A randomized trial of AmBisome monotherapy and AmBisome and miltefosine combination to treat visceral leishmaniasis in HIV co-infected patients in Ethiopia

Ermias Diro, Severine Blesson, Tansy Edwards, Koert Ritmeijer, Helina Fikre, Henok Admassu, Aderajew Kibret, Sally J Ellis, Clelia Bardonneau, Eduard E Zijlstra, Peninah Soipei, Brian Mutinda, Raymond Omollo, Robert Kimutai, Gabriel Omwalo, Monique Wasunna, Fentahun Tadesse, Fabiana Alves, Nathalie Strub-Wourgaft, Asrat Hailu, Neal Alexander, Jorge Alvar, Ermias Diro, Severine Blesson, Tansy Edwards, Koert Ritmeijer, Helina Fikre, Henok Admassu, Aderajew Kibret, Sally J Ellis, Clelia Bardonneau, Eduard E Zijlstra, Peninah Soipei, Brian Mutinda, Raymond Omollo, Robert Kimutai, Gabriel Omwalo, Monique Wasunna, Fentahun Tadesse, Fabiana Alves, Nathalie Strub-Wourgaft, Asrat Hailu, Neal Alexander, Jorge Alvar

Abstract

Background: Visceral leishmaniasis (VL) in human immunodeficiency virus (HIV) co-infected patients requires special case management. AmBisome monotherapy at 40 mg/kg is recommended by the World Health Organization. The objective of the study was to assess if a combination of a lower dose of AmBisome with miltefosine would show acceptable efficacy at the end of treatment.

Methodology/principal findings: An open-label, non-comparative randomized trial of AmBisome (30 mg/kg) with miltefosine (100 mg/day for 28 days), and AmBisome monotherapy (40 mg/kg) was conducted in Ethiopian VL patients co-infected with HIV (NCT02011958). A sequential design was used with a triangular continuation region. The primary outcome was parasite clearance at day 29, after the first round of treatment. Patients with clinical improvement but without parasite clearance at day 29 received a second round of the allocated treatment. Efficacy was evaluated again at day 58, after completion of treatment. Recruitment was stopped after inclusion of 19 and 39 patients in monotherapy and combination arms respectively, as per pre-specified stopping rules. At D29, intention-to-treat efficacy in the AmBisome arm was 70% (95% CI 45-87%) in the unadjusted analysis, and 50% (95% CI 27-73%) in the adjusted analysis, while in the combination arm, it was 81% (95% CI 67-90%) and 67% (95% CI 48-82%) respectively. At D58, the adjusted efficacy was 55% (95% CI 32-78%) in the monotherapy arm, and 88% (95% CI 79-98%) in the combination arm. No major safety concerns related to the study medication were identified. Ten SAEs were observed within the treatment period, and 4 deaths unrelated to the study medication.

Conclusions/significance: The extended treatment strategy with the combination regimen showed the highest documented efficacy in HIV-VL patients; these results support a recommendation of this regimen as first-line treatment strategy for HIV-VL patients in eastern Africa.

Trial registration number: www.clinicaltrials.gov NCT02011958.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Treatment strategy.
Fig 1. Treatment strategy.
Fig 2. Sequential analysis.
Fig 2. Sequential analysis.
The vertical axis, (Z), is the number of observed treatment successes minus the number of expected successes. The higher the value represented on that vertical axis, the more favorable (better efficacy observed). The horizontal axis (V) is proportional to the number of patients who, over time, have been evaluated at day 29. Hence, over time, each arm’s data points extend to the right. Each arm starts inside the triangle (green area) and recruitment is stopped on crossing either the upper (blue area) or the lower boundary (pink area). Recruitment to the AmBisome monotherapy arm was stopped after the first interim analysis (I1) and to the combination arm after the second interim analysis (I2). Also shown are the final results for each arm, including those who had not reached the primary endpoint (day29) when the interim analysis was performed and the recruitment was stopped. These final results take the trajectory of each arm well away from the triangular boundary, which is known as ‘over-run'. The two numbers after each label are the numbers of patients included in that analysis and the number of these with treatment success at day 29.
Fig 3. Trial participant flow.
Fig 3. Trial participant flow.
Fig 4. CD4-cell counts at baseline (D0),…
Fig 4. CD4-cell counts at baseline (D0), D29 and D58.
Categories are Cat 1 =

References

    1. Cota GF, de Sousa MR, Rabello A. Predictors of visceral leishmaniasis relapse in hiv-infected patients: A systematic review. PLoS Negl Trop Dis. 2011;5 10.1371/journal.pntd.0001153
    1. Diro E, Lynen L, Ritmeijer K, Boelaert M, Hailu A, van Griensven J. Visceral Leishmaniasis and HIV Coinfection in East Africa. PLoS Negl Trop Dis. 2014;8 10.1371/journal.pntd.0002869
    1. Alvar J, Aparicio P, Aseffa A, Den Boer M, Cañavate C, Dedet JP, et al. The relationship between leishmaniasis and AIDS: The second 10 years. Clinical Microbiology Reviews. 2008. pp. 334–359. 10.1128/CMR.00061-07
    1. Monge-Maillo B, Norman FF, Cruz I, Alvar J, López-Vélez R. Visceral Leishmaniasis and HIV Coinfection in the Mediterranean Region. PLoS Negl Trop Dis. 2014;8 10.1371/journal.pntd.0003021
    1. Lindoso JA, Cota GF, da Cruz AM, Goto H, Maia-Elkhoury ANS, Romero GAS, et al. Visceral Leishmaniasis and HIV Coinfection in Latin America. PLoS Negl Trop Dis. 2014;8 10.1371/journal.pntd.0003136
    1. Burza S, Mahajan R, Sanz MG, Sunyoto T, Kumar R, Mitra G, et al. HIV and visceral leishmaniasis coinfection in Bihar, India: An underrecognized and underdiagnosed threat against elimination. Clin Infect Dis. 2014;59: 552–555. 10.1093/cid/ciu333
    1. López-Vélez R. The impact of highly active antiretroviral therapy (HAART) on visceral leishmaniasis in Spanish patients who are co-infected with HIV. Ann Trop Med Parasitol. 2003;97 Suppl 1: 143–147. 10.1179/000349803225002615
    1. Argaw D, Mulugeta A, Herrero M, Nombela N, Teklu T, Tefera T, et al. Risk factors for visceral Leishmaniasis among residents and migrants in Kafta-Humera, Ethiopia. PLoS Negl Trop Dis. 2013;7: e2543 10.1371/journal.pntd.0002543
    1. Tsegaw T, Gadisa E, Seid A, Abera A, Teshome A, Mulugeta A, et al. Identification of environmental parameters and risk mapping of visceral leishmaniasis in Ethiopia by using geographical information systems and a statistical approach. Geospat Health. 2013;7: 299–308. 10.4081/gh.2013.88
    1. WHO. Control of the Leishmaniases: Report of a meeting of the WHO expert committee on the control of the Leishmaniases, Geneva, 22–26 March 2010. 2010.
    1. Guideline for diagnosis, treatment and prevention of leishmaniasis in Ethiopia. 2013.
    1. Ritmeijer K, Ter Horst R, Chane S, Aderie EM, Piening T, Collin SM, et al. 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. 2011;53 10.1093/cid/cir674
    1. Diro E, Lynen L, Mohammed R, Boelaert M, Hailu A, van Griensven J. High Parasitological Failure Rate of Visceral Leishmaniasis to Sodium Stibogluconate among HIV Co-infected Adults in Ethiopia. PLoS Negl Trop Dis. 2014;8 10.1371/journal.pntd.0002875
    1. Ritmeijer K, Veeken H, Melaku Y, Leal G, Amsalu R, Seaman J, et al. Ethiopian visceral leishmaniasis: Generic and proprietary sodium stibogluconate are equivalent; HIV co-infected patients have a poor outcome. Trans R Soc Trop Med Hyg. 2001;95: 668–672. 10.1016/S0035-9203(01)90110-5
    1. Molina R, Lohse JM, Pulido F, Laguna F, López-Vélez R, Alvar J. Infection of sand flies by humans coinfected with Leishmania infantum and human immunodeficiency virus. Am J Trop Med Hyg. 1999;60: 51–53.
    1. ter Horst R, Collin SM, Ritmeijer K, Bogale A, Davidson RN. Concordant HIV Infection and Visceral Leishmaniasis in Ethiopia: The Influence of Antiretroviral Treatment and Other Factors on Outcome. Clin Infect Dis. 2008;46: 1702–1709. 10.1086/587899
    1. Diro E, Ritmeijer K, Boelaert M, Alves F, Mohammed R, Abongomera C, et al. Use of Pentamidine As Secondary Prophylaxis to Prevent Visceral Leishmaniasis Relapse in HIV Infected Patients, the First Twelve Months of a Prospective Cohort Study. PLoS Negl Trop Dis. 2015;9 10.1371/journal.pntd.0004087
    1. Ranque S, Badiaga S, Delmont J, Brouqui P. Triangular test applied to the clinical trial of azithromycin against relapses in Plasmodium vivax infections. Malar J. 2002;1: 1–7. 10.1186/1475-2875-1-1
    1. Wasunna M, Njenga S, Balasegaram M, Alexander N, Omollo R, Edwards T, et al. Efficacy and Safety of AmBisome in Combination with Sodium Stibogluconate or Miltefosine and Miltefosine Monotherapy for African Visceral Leishmaniasis: Phase II Randomized Trial. PLoS Negl Trop Dis. 2016; 10.1371/journal.pntd.0004880
    1. Gelanew T, Cruz I, Kuhls K, Alvar J, Cañavate C, Hailu A, et al. 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. 2011; 10.1016/j.micinf.2011.02.003
    1. Dereure J, El-Safi SH, Bucheton B, Boni M, Kheir MM, Davoust B, et al. Visceral leishmaniasis in eastern Sudan: Parasite identification in humans and dogs; Host-parasite relationships. Microbes Infect. 2003; 10.1016/j.micinf.2003.07.003
    1. National Institute of Cancer. Common Terminology Criteria for Adverse Events (CTCAE), Version 4.0, DCTD, CTI, NIH, DHHS [Internet]. NIH Publication. 2009. 10.1080/00140139.2010.489653
    1. Omollo R, Ochieng M, Mutinda B, Omollo T, Owiti R, Okeyo S, et al. Innovative Approaches to Clinical Data Management in Resource Limited Settings Using Open-Source Technologies. PLoS Negl Trop Dis. 2014;8 10.1371/journal.pntd.0003134
    1. Lakens D, Lakens D. Performing high-powered studies efficiently with sequential analyses Special issue article: Methods and statistics in social psychology: Refinements and new developments Performing high-powered studies efficiently with sequential analyses. Eur J Soc Psychol. 2014;44: 701–710. 10.1002/ejsp.2023
    1. Whitehead J. On the bias of maximum likelihood estimation following a sequential test. Biometrika. 1986;73: 573–581.
    1. Whitehead J. The Design and Analysis of Sequential Clinical Trials. 1st edn. Horwood CE, editor. 1983.
    1. Liu B. A Simple Low-Bias Estimate following a Sequential Test with Linear Boundaries Crossing boundaries: Statistical Essays in Honor of Jack Hall Lecture Notes—Monograph Series Beachwood, Ohio: Institute of Mathematical Statistics. 2003. pp. 47–58.
    1. Allison A, Edwards T, Omollo R, Alves F, Magirr D, Alexander NDE. Generalizing boundaries for triangular designs, and efficacy estimation at extended follow-ups. Trials. Trials; 2015;16: 1–11. 10.1186/1745-6215-16-1
    1. Gilead Sciences. Ambisome Package Insert. 2012. pp. 1–27.
    1. Paladin Therapeutics. Impavido Package Insert US. 2014. pp. 1–27.
    1. Ritmeijer K, Dejenie A, Assefa Y, Hundie TB, Mesure J, Boots G, 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. 2006;43: 357–364. 10.1086/505217
    1. Laguna F, Videla S, Jiménez-Mejías ME, Sirera G, Torre-Cisneros J, Ribera E, et al. Amphotericin B lipid complex versus meglumine antimoniate in the treatment of visceral leishmaniasis in patients infected with HIV: a randomized pilot study. J Antimicrob Chemother. 2003;52: 464–8. 10.1093/jac/dkg356
    1. Sundar S, Mehta H, Suresh a V, Singh SP, Rai M, Murray HW. Amphotericin B treatment for Indian visceral leishmaniasis: conventional versus lipid formulations. Clin Infect Dis. 2004;38: 377–83. 10.1086/380971
    1. Mahajan R, Das P, Isaakidis P, Sunyoto T, Sagili KD, Lima MA, et al. Combination Treatment for Visceral Leishmaniasis Patients Coinfected with Human Immunodeficiency Virus in India. Clin Infect Dis. 2015;61: 1255–1262. 10.1093/cid/civ530

Source: PubMed

3
Iratkozz fel