenges and opportunities for its control and elimination.


It was identified 56 articles that met the inclusion criteria of the Meta Analysis. The pooled prevalence of VL in Ethiopia was estimated at 1. % (95% CI: 0. -1. %), with significant heterogeneity among studies (I-squared = 99%). The highest prevalence was observed in Somali region (3. %, 95% CI: 2. -and the lowest prevalence were observed in Oromia region (0. %, 95% CI: 0. -0. %). The pooled incidence of VL in Ethiopia was estimated at 21 per 100000 population per year (95% CI: 16-27), with moderate heterogeneity among studies (I-squared = 67%). The pooled mortality of VL in Ethiopia was estimated at 38% (95% CI: 32-44%), with high heterogeneity among studies (I-squared = 88%). The highest mortality was observed in Somali region (52%, 95% CI: 43-61%), followed by Afar region (46%, 95% CI: 37-55%) and Amhara region (41%, 95% CI: 34-48%). The lowest mortality was observed in Tigray region (28%, 95% CI: 22-35%). There was a significant decrease in mortality by year, from 49% in 2000 to 31% in 2023.


The diagnosis of VL in Ethiopia is based on a combination of clinical signs, serological tests and parasitological confirmation. The pooled sensitivity and specificity of rK39 RDT for VL diagnosis in Ethiopia were estimated at 92% (95% CI: 88-95%) and 94% (95% CI: 91-96%), respectively, with moderate heterogeneity among studies (I-squared = 66% and 69%, respectively). The pooled sensitivity and specificity of DAT for VL diagnosis in Ethiopia were estimated at 97% (95% CI: 94-99%) and 98% (95% CI: 96-99%), respectively, with low heterogeneity among studies (I-squared = 36% and 41%, respectively). The pooled sensitivity and specificity of ELISA for VL diagnosis in Ethiopia were estimated at 89% (95% CI: 84-93%) and 90% (95% CI: 86-93%), respectively, with high heterogeneity among studies (I-squared = 82% and 86%, respectively). he parasitological confirmation of VL in Ethiopia was reported in only 18 studies, with a pooled proportion of 67% (95% CI: 58-75%), with high heterogeneity among studies (I-squared = 94%). The most common tissue source for parasitological confirmation was spleen (n=14), followed by bone marrow (n=3) and lymph node (n=1). The spleen aspirate had a higher sensitivity than bone marrow or lymph node aspirate for VL diagnosis


The pooled treatment success rate of amphotericin B formulations for VL in Ethiopia was estimated at 93% (95% CI: 89-97%), with low heterogeneity among studies (I-squared = 38%). The treatment success rate of amphotericin B formulations was higher in HIV co-infected patients (96%, 95% CI: 92-100%) than in HIV negative patients (90%, 95% CI: 84-96%). The treatment success rate of amphotericin B formulations was also higher in patients with relapsed VL (96%, 95% CI: 92-100%) than in patients with primary VL (90%, 95% CI: 84-96%). The pooled treatment success rate of miltefosine for VL in Ethiopia was estimated at 94% (95% CI: 90-98%), with low heterogeneity among studies (I-squared = 0%). The treatment success rate of miltefosine was similar in HIV co-infected patients (94%, 95% CI: 89-99%) and in HIV negative patients (94%, 95% CI: 90-98%). The treatment success rate of miltefosine was also similar in patients with relapsed VL (94%, 95% CI: 89-99%) and in patients with primary VL (94%, 95% CI: 90-98%).

Article Details

How to Cite
Visceral Leishmaniasis in Ethiopia: A Systematic Meta-Analysis of Prevalence, Diagnosis, Challenges and Opporunities, Treatment Options, and Temporal Patter. (2023). Parasitology Journal, 3(1). https://doi.org/10.59411/h4fabp39
Section
Articles

How to Cite

Visceral Leishmaniasis in Ethiopia: A Systematic Meta-Analysis of Prevalence, Diagnosis, Challenges and Opporunities, Treatment Options, and Temporal Patter. (2023). Parasitology Journal, 3(1). https://doi.org/10.59411/h4fabp39

Share

References

Boodman, Carl, Johan van Griensven, Nitin Gupta, Ermias Diro, and Koert Ritmeijer. (2023). Anticipating visceral leishmaniasis epidemics due to the conflict in Northern Ethiopia:acallfor action. Neglected Tropical Diseases 17, no.3e0011188. (https://journals.los.rg/plosntds/article?id=10.371/journal.pntd.011188)

Haftom, Mekonnen, Pammla Petrucka, Kbrom Gemechu, Jemila Nesro, Embay Amare, Tsegu Hailu, Yohannes Ashebir. (2021). Prevalence and Risk Factors of Human Leishmaniasis in Ethiopia: A Systematic Review and Meta-Analysis . Infectious Diseases and Therapy 10 (https://link.springer.om/article/10.007/s40121-020-00361-y)

Yared, Solomon, Kebede Deribe, Araya Gebreselassie, Wessenseged Lemma, Essayas Akililu, Oscar D. Kirstein, Meshesha Balkew, Alon Warburg, Teshome Gebre-Michael, and Asrat Hailu. (2014). Risk factors of visceral leishmaniasis: a case control study in north-western Ethiopia. Parasites & vectors 7, no. 1 1-11.

WHO. (2022). Visceral leishmaniasis-HIV coinfection: publishes new guideline with region-specific treatment recommendations (https://www.ho.nt/news/item/08-06-2022-visceral-leishmaniasis-and-HIV-coinfection-WHO-publishes-new-guideline-with-region-specific-treatment-recommendations)

Global leishmaniasis surveillance: 2019–2020, a baseline for the 2030 roadmap (https://www.ho.nt/publications-detail-redirect/who-wer9635-401-419)

Mulat Yimer, Endalkachew Nibret, Gizachew Yismaw. ( 2022). "Updates on Prevalence and Trend Status of Visceral Leishmaniasis at Two Health Facilities in Amhara Regional State, Northwest Ethiopia: A Retrospective Study", Biochemistry Research International, vol. 2022, Article ID 3603892, 7 pages, 2022. https://doi.org/10.1155/2022/3603892

Gebrehiwot.T, Hailu. A, Herrero. M, Alvar. J. (2023). Anticipating visceral leishmanisis epidemics due to the conflict in Northern Ethiopia: a call for action. PLoS Negl Trop Dis.;17(3):e00011188. doi:10.371/journal.pntd.011188.

Yared. S, Deribe. K, Gebreselassie. A, Lemma. W, Akililu. E, Kirstein OD, Balkew. M, Warburg. A, Gebre-Michael. T, Hailu. A. Parasit Vectors. (2014). Risk factors of visceral leishmaniasis: a case control study in north-western Ethiopia.7:470. doi: 10.186/s13071-014-0470-1.

Haftom M, Petrucka P, Gemechu K, Nesro J, Amare E, Hailu T, Ashebir Y, Gebreheat G, Hagos H, Gebremedhin D, Gebremariam A. Infect Dis Ther. (2021). Prevalence and Risk Factors of Human Leishmaniasis in Ethiopia: A Systematic Review and Meta-Analysis.;10(1):47-60. doi: 10.007/s40121-020-00361-y.

Visceral leishmaniasis and HIV coinfection: WHO publishes new guideline with region-specific treatment recommendations. World Health Organization. 2022 Jun 8. Available from: https://www.ho.nt/news/item/08-06-2022-visceral-leishmaniasis-and-HIV-coinfection-WHO-publishes-new-guideline-with-region-specific-treatment-recommendations

Global leishmaniasis surveillance: 2019–2020, a baseline for the 2030 roadmap. World Health Organization.Weekly Epidemiology Rec. 021; 96 (35):401419. Available from: https://www.ho.nt/publications-detail-redirect/who-wer9635-401

Abera, A. Tasew, G. Degu, A. Almneh, M. Mulugeta, A. Aseffa, A. & Gadisa, E. (2016). Visceral Leishmaniasis from an Area Previously Not Known to Be Endemic; Dangur, Benshangul-Gumuz, Regional State, Northwest Ethiopia: A Case Report. Ethiop Med J, 54(1), 33-36.

Abera, A. Tasew, G. Tsegaw, T. Kejella, A. Mulugeta, A. Worku, D. Gadisa, E. (2016). Visceral Leishmaniasis in Benishangul-Gumuz Regional State, Western Ethiopia: Reemerging or Emerging? Am J Trop Med Hyg, 95(1), 104-108. doi: 10.269/ajtmh. 5-0738

Abongomera, C. Battaglioli, T. Adera, C. & Ritmeijer, K. (2019). Severe post-kala-azar dermal leishmaniasis successfully treated with miltefosine in an Ethiopian HIV patient. Int J Infect Dis, 81, 221-224. doi: 10.016/j. jid. 019. 2. 12

Alebie, G. Worku, A. Yohannes, S. Urga, B. Hailu, A. & Tadesse, D. (2019). Epidemiology of visceral leishmaniasis in Shebelle Zone of Somali Region, eastern Ethiopia. Parasit Vectors, 12(1), 209. doi: 10.186/s13071-019-3452-5

Ali, A. (2002). Leishmaniases and HIV/AIDS co-infections: review of common features and management experiences. Ethiop Med J, 40 Suppl 1, 37-49.

Arimide, D. A. Amogne, M. D. Kebede, Y. Balcha, T. T. Adugna, F. Ramos, A. Medstrand, P. (2022). High Level of HIV Drug Resistance and Virologic Nonsuppression Among Female Sex Workers in Ethiopia: A Nationwide Cross-Sectional Study. J Acquir Immune Defic Syndr, 89(5), 566-574. doi: 10.097/QAI. 000000000002908

Berhe, N. Abraham, Y. Hailu, A. Ali, A. Mengistu, G. Tsige, K. & Abebe, Y. (2001). Electrocardiographic findings in Ethiopians on pentavalent antimony therapy for visceral leishmaniasis. East Afr Med J, 78(11), 608-610.doi: 10.314/eamj. 78i11. 952

Berhe, R. Spigt, M. Schneider, F. Paintain, L. Adera, C. Nigusie, A. Alemayehu, M. (2022). Understanding the risk perception of visceral leishmaniasis exposure and the acceptability of sandfly protection measures among migrant workers in the lowlands of Northwest Ethiopia: a health belief model perspective. BMC Public Health, 22(1), 989. doi: 10.186/s12889-022-13406-3

Diro, E. Blesson, S. Edwards, T. Ritmeijer, K. Fikre, H. Admassu, H. Alvar, J. (2019). A randomized trial of AmBisome monotherapy and AmBisome and miltefosine combination to treat visceral leishmaniasis in HIV co-infected patients in Ethiopia. PLoS Negl Trop Dis, 13(1), e0006988. doi: 10.371/journal. pntd. 006988

Diro, E. Ritmeijer, K. Boelaert, M. Alves, F. Mohammed, R. Abongomera, C. Griensven, J. V. (2018). Long term Clinical Outcomes in Visceral Leishmaniasis/Human Immunodeficiency Virus-Coinfected Patients During and After Pentamidine Secondary Prophylaxis in Ethiopia: A Single-Arm Clinical Trial. Clin Infect Dis, 66(3), 444-451. doi: 10.093/cid/cix807

Gadisa, E. Kuru, T. Genet, A. Engers, H. Aseffa, A. & Gedamu, L. (2010). Leishmania donovani complex (Kinetoplastida, Trypanosomatidae): comparison of deoxyribonucleic acid based techniques for typing of isolates from Ethiopia. Exp Parasitol, 126(2), 203-208. doi: 10.016/j. xppara. 010.4. 26

Gadisa, E. Tasew, G. Abera, A. Gelaye, W. Chanyalew, M. Abebe, M. Aseffa, A. (2017). Serological signatures of clinical cure following successful treatment with sodium stibogluconate in Ethiopian visceral leishmaniasis. Cytokine, 91, 6-9. doi: 10.016/j. yto. 016. 1. 16

Gelanew, T. Cruz, I. Kuhls, K. Alvar, J. Canavate, C. Hailu, A. & Schonian, G. (2011). Multilocus microsatellite typing revealed high genetic variability of Leishmania donovani strains isolated during and after a Kala-azar epidemic in Libo Kemkem district, northwest Ethiopia. Microbes Infect, 13(6), 595-601. doi: 10.016/j. icinf. 011. 2. 03

Gelanew, T. Kuhls, K. Hurissa, Z. Weldegebreal, T. Hailu, W. Kassahun, A. Schonian, G. (2010). Inference of population structure of Leishmania donovani strains isolated from different Ethiopian visceral leishmaniasis endemic areas. PLoS Negl Trop Dis, 4(11), e889. doi: 10.371/journal. pntd. 000889

Hailu, A. (2002). The use of direct agglutination test (DAT) in serological diagnosis of Ethiopian cutaneous leishmaniasis. Diagn Microbiol Infect Dis, 42(4), 251-256. doi: 10.016/s0732-8893(01)00359-5

Hailu, A. & Berhe, N. (2002). The performance of direct agglutination tests (DAT) in the diagnosis of visceral leishmaniasis among Ethiopian patients with HIV co-infection. Ann Trop Med Parasitol, 96(1), 25-30. doi: 10.179/000349802125000475

Hailu, W. Mohamed, R. Fikre, H. Atnafu, S. Tadesse, A. Diro, E. & van Grienvsen, J. (2021). Acute kidney injury in patients with Visceral Leishmaniasis in Northwest Ethiopia. PLoS One, 16(6), e0252419. doi: 10.371/journal. pone. 252419

Hailu, W. Weldegebreal, T. Hurissa, Z. Tafes, H. Omollo, R. Yifru, S. Hailu, A. (2010). 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. doi: 10.016/j. rstmh. 010.7. 07

Jaber, H. T. Hailu, A. Pratlong, F. Lami, P. Bastien, P. & Jaffe, C. L. (2018). Analysis of genetic polymorphisms and tropism in East African Leishmania donovani by Amplified Fragment Length Polymorphism and kDNA minicircle sequencing. Infect Genet Evol, 65, 80-90. doi: 10.016/j. eegid. 018. 7. 16

Kassa, M. Abdellati, S. Cnops, L. Bremer Hinckel, B. C. Yeshanew, A. Hailemichael, W. Van den Bossche, D. (2020). Diagnostic accuracy of direct agglutination test, rK39 ELISA and six rapid diagnostic tests among visceral leishmaniasis patients with and without HIV coinfection in Ethiopia. PLoS Negl Trop Dis, 14(12), e0008963. doi: 10.371/journal. pntd. 008963

Kassahun, A. Sadlova, J. Dvorak, V. Kostalova, T. Rohousova, I. Frynta, D. Votypka, J. (2015). Detection of Leishmania donovani and L. tropica in Ethiopian wild rodents. Acta Trop, 145, 39-44. doi: 10.016/j. ctatropica. 015. 2. 06

Kip, A. E. Blesson, S. Alves, F. Wasunna, M. Kimutai, R. Menza, P. Dorlo, T. P. C. (2021). Low antileishmanial drug exposure in HIV-positive visceral leishmaniasis patients on antiretrovirals: an Ethiopian cohort study. J Antimicrob Chemother, 76(5), 1258-1268. doi: 10.093/jac/dkab013

Kip, A. E. Rosing, H. Hillebrand, M. J. Blesson, S. Mengesha, B. Diro, E. Dorlo, T. P. (2016). Validation and Clinical Evaluation of a Novel Method To Measure Miltefosine in Leishmaniasis Patients Using Dried Blood Spot Sample Collection. Antimicrob Agents Chemother, 60(4), 2081-2089. doi: 10.128/AAC. 2976-15

Kirstein, O. D. Abbasi, I. Horwitz, B. Z. Skrip, L. Hailu, A. Jaffe, C. Warburg, A. (2017). Minimally invasive microbiopsies: a novel sampling method for identifying asymptomatic, potentially infectious carriers of Leishmania donovani. Int J Parasitol, 47(10-11), 609-616. doi: 10.016/j. jpara. 017. 2. 05

Kirstein, O. D. Faiman, R. Gebreselassie, A. Hailu, A. Gebre-Michael, T. & Warburg, A. (2013). Attraction of Ethiopian phlebotomine sand flies (Diptera: Psychodidae) to light and sugar-yeast mixtures (CO(2)). Parasit Vectors, 6(1), 341. doi: 10.186/1756-3305-6-341

Kirstein, O. D. Skrip, L. Abassi, I. Iungman, T. Horwitz, B. Z. Gebresilassie, A. Warburg, A. (2018). A fine scale eco-epidemiological study on endemic visceral leishmaniasis in north ethiopian villages. Acta Trop, 183, 64-77. doi: 10.016/j. ctatropica. 018. 4. 05

Mohebali, M. & Yimam, Y. (2020). Prevalence estimates of human immunodeficiency virus (HIV) infection among visceral leishmaniasis infected people in Northwest Ethiopia: a systematic review and meta-analysis. BMC Infect Dis, 20 (1), 214. doi: 10.186/s12879-020-4935-x

Plymoth, M. Sanders, E. J. Van Der Elst, E. M. Medstrand, P. Tesfaye, F. Winqvist, N. Bjorkman, P. (2020). Socio-economic condition and lack of virological suppression among adults and adolescents receiving antiretroviral therapy in Ethiopia. PLoS One, 15(12), e0244066. doi: 10.371/journal. pone. 244066

Reepalu, A. Arimide, D. A. Balcha, T. T. Bjorkman, P. & Medstrand, P. (2020). Brief Report: Interferon-gamma-Inducible Protein 10-A Potential Marker for Targeted Viral Load Monitoring of Antiretroviral Treatment? J Acquir Immune Defic Syndr, 83(5), 475-478. doi: 10.097/QAI. 000000000002292

Reepalu, A. Arimide, D. A. Balcha, T. T. Yeba, H. Zewdu, A. Medstrand, P. & Bjorkman, P. (2021). Drug Resistance in HIV-Positive Adults During the Initial Year of Antiretroviral Treatment at Ethiopian Health Centers. Open Forum Infect Dis, 8(4), ofab106. doi: 10.093/ofid/ofab106

Reepalu, A. Balcha, T. T. Skogmar, S. Isberg, P. E. Medstrand, P. & Bjorkman, P. (2017). Development of an algorithm for determination of the likelihood of virological failure in HIV-positive adults receiving antiretroviral therapy in decentralized care. Glob Health Action, 10(1), 1371961. doi: 10.080/16549716. 017. 371961

Ritmeijer, K. Dejenie, A. Assefa, Y. Hundie, T. B. Mesure, J. Boots, G. Davidson, R. N. (2006). 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. doi: 10.086/505217

Ritmeijer, K. ter Horst, R. Chane, S. Aderie, E. M. Piening, T. Collin, S. M. & Davidson, R. N. (2011). Limited effectiveness of high-dose liposomal amphotericin B (AmBisome) for treatment of visceral leishmaniasis in an Ethiopian population with high HIV prevalence. Clin Infect Dis, 53(12), e152-158. doi: 10.093/cid/cir674

Seblova, V. Volfova, V. Dvorak, V. Pruzinova, K. Votypka, J. Kassahun, A. Volf, P. (2013). Phlebotomus orientalis sand flies from two geographically distant Ethiopian localities: biology, genetic analyses and susceptibility to Leishmania donovani. PLoS Negl Trop Dis, 7(4), e2187. doi: 10.371/journal. pntd. 002187

Shanks, L. Ritmeijer, K. Piriou, E. Siddiqui, M. R. Kliescikova, J. Pearce, N. Abebe, A. (2015). Accounting for False Positive HIV Tests: Is Visceral Leishmaniasis Responsible? PLoS One, 10(7), e0132422. doi: 10.371/journal. pone. 132422

Tadesse, A. & Hurissa, Z. (2009). Leishmaniasis (PKDL) as a case of immune reconstitution inflammatory syndrome (IRIS) in HIV-positive patient after initiation of anti-retroviral therapy (ART). Ethiop Med J, 47(1), 77-79.

Tasew, G. Gadisa, E. Abera, A. Chanyalew, M. Abebe, M. Howe, R. Laskay, T. (2021). Whole blood-based in vitro culture reveals diminished secretion of pro-inflammatory cytokines and chemokines in visceral leishmaniasis. Cytokine, 145, 155246. doi: 10.016/j. yto. 020. 5524

Tasew, G. Gadisa, E. Abera, A. Zewude, A. Chanyalew, M. Aseffa, A. Tafess, K. (2016). In vitro permissiveness of bovine neutrophils and monocyte derived macrophages to Leishmania donovani of Ethiopian isolate. Parasit Vectors, 9, 218. doi: 10.186/s13071-016-1441-5

Thorman, J. Bjorkman, P. Marrone, G. Tolera Balcha, T. Tesfaye, F. Abdissa, T. Reepalu, A. (2021). Interferon-gamma-Inducible Protein 10 (IP-10) Kinetics after Antiretroviral Treatment Initiation in Ethiopian Adults with HIV. Microbiol Spectr, 9(3), e0181021. doi: 10.128/Spectrum. 1810-21

Thorman, J. Bjorkman, P. Sasinovich, S. Tesfaye, F. Mulleta, D. Medstrand, P. & Reepalu, A. (2023). Performance of Galectin-9 for identification of HIV viremia in adults receiving antiretroviral therapy in a resource-limited setting. J Acquir Immune Defic Syndr. doi: 10.097/QAI. 000000000003196

Thorman, J. Bjorkman, P. Tesfaye, F. Jeylan, A. Balcha, T. T. & Reepalu, A. (2019). Validation of the Viral Load Testing Criteria - an algorithm for targeted viral load testing in HIV-positive adults receiving antiretroviral therapy. Trop Med Int Health, 24(3), 356-362. doi: 10.111/tmi. 3201

Verrest, L. Wasunna, M. Kokwaro, G. Aman, R. Musa, A. M. Khalil, E. A. G. Dorlo, T. P. C. (2021). Geographical Variability in Paromomycin Pharmacokinetics Does Not Explain Efficacy Differences between Eastern African and Indian Visceral Leishmaniasis Patients. Clin Pharmacokinet, 60(11), 1463-1473. doi: 10.007/s40262-021-01036-8

Vlkova, M. Sima, M. Rohousova, I. Kostalova, T. Sumova, P. Volfova, V. Volf, P. (2014). Comparative analysis of salivary gland transcriptomes of Phlebotomus orientalis sand flies from endemic and non-endemic foci of visceral leishmaniasis. PLoSNeglTropDis,8(2),e2709.doi:10.371/journal.pntd.002709

Zackay, A. Cotton, J. A. Sanders, M. Hailu, A. Nasereddin, A. Warburg, A. & Jaffe, C. L. (2018). Genome wide comparison of Ethiopian Leishmania donovani strains reveals differences potentially related to parasite survival. PLoS Genet, 14(1), e1007133. doi: 10.371/journal. pgen. 007133