1
|
Monge-Maillo B, Roger-Zapata D, Dronda F, Carrillo E, Moreno J, Corbacho-Loarte MD, Gayoso Cantero D, Martín O, Chamorro-Tojeiro S, Perez-Molina JA, Norman F, González-Sanz M, López-Vélez R. Case Report: Long-Term Follow-Up of Visceral Leishmaniasis and HIV Coinfected Patients Without Relapse: Lymphoproliferative Response After Stimulation with Soluble Leishmania Antigen. Microorganisms 2025; 13:686. [PMID: 40142579 PMCID: PMC11944632 DOI: 10.3390/microorganisms13030686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Highly active antiretroviral therapy (HAART) has reduced the incidence of VL/HIV dramatically. However, HAART only partially prevents relapses, with one-year relapse rates ranging from 30 to 60%. Consequently, secondary prophylaxis is recommended for patients with <200 CD4+ cells/μL. In clinical practice, characterizing cellular immune response could help estimate the risk of relapse in VL/HIV coinfected patients. In this study, the lymphoproliferative response after stimulation with soluble Leishmania antigen was assessed in 2022 and 2023 in three cases of VL/HIV coinfection with long-term follow-up (17, 8 and 19 years). PCR and rK-39 results for Leishmania, HIV viral load, CD4 cell count, proliferation index, IFN-γ, IL-2, IP-10, IL-10 and TNF-α were determined. Heterogeneous results were obtained, with only one patient having developed specific cellular immunity against Leishmania. No cases of relapse were observed. The heterogeneity of lymphoproliferative test results in the three cases described highlights the need to identify surrogate markers of cure to guide maintenance or withdrawal of prophylaxis.
Collapse
Affiliation(s)
- Begoña Monge-Maillo
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
| | | | - Fernando Dronda
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
- Department of Infectious Diseases, Ramón y Cajal University Hospital, IRICYS, 28034 Madrid, Spain
| | - Eugenia Carrillo
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
- Unidad de Leishmaniasis y Enfermedad de Chagas, WHO Collaborating Center for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Moreno
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
- Unidad de Leishmaniasis y Enfermedad de Chagas, WHO Collaborating Center for Leishmaniasis, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Dolores Corbacho-Loarte
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
| | - Diego Gayoso Cantero
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
| | - Oihane Martín
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
- Microbiology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Sandra Chamorro-Tojeiro
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
| | - Jose A. Perez-Molina
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
| | - Francesca Norman
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
| | - Marta González-Sanz
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
- University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Rogelio López-Vélez
- National Reference Unit for Tropical Diseases, Department of Infectious Diseases, Ramón y Cajal University Hospital, WHO Collaborating Center for Clinical Management of Leishmaniasis, Ramón y Cajal Research Institute (IRYCIS), 28034 Madrid, Spain; (M.D.C.-L.); (D.G.C.); (S.C.-T.); (F.N.); (M.G.-S.); (R.L.-V.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.D.); (E.C.); (J.M.); (O.M.)
| |
Collapse
|
2
|
de Vrij N, Pollmann J, Rezende AM, Ibarra-Meneses AV, Pham TT, Hailemichael W, Kassa M, Bogale T, Melkamu R, Yeshanew A, Mohammed R, Diro E, Maes I, Domagalska MA, Landuyt H, Vogt F, van Henten S, Laukens K, Cuypers B, Meysman P, Beyene H, Sisay K, Kibret A, Mersha D, Ritmeijer K, van Griensven J, Adriaensen W. Persistent T cell unresponsiveness associated with chronic visceral leishmaniasis in HIV-coinfected patients. Commun Biol 2024; 7:524. [PMID: 38702419 PMCID: PMC11068874 DOI: 10.1038/s42003-024-06225-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
A large proportion of HIV-coinfected visceral leishmaniasis (VL-HIV) patients exhibit chronic disease with frequent VL recurrence. However, knowledge on immunological determinants underlying the disease course is scarce. We longitudinally profiled the circulatory cellular immunity of an Ethiopian HIV cohort that included VL developers. We show that chronic VL-HIV patients exhibit high and persistent levels of TIGIT and PD-1 on CD8+/CD8- T cells, in addition to a lower frequency of IFN-γ+ TIGIT- CD8+/CD8- T cells, suggestive of impaired T cell functionality. At single T cell transcriptome and clonal resolution, the patients show CD4+ T cell anergy, characterised by a lack of T cell activation and lymphoproliferative response. These findings suggest that PD-1 and TIGIT play a pivotal role in VL-HIV chronicity, and may be further explored for patient risk stratification. Our findings provide a strong rationale for adjunctive immunotherapy for the treatment of chronic VL-HIV patients to break the recurrent disease cycle.
Collapse
Affiliation(s)
- Nicky de Vrij
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Julia Pollmann
- Department of Medical Oncology, University Hospital Heidelberg, National Center for Tumor Diseases (NCT) Heidelberg, 69120, Heidelberg, Germany
| | - Antonio M Rezende
- Department of Microbiology, Aggeu Magalhães Institute-FIOCRUZ/PE, Recife, Brazil
| | - Ana V Ibarra-Meneses
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Thao-Thy Pham
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Wasihun Hailemichael
- Department of Immunology and Molecular Biology, Faculty of Biomedical Sciences, University of Gondar, Gondar, Ethiopia
| | - Mekibib Kassa
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Tadfe Bogale
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Roma Melkamu
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Arega Yeshanew
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Rezika Mohammed
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Ermias Diro
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Ilse Maes
- Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Malgorzata A Domagalska
- Molecular Parasitology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Hanne Landuyt
- Clinical Trial Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Florian Vogt
- National Centre for Epidemiology and Population Health, The Australian National University, Canberra, 2601, Australia
- The Kirby Institute, University of New South Wales, Sydney, 2052, Australia
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Saskia van Henten
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Bart Cuypers
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, 2020, Antwerp, Belgium
| | | | | | | | | | | | - Johan van Griensven
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Wim Adriaensen
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium.
| |
Collapse
|
3
|
Singh R, Kashif M, Srivastava P, Manna PP. Recent Advances in Chemotherapeutics for Leishmaniasis: Importance of the Cellular Biochemistry of the Parasite and Its Molecular Interaction with the Host. Pathogens 2023; 12:pathogens12050706. [PMID: 37242374 DOI: 10.3390/pathogens12050706] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Leishmaniasis, a category 1 neglected protozoan disease caused by a kinetoplastid pathogen called Leishmania, is transmitted through dipteran insect vectors (phlebotomine, sand flies) in three main clinical forms: fatal visceral leishmaniasis, self-healing cutaneous leishmaniasis, and mucocutaneous leishmaniasis. Generic pentavalent antimonials have long been the drug of choice against leishmaniasis; however, their success is plagued with limitations such as drug resistance and severe side effects, which makes them redundant as frontline therapy for endemic visceral leishmaniasis. Alternative therapeutic regimens based on amphotericin B, miltefosine, and paromomycin have also been approved. Due to the unavailability of human vaccines, first-line chemotherapies such as pentavalent antimonials, pentamidine, and amphotericin B are the only options to treat infected individuals. The higher toxicity, adverse effects, and perceived cost of these pharmaceutics, coupled with the emergence of parasite resistance and disease relapse, makes it urgent to identify new, rationalized drug targets for the improvement in disease management and palliative care for patients. This has become an emergent need and more relevant due to the lack of information on validated molecular resistance markers for the monitoring and surveillance of changes in drug sensitivity and resistance. The present study reviewed the recent advances in chemotherapeutic regimens by targeting novel drugs using several strategies including bioinformatics to gain new insight into leishmaniasis. Leishmania has unique enzymes and biochemical pathways that are distinct from those of its mammalian hosts. In light of the limited number of available antileishmanial drugs, the identification of novel drug targets and studying the molecular and cellular aspects of these drugs in the parasite and its host is critical to design specific inhibitors targeting and controlling the parasite. The biochemical characterization of unique Leishmania-specific enzymes can be used as tools to read through possible drug targets. In this review, we discuss relevant metabolic pathways and novel drugs that are unique, essential, and linked to the survival of the parasite based on bioinformatics and cellular and biochemical analyses.
Collapse
Affiliation(s)
- Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Mohammad Kashif
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Prateek Srivastava
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
4
|
Saini I, Joshi J, Kaur S. Unwelcome prevalence of leishmaniasis with several other infectious diseases. Int Immunopharmacol 2022; 110:109059. [DOI: 10.1016/j.intimp.2022.109059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022]
|
5
|
Li S, Li Y, Deng B, Yan J, Wang Y. Identification of the Differentially Expressed Genes Involved in the Synergistic Neurotoxicity of an HIV Protease Inhibitor and Methamphetamine. Curr HIV Res 2020; 17:290-303. [PMID: 31550215 DOI: 10.2174/1570162x17666190924200354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/23/2019] [Accepted: 09/05/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND The abuse of psychostimulants such as methamphetamine (METH) is common in human immunodeficiency virus (HIV)-infected individuals. Acquired immunodeficiency syndrome (AIDS) patients taking METH and antiretroviral drugs could suffer severe neurologic damage and cognitive impairment. OBJECTIVE To reveal the underlying neuropathologic mechanisms of an HIV protease inhibitor (PI) combined with METH, growth-inhibition tests of dopaminergic cells and RNA sequencing were performed. METHODS A combination of METH and PI caused more growth inhibition of dopaminergic cells than METH alone or a PI alone. Furthermore, we identified differentially expressed gene (DEG) patterns in the METH vs. untreated cells (1161 genes), PI vs. untreated cells (16 genes), METH-PI vs. PI (3959 genes), and METH-PI vs. METH groups (14 genes). RESULTS The DEGs in the METH-PI co-treatment group were verified in the brains of a mouse model using quantitative polymerase chain reaction and were involved mostly in the regulatory functions of cell proliferation and inflammation. CONCLUSION Such identification of key regulatory genes could facilitate the study of their neuroprotective potential in the users of METH and PIs.
Collapse
Affiliation(s)
- Sangsang Li
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Immunology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yanfei Li
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Department of Immunology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bingpeng Deng
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Reguera RM, Elmahallawy EK, García-Estrada C, Carbajo-Andrés R, Balaña-Fouce R. DNA Topoisomerases of Leishmania Parasites; Druggable Targets for Drug Discovery. Curr Med Chem 2019; 26:5900-5923. [DOI: 10.2174/0929867325666180518074959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/15/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
DNA topoisomerases (Top) are a group of isomerase enzymes responsible for controlling the topological problems caused by DNA double helix in the cell during the processes of replication, transcription and recombination. Interestingly, these enzymes have been known since long to be key molecular machines in several cellular processes through overwinding or underwinding of DNA in all living organisms. Leishmania, a trypanosomatid parasite responsible for causing fatal diseases mostly in impoverished populations of low-income countries, has a set of six classes of Top enzymes. These are placed in the nucleus and the single mitochondrion and can be deadly targets of suitable drugs. Given the fact that there are clear differences in structure and expression between parasite and host enzymes, numerous studies have reported the therapeutic potential of Top inhibitors as antileishmanial drugs. In this regard, numerous compounds have been described as Top type IB and Top type II inhibitors in Leishmania parasites, such as camptothecin derivatives, indenoisoquinolines, indeno-1,5- naphthyridines, fluoroquinolones, anthracyclines and podophyllotoxins. The aim of this review is to highlight several facts about Top and Top inhibitors as potential antileishmanial drugs, which may represent a promising strategy for the control of this disease of public health importance.
Collapse
Affiliation(s)
- Rosa M. Reguera
- Department of Biomedical Sciences, University of Leon (ULE), Leon, Spain
| | | | | | | | | |
Collapse
|
7
|
Wang JL, Elsheikha HM, Li TT, He JJ, Bai MJ, Liang QL, Zhu XQ, Cong W. Efficacy of antiretroviral compounds against Toxoplasma gondii in vitro. Int J Antimicrob Agents 2019; 54:814-819. [PMID: 31479744 DOI: 10.1016/j.ijantimicag.2019.08.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/14/2019] [Accepted: 08/24/2019] [Indexed: 12/17/2022]
Abstract
The obligate intracellular parasite Toxoplasma gondii can infect nearly all warm-blooded animals, including humans. Although infection with this parasite is generally benign, severe illness may occur in infected individuals if their immunity becomes less competent, such as in human immunodeficiency virus (HIV)-infected patients. In this study, the inhibitory activity of 44 commonly used antiretroviral compounds was determined against T. gondii in vitro. Of the 44 tested antiretroviral compounds, 14 showed potency against T. gondii at IC50 concentrations (concentration inhibiting T. gondii tachyzoite growth by 50%) ranging from 1.18 ± 2.21 µM (nelfinavir) to 18.89 ± 1.87 µM (trovirdine). Of the 14 potent antiretroviral compounds, 7 are HIV-1 protease inhibitors. This study also investigated whether co-administration of these 14 antiretroviral compounds interferes with the anti-T. gondii activity of existing anti-T. gondii drugs, namely sulfadiazine and pyrimethamine. The results showed no significant interaction between any of the 14 tested antiretroviral compounds and pyrimethamine or sulfadiazine. These results warrant investigation of whether administration of the lead antiretroviral drugs with highly potent anti-T. gondii activity to HIV patients may help to limit the occurrence of toxoplasmic encephalitis.
Collapse
Affiliation(s)
- Jin-Lei Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China.
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | - Ting-Ting Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China
| | - Meng-Jie Bai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China
| | - Qin-Li Liang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China
| | - Wei Cong
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, P.R. China; College of Marine Science, Shandong University at Weihai, Weihai, Shandong Province 264209, P.R. China.
| |
Collapse
|
8
|
Mastroianni A, Gaibani P, Rossini G, Vocale C, Re MC, Ravaglia G, Sambri V, Varani S. Two cases of relapsed HIV-associated visceral leishmaniasis successfully treated with combination therapy. AIDS Res Ther 2018; 15:27. [PMID: 30572924 PMCID: PMC6300903 DOI: 10.1186/s12981-018-0215-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/11/2018] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The management of visceral leishmaniasis (VL) in HIV-infected patients is often complex with patients experiencing higher mortality rates, more toxic side effects and a higher possibility of treatment failure and relapse than HIV-negative individuals with VL. CASE PRESENTATION We report on successful salvage therapy in two HIV-infected patients suffering with disseminated cutaneous and visceral leishmaniasis, recalcitrant to therapy with liposomal amphotericin B. After the employment of combination anti-leishmanial treatment, parasite genomes were not detectable up to the last follow up visit, 57 and 78 weeks after treatment onset, respectively. CD4+ lymphocyte counts fluctuated over time, but were generally higher than counts detected at treatment onset, which likely contributed to protection against VL relapse. CONCLUSIONS Results achieved with the anti-leishmanial combination treatment were promising, but are based on only two patients. Future investigation is necessary to confirm the efficacy of this salvage therapy in sustaining the immunological response and control of VL.
Collapse
Affiliation(s)
- Antonio Mastroianni
- Unit of Infectious and Tropical Diseases, St. Annunziata Hospital, Cosenza, Italy
| | - Paolo Gaibani
- Unit of Microbiology, Regional Reference Centre for Microbiological Emergencies (CRREM), St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Giada Rossini
- Unit of Microbiology, Regional Reference Centre for Microbiological Emergencies (CRREM), St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Caterina Vocale
- Unit of Microbiology, Regional Reference Centre for Microbiological Emergencies (CRREM), St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Maria Carla Re
- Unit of Microbiology, Regional Reference Centre for Microbiological Emergencies (CRREM), St. Orsola-Malpighi University Hospital, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | - Vittorio Sambri
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Unit of Microbiology, the Greater Romagna Area Hub Laboratory, Pievesestina (Forlì-Cesena), Italy
| | - Stefania Varani
- Unit of Microbiology, Regional Reference Centre for Microbiological Emergencies (CRREM), St. Orsola-Malpighi University Hospital, Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| |
Collapse
|
9
|
Ferreira GR, Castelo Branco Ribeiro JC, Meneses Filho A, de Jesus Cardoso Farias Pereira T, Parente DM, Pereira HF, Carlos da Silva J, Zacarias DA, Vieira da Silva L, Medeiros Faustino SK, Almeida Neto WS, Costa DL, Lopes de Mendonça I, Nery Costa CH. Human Competence to Transmit Leishmania infantum to Lutzomyia longipalpis and the Influence of Human Immunodeficiency Virus Infection. Am J Trop Med Hyg 2018; 98:126-133. [PMID: 29141704 DOI: 10.4269/ajtmh.16-0883] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Visceral leishmaniasis (VL) caused by Leishmania infantum is a lethal disease transmitted by sand flies. Although, considered a zoonosis with dogs held as the main reservoirs, humans are also sources of infection. Therefore, control policies currently focused on dog culling may need to consider that VL and human immunodeficiency virus (HIV)/VL patients may also be infectious, contributing to transmission. Reservoir competence of patients with VL without and with HIV infection and of persons asymptomatically infected with Leishmania was assessed by xenodiagnosis with the vector Lutzomyia longipalpis. Parasites in sand fly's guts were identified by using optical microscopy and by conventional polymerase chain reaction (PCR). Leishmania infantum blood parasite burden was determined by quantitative PCR. Among the 61 participants, 27 (44%) infected sand flies as seen by microscopy or PCR. When infectiousness was assessed by microscopy, xenodiagnosis was positive in five (25%) patients not infected with HIV, whereas nine (45%) of those harboring HIV were positive. Among the 19 asymptomatic patients four (21%) infected sand flies only demonstrated by PCR. One (50%) asymptomatic patient with HIV had a positive xenodiagnosis by microscopy. 9/372 (2.4%) and 37/398 (9.2%) sand flies were infected when feeding in patients without and with HIV, respectively. Infectiousness was poorly correlated with quantitative PCR. The study shows that asymptomatic humans are capable of transmitting L. infantum, that ill persons with HIV infection are more infectious to sand flies, and that humans are more important reservoirs than previously thought. This fact may be considered when designing control policies for zoonotic VL.
Collapse
Affiliation(s)
- Gabriel Reis Ferreira
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil.,Graduation Program in Sciences and Health, Federal University of Piauí, Teresina, Brazil
| | - José Carlos Castelo Branco Ribeiro
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil.,Graduation Program in Sciences and Health, Federal University of Piauí, Teresina, Brazil
| | - Antônio Meneses Filho
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil
| | | | - Daniela Moura Parente
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil
| | - Humberto Feitosa Pereira
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil
| | - Jailthon Carlos da Silva
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil.,Graduation Program in Sciences and Health, Federal University of Piauí, Teresina, Brazil
| | - Danielle Alves Zacarias
- Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil
| | | | | | | | - Dorcas Lamounier Costa
- Graduation Program in Sciences and Health, Federal University of Piauí, Teresina, Brazil.,Maternal and Child Department, Federal University of Piauí, Teresina, Brazil.,Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil
| | | | - Carlos Henrique Nery Costa
- Graduation Program in Sciences and Health, Federal University of Piauí, Teresina, Brazil.,Department of Community Medicine, Federal University of Piauí, Teresina, Brazil.,Leishmaniasis Research Laboratory at Natan Portella Tropical Diseases Institute, Teresina, Brazil
| |
Collapse
|
10
|
Lindoso JAL, Moreira CHV, Cunha MA, Queiroz IT. Visceral leishmaniasis and HIV coinfection: current perspectives. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2018; 10:193-201. [PMID: 30410407 PMCID: PMC6197215 DOI: 10.2147/hiv.s143929] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Visceral leishmaniasis (VL) is caused by Leishmania donovani and Leishmania infantum. The burden of VL is concentrated in tropical and subtropical areas; however, HIV infection has spread VL over a hyperendemic area. Several outcomes are observed as a result of VL–HIV coinfection. Impacts are observed in immunopathogenesis, clinical manifestation, diagnosis, and therapeutic response. Concerning clinical manifestation, typical and unusual manifestation has been observed during active VL in HIV-infected patient, as well as alteration in immunoresponse, inducing greater immunosuppression by low CD4 T-lymphocyte count or even by induction of immunoactivation, with cell senescence. Serological diagnosis of VL in the HIV-infected is poor, due to low humoral response, characterized by antibody production, so parasitological methods are more recommended. Another important and even more challenging point is the definition of the best therapeutic regimen for VL in HIV-coinfected patients, because in this population there is greater failure and consequently higher mortality. The challenge of better understanding immunopathogenesis in order to obtain more effective therapies is one of the crucial points to be developed. The combination of drugs and the use of secondary prophylaxis associated with highly active antiretroviral therapy may be the best tool for treatment of HIV coinfection. Some derivatives from natural sources have action against Leishmania; however, studies have been limited to in vitro evaluation, without clinical trials.
Collapse
Affiliation(s)
- José Angelo Lauletta Lindoso
- Instituto de Infectologia Emilio Ribas, São Paulo, Brazil, .,Nucleo de Medicina Tropical, Universidade de Brasília, Brasília, Brazil, .,Laboratorio de Soroepidemiologia, Institutode Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil,
| | - Carlos Henrique Valente Moreira
- Instituto de Infectologia Emilio Ribas, São Paulo, Brazil, .,Laboratorio de Parasitologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo, Brazil
| | - Mirella Alves Cunha
- Departamento de Infectologia, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Igor Thiago Queiroz
- Universidade Potiguar (UnP), Laureate International Universities, Natal, Brazil.,Hospital Giselda Trigueiro (SESAP/RN), Natal, Brazil
| |
Collapse
|
11
|
Rebello KM, Andrade-Neto VV, Zuma AA, Motta MCM, Gomes CRB, de Souza MVN, Atella GC, Branquinha MH, Santos ALS, Torres-Santos EC, d'Avila-Levy CM. Lopinavir, an HIV-1 peptidase inhibitor, induces alteration on the lipid metabolism of Leishmania amazonensis promastigotes. Parasitology 2018; 145:1304-1310. [PMID: 29806577 PMCID: PMC6137378 DOI: 10.1017/s0031182018000823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Abstract
The anti-leishmania effects of HIV peptidase inhibitors (PIs) have been widely reported; however, the biochemical target and mode of action are still a matter of controversy in Leishmania parasites. Considering the possibility that HIV-PIs induce lipid accumulation in Leishmania amazonensis, we analysed the effects of lopinavir on the lipid metabolism of L. amazonensis promastigotes. To this end, parasites were treated with lopinavir at different concentrations and analysed by fluorescence microscopy and spectrofluorimetry, using a fluorescent lipophilic marker. Then, the cellular ultrastructure of treated and control parasites was analysed by transmission electron microscopy (TEM), and the lipid composition was investigated by thin-layer chromatography (TLC). Finally, the sterol content was assayed by gas chromatography-mass spectrometry (GC/MS). TEM analysis revealed an increased number of lipid inclusions in lopinavir-treated cells, which was accompanied by an increase in the lipophilic content, in a dose-dependent manner. TLC and GC-MS analysis revealed a marked increase of cholesterol-esters and cholesterol. In conclusion, lopinavir-induced lipid accumulation and affected lipid composition in L. amazonensis in a concentration-response manner. These data contribute to a better understanding of the possible mechanisms of action of this HIV-PI in L. amazonensis promastigotes. The concerted action of lopinavir on this and other cellular processes, such as the direct inhibition of an aspartyl peptidase, may be responsible for the arrested development of the parasite.
Collapse
Affiliation(s)
- Karina M Rebello
- Laboratório de Estudos Integrados em Protozoologia,Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ),Rio de Janeiro,Brazil
| | - Valter V Andrade-Neto
- Laboratório de Bioquímica de Tripanosomatídeos,Instituto Oswaldo Cruz, FIOCRUZ,Rio de Janeiro,Brazil
| | - Aline A Zuma
- Laboratório de Ultraestrutura Celular Hertha Meyer,Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ),Rio de Janeiro,Brazil
| | - Maria Cristina M Motta
- Laboratório de Ultraestrutura Celular Hertha Meyer,Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ),Rio de Janeiro,Brazil
| | | | | | | | - Marta H Branquinha
- Laboratório de Investigação de Peptidases,Instituto de Microbiologia Paulo de Góes, UFRJ,Rio de Janeiro,Brazil
| | - André L S Santos
- Laboratório de Investigação de Peptidases,Instituto de Microbiologia Paulo de Góes, UFRJ,Rio de Janeiro,Brazil
| | | | - Claudia M d'Avila-Levy
- Laboratório de Estudos Integrados em Protozoologia,Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ),Rio de Janeiro,Brazil
| |
Collapse
|
12
|
Adriaensen W, Dorlo TPC, Vanham G, Kestens L, Kaye PM, van Griensven J. Immunomodulatory Therapy of Visceral Leishmaniasis in Human Immunodeficiency Virus-Coinfected Patients. Front Immunol 2018; 8:1943. [PMID: 29375567 PMCID: PMC5770372 DOI: 10.3389/fimmu.2017.01943] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/18/2017] [Indexed: 12/23/2022] Open
Abstract
Patients with visceral leishmaniasis (VL)–human immunodeficiency virus (HIV) coinfection experience increased drug toxicity and treatment failure rates compared to VL patients, with more frequent VL relapse and death. In the era of VL elimination strategies, HIV coinfection is progressively becoming a key challenge, because HIV-coinfected patients respond poorly to conventional VL treatment and play an important role in parasite transmission. With limited chemotherapeutic options and a paucity of novel anti-parasitic drugs, new interventions that target host immunity may offer an effective alternative. In this review, we first summarize current views on how VL immunopathology is significantly affected by HIV coinfection. We then review current clinical and promising preclinical immunomodulatory interventions in the field of VL and discuss how these may operate in the context of a concurrent HIV infection. Caveats are formulated as these interventions may unpredictably impact the delicate balance between boosting of beneficial VL-specific responses and deleterious immune activation/hyperinflammation, activation of latent provirus or increased HIV-susceptibility of target cells. Evidence is lacking to prioritize a target molecule and a more detailed account of the immunological status induced by the coinfection as well as surrogate markers of cure and protection are still required. We do, however, argue that virologically suppressed VL patients with a recovered immune system, in whom effective antiretroviral therapy alone is not able to restore protective immunity, can be considered a relevant target group for an immunomodulatory intervention. Finally, we provide perspectives on the translation of novel theories on synergistic immune cell cross-talk into an effective treatment strategy for VL–HIV-coinfected patients.
Collapse
Affiliation(s)
- Wim Adriaensen
- Unit of HIV and Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Thomas P C Dorlo
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek Hospital, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Guido Vanham
- Unit of Virology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Luc Kestens
- Unit of Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Paul M Kaye
- Centre for Immunology and Infection, Department of Biology, Hull York Medical School, University of York, Heslington, York, United Kingdom
| | - Johan van Griensven
- Unit of HIV and Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
13
|
Understanding serine proteases implications on Leishmania spp lifecycle. Exp Parasitol 2018; 184:67-81. [DOI: 10.1016/j.exppara.2017.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/14/2017] [Accepted: 11/20/2017] [Indexed: 12/14/2022]
|
14
|
Evans EE, Siedner MJ. Tropical Parasitic Infections in Individuals Infected with HIV. CURRENT TROPICAL MEDICINE REPORTS 2017; 4:268-280. [PMID: 33842194 PMCID: PMC8034600 DOI: 10.1007/s40475-017-0130-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Neglected tropical diseases share both geographic and socio-behavioral epidemiological risk factors with HIV infection. In this literature review, we describe interactions between parasitic diseases and HIV infection, with a focus on the impact of parasitic infections on HIV infection risk and disease progression, and the impact of HIV infection on clinical characteristics of tropical parasitic infections. We limit our review to tropical parasitic infections of the greatest public health burden, and exclude discussion of classic HIV-associated opportunistic infections that have been well reviewed elsewhere. RECENT FINDINGS Tropical parasitic infections, HIV-infection, and treatment with antiretroviral therapy alter host immunity, which can impact susceptibility, transmissibility, diagnosis, and severity of both HIV and parasitic infections. These relationships have a broad range of consequences, from putatively increasing susceptibility to HIV acquisition, as in the case of schistosomiasis, to decreasing risk of protozoal infections through pharmacokinetic interactions between antiretroviral therapy and antiparasitic agents, as in the case of malaria. However, despite this intimate interplay in pathophysiology and a broad overlap in epidemiology, there is a general paucity of data on the interactions between HIV and tropical parasitic infections, particularly in the era of widespread antiretroviral therapy availability. SUMMARY Additional data are needed to motivate clinical recommendations for detection and management of parasitic infections in HIV-infected individuals, and to consider the implications of and potential opportunity granted by HIV treatment programs on parasitic disease control.
Collapse
Affiliation(s)
| | - Mark J Siedner
- Massachusetts General Hospital
- Harvard Medical School
- Mbarara University of Science and Technology
| |
Collapse
|
15
|
Abstract
New drugs and treatments for diseases caused by intracellular pathogens, such as leishmaniasis and the Leishmania species, have proved to be some of the most difficult to discover and develop. The focus of discovery research has been on the identification of potent and selective compounds that inhibit target enzymes (or other essential molecules) or are active against the causative pathogen in phenotypic in vitro assays. Although these discovery paradigms remain an essential part of the early stages of the drug R & D pathway, over the past two decades additional emphasis has been given to the challenges needed to ensure that the potential anti-infective drugs distribute to infected tissues, reach the target pathogen within the host cell and exert the appropriate pharmacodynamic effect at these sites. This review will focus on how these challenges are being met in relation to Leishmania and the leishmaniases with lessons learned from drug R & D for other intracellular pathogens.
Collapse
|
16
|
Abstract
Cutaneous and visceral leishmaniasis are amongst the most devastating infectious diseases of our time, affecting millions of people worldwide. The treatment of these serious diseases rely on a few chemotherapeutic agents, most of which are of parenteral use and induce severe side-effects. Furthermore, rates of treatment failure are high and have been linked to drug resistance in some areas. Here, we reviewed data on current chemotherapy practice in leishmaniasis. Drug resistance and mechanisms of resistance are described as well as the prospects for applying drug combinations for leishmaniasis chemotherapy. It is clear that efforts for discovering new drugs applicable to leishmaniasis chemotherapy are essential. The main aspects on the various steps of drug discovery in the field are discussed.
Collapse
|
17
|
Aronson N, Herwaldt BL, Libman M, Pearson R, Lopez-Velez R, Weina P, Carvalho EM, Ephros M, Jeronimo S, Magill A. Diagnosis and Treatment of Leishmaniasis: Clinical Practice Guidelines by the Infectious Diseases Society of America (IDSA) and the American Society of Tropical Medicine and Hygiene (ASTMH). Clin Infect Dis 2016; 63:e202-e264. [PMID: 27941151 DOI: 10.1093/cid/ciw670] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 09/22/2016] [Indexed: 12/25/2022] Open
Abstract
It is important to realize that leishmaniasis guidelines cannot always account for individual variation among patients. They are not intended to supplant physician judgment with respect to particular patients or special clinical situations. The IDSA and ASTMH consider adherence to these guidelines to be voluntary, with the ultimate determinations regarding their application to be made by the physician in the light of each patient's individual circumstances.
Collapse
Affiliation(s)
- Naomi Aronson
- Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Michael Libman
- McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | - Peter Weina
- Walter Reed National Military Medical Center, Bethesda, Maryland
| | | | | | - Selma Jeronimo
- Federal University of Rio Grande do Norte, Natal, Brazil
| | - Alan Magill
- Bill and Melinda Gates Foundation, Seattle, Washington
| |
Collapse
|
18
|
Lindoso JAL, Cunha MA, Queiroz IT, Moreira CHV. Leishmaniasis-HIV coinfection: current challenges. HIV AIDS-RESEARCH AND PALLIATIVE CARE 2016; 8:147-156. [PMID: 27785103 PMCID: PMC5063600 DOI: 10.2147/hiv.s93789] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Leishmaniasis – human immunodeficiency virus (HIV) coinfection can manifest itself as tegumentary or visceral leishmaniasis. Almost 35 countries have reported autochthonous coinfections. Visceral leishmaniasis is more frequently described. However, usual and unusual manifestations of tegumentary leishmaniasis have been reported mainly in the Americas, but the real prevalence of Leishmania infection in HIV-infected patients is not clear. Regarding the clinical manifestations, there are some reports showing unusual manifestations in visceral leishmaniasis and tegumentary leishmaniasis in HIV-infected patients; yet, the usual manifestations are more frequent. Leishmaniasis diagnosis relies on clinical methods, but serological tests are used to diagnose visceral leishmaniasis despite them having a low sensitivity to tegumentary leishmaniasis. The search for the parasite is used to diagnose both visceral leishmaniasis and tegumentary leishmaniasis. Nevertheless, in HIV-infected patients, the sensitivity of serology is very low. Drugs available to treat leishmaniasis are more restricted and cause severe side effects. Furthermore, in HIV-infected patients, these side effects are more prominent and relapses and lethality are more recurrent. In this article, we discuss the current challenges of tegumentary leishmaniasis and visceral leishmaniasis–HIV infection, focusing mainly on the clinical manifestations, diagnosis, and treatment of leishmaniasis.
Collapse
Affiliation(s)
- José Angelo Lauletta Lindoso
- Laboratory of Soroepidemiology (LIM HC-FMUSP), São Paulo University, São Paulo; Instituto de Infectologia Emilio Ribas-SES, São Paulo
| | - Mirella Alves Cunha
- Department of Infectious Disease, Faculty of Medicine, São Paulo University, São Paulo
| | | | | |
Collapse
|
19
|
Relapse of visceral leishmaniasis in an HIV-infected patient successfully treated with a combination of miltefosine and amphotericin B. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2016; 26:325-9. [PMID: 26744591 PMCID: PMC4692303 DOI: 10.1155/2015/176545] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The present report documents a 49-year-old HIV-infected man receiving antiretroviral therapy with a suboptimal immune response and a CD4 count of 95 cells/mm(3), despite virological suppression. Investigation of bone marrow was conducted and yielded a diagnosis of visceral leishmaniasis. The clinical course was complicated by gastrointestinal involvment and relapse occurred after amphotericin B therapy. With the addition of miltefosine, the patient no longer presented with bone marrow amastigotes, and displayed an increased CD4 count and negative Leishmania polymerase chain reaction results. The present case highlights atypical presentation of visceral leishmaniasis, including poor immune reconstitution and gastrointestinal involvement. The high likelihood of relapse and response to combination therapy are illustrated.
Collapse
|
20
|
Singh J, Srivastava A, Jha P, Sinha KK, Kundu B. l-Asparaginase as a new molecular target against leishmaniasis: insights into the mechanism of action and structure-based inhibitor design. MOLECULAR BIOSYSTEMS 2015; 11:1887-96. [DOI: 10.1039/c5mb00251f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
l-Asparaginases belong to a family of amidohydrolases that catalyze the conversion of l-asparagine into l-aspartic acid and ammonia.
Collapse
Affiliation(s)
- Jasdeep Singh
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi-110016
- India
| | - Ankit Srivastava
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi-110016
- India
| | - Pravin Jha
- National Institute of Pharmaceutical Education & Research
- Vaishali-844102
- India
| | - Kislay K. Sinha
- National Institute of Pharmaceutical Education & Research
- Vaishali-844102
- India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences
- Indian Institute of Technology Delhi
- New Delhi-110016
- India
| |
Collapse
|
21
|
Huang Y, Zhang Q, Liu G, Zhao R. A continuous-flow mass biosensor for the real-time dynamic analysis of protease inhibition. Chem Commun (Camb) 2015; 51:6601-4. [DOI: 10.1039/c5cc00885a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A flow injection analysis–quartz crystal microbalance (FIA–QCM) biosensor system was introduced for probing the dynamic interactions during protease inhibition.
Collapse
Affiliation(s)
- Yanyan Huang
- Beijing National Laboratory for Molecular Sciences
- CAS Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Qundan Zhang
- Beijing National Laboratory for Molecular Sciences
- CAS Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Guoquan Liu
- Beijing National Laboratory for Molecular Sciences
- CAS Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences
- CAS Key Laboratory of Analytical Chemistry for Living Biosystems
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| |
Collapse
|
22
|
Vasconcelos MFD, Cunha-Júnior EFD, Andrade-Neto VVD, Siqueira LM, d'Avila-Levy CM, Moreth M, Cunico W, Souza MVND, Gomes CRB, Torres-Santos EC. Oral effectiveness of PMIC4, a novel hydroxyethylpiperazine analogue, in Leishmania amazonensis. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2014; 4:210-3. [PMID: 25516829 PMCID: PMC4266774 DOI: 10.1016/j.ijpddr.2014.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/10/2014] [Accepted: 10/16/2014] [Indexed: 12/21/2022]
Abstract
Pentavalent antimonials have saved the lives of thousands of Leishmania-infected patients more than seventy years but, unfortunately, they are highly toxic and require parenteral delivery. Therefore, the search for safer and orally delivered alternative is a need. This paper describes the antileishmanial properties of PMIC4, a novel hydroxyethylpiperazine analogue. PMIC4 showed potent activity against intracellular amastigotes of Leishmania amazonensis, with IC50 of 1.8 μM and selectivity index higher than 100-fold, calculated in relation to the toxicity on the host cell. Following laboratory animal welfare policies, we analyzed the absorption, distribution, metabolism, excretion and toxicity (ADMET) properties and calculated the Lipinski's rule of five of PMIC4 before proceeding to in vivo tests. PMIC4 satisfied Lipinski's rule of five and presented high probability of human intestinal absorption, suggesting a good chance of druglikeness and oral bioavailability. For in vivo studies, PMIC4 was administered via intralesional injection (3.4 mg/kg/day, three times a week) or orally (34.0 mg/kg/day, five times a week) to L. amazonensis-infected BALB/c mice throughout the 98 day experiments. At the end of the treatment period, serum markers of toxicity were measured. When administered orally, PMIC4 controlled the lesions in L. amazonensis-infected BALB/c mice without altering serological markers of toxicity. These results demonstrate that PMIC4 is a promising molecular scaffold, orally effective against experimental leishmaniasis.
Collapse
Affiliation(s)
| | | | | | - Larissa Moreira Siqueira
- Laboratório de Bioquímica de Tripanosomatídeos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Cláudia Masini d'Avila-Levy
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Marcele Moreth
- Departamento de Síntese de Fármacos, Farmanguinhos, FIOCRUZ, Rio de Janeiro, Brazil
| | - Wilson Cunico
- Laboratório de Química Aplicada à Bioativos, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | | | | | | |
Collapse
|
23
|
Lindoso JA, Cota GF, da Cruz AM, Goto H, Maia-Elkhoury ANS, Romero GAS, de Sousa-Gomes ML, Santos-Oliveira JR, Rabello A. Visceral leishmaniasis and HIV coinfection in Latin America. PLoS Negl Trop Dis 2014; 8:e3136. [PMID: 25233461 PMCID: PMC4169383 DOI: 10.1371/journal.pntd.0003136] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Visceral leishmaniasis (VL) is an endemic zoonotic disease in Latin America caused by Leishmania (Leishmania) infantum, which is transmitted by sand flies from the genus Lutzomyia. VL occurs in 12 countries of Latin America, with 96% of cases reported in Brazil. Recently, an increase in VL, primarily affecting children and young adults, has been observed in urban areas of Latin America. The area in which this spread of VL is occurring overlaps regions with individuals living with HIV, the number of whom is estimated to be 1.4 million people by the World Health Organization. This overlap is suggested to be a leading cause of the increased number of reported VL-HIV coinfections. The clinical progression of HIV and L. infantum infections are both highly dependent on the specific immune response of an individual. Furthermore, the impact on the immune system caused by either pathogen and by VL-HIV coinfection can contribute to an accelerated progression of the diseases. Clinical presentation of VL in HIV positive patients is similar to patients without HIV, with symptoms characterized by fever, splenomegaly, and hepatomegaly, but diarrhea appears to be more common in coinfected patients. In addition, VL relapses are higher in coinfected patients, affecting 10% to 56.5% of cases and with a lethality ranging from 8.7% to 23.5% in Latin America, depending on the study. With regards to the diagnosis of VL, parasitological tests of bone marrow aspirates have proven to be the most sensitive test in HIV-infected patients. Serologic tests have demonstrated a variable sensitivity according to the method and antigens used, with the standard tests used for diagnosing VL in Latin America displaying lower sensitivity. For this review, few articles were identified that related to VL-HIV coinfections and originated from Latin America, highlighting the need for improving research within the regions most greatly affected. We strongly support the formation of a Latin American network for coinfections of Leishmania and HIV to improve the consistency of research on the current situation of VL-HIV coinfections. Such a network would improve the collection of vital data and samples for better understanding of the clinical manifestations and immunopathogenic aspects of VL in immunosuppressed patients. Ultimately, a concerted effort would improve trials for new diagnostic methodologies and therapeutics, which could accelerate the implementation of more specific and effective diagnosis as well as public policies for treatments to reduce the impact of VL-HIV coinfections on the Latin American population.
Collapse
Affiliation(s)
- José Angelo Lindoso
- Instituto de Infectologia Emilio Ribas, São Paulo, São Paulo, Brasil
- Laboratório de Soroepidemiologia (LIM-38) Hospital das Clínicas da Faculdade de Mediciina da Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- * E-mail: (JAL); (AR)
| | - Gláucia Fernandes Cota
- Hospital Eduardo de Menezes, Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
- Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil
| | - Alda Maria da Cruz
- Laboratório Interdisciplinar de Pesquisas Medicas, Instituto Oswaldo Cruz–FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
- Disciplina de Parasitologia/FCM-UERJ, Manguinhos, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hiro Goto
- Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Departamento de Medicina Preventiva da Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Gustavo Adolfo Sierra Romero
- Núcleo de Medicina Tropical, Universidade de Brasilia, Distrito Federal, Brazil
- Instituto Nacional de Ciência e Tecnologia de Avaliação de Tecnologia em Saúde, Porto Alegre, Rio Grande do Sul, Brazil
- Fundação de Amparo à Pesquisa do Estado do Amazonas (FAPEAM), Manaus, Amazonas, Brazil
| | | | - Joanna Reis Santos-Oliveira
- Laboratório Interdisciplinar de Pesquisas Medicas, Instituto Oswaldo Cruz–FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Rabello
- Centro de Pesquisa René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil
- * E-mail: (JAL); (AR)
| |
Collapse
|
24
|
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:e3021. [PMID: 25144380 PMCID: PMC4140663 DOI: 10.1371/journal.pntd.0003021] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Visceral leishmaniasis is hypoendemic in Mediterranean countries, where it is caused by the flagellate protozoan Leishmania infantum. VL cases in this area account for 5%–6% of the global burden. Cases of Leishmania/HIV coinfection have been reported in the Mediterranean region, mainly in France, Italy, Portugal, and Spain. Since highly active antiretroviral therapy was introduced in 1997, a marked decrease in the number of coinfected cases in this region has been reported. The development of new diagnostic methods to accurately identify level of parasitemia and the risk of relapse is one of the main challenges in improving the treatment of coinfected patients. Clinical trials in the Mediterranean region are needed to determine the most adequate therapeutic options for Leishmania/HIV patients as well as the indications and regimes for secondary prophylaxis. This article reviews the epidemiological, diagnostic, clinical, and therapeutic aspects of Leishmania/HIV coinfection in the Mediterranean region.
Collapse
Affiliation(s)
- Begoña Monge-Maillo
- Tropical Medicine & Clinical Parasitology, Infectious Diseases Department, Ramón y Cajal Hospital, Madrid, Spain
| | - Francesca F. Norman
- Tropical Medicine & Clinical Parasitology, Infectious Diseases Department, Ramón y Cajal Hospital, Madrid, Spain
| | - Israel Cruz
- WHO Collaborating Centre for Leishmaniasis, Servicio de Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Jorge Alvar
- Visceral Leishmaniasis Program, Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Rogelio López-Vélez
- Tropical Medicine & Clinical Parasitology, Infectious Diseases Department, Ramón y Cajal Hospital, Madrid, Spain
- * E-mail:
| |
Collapse
|
25
|
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:e2869. [PMID: 24968313 PMCID: PMC4072530 DOI: 10.1371/journal.pntd.0002869] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Visceral Leishmaniasis (VL) is an important protozoan opportunistic disease in HIV patients in endemic areas. East Africa is second to the Indian subcontinent in the global VL caseload and first in VL-HIV coinfection rate. Because of the alteration in the disease course, the diagnostic challenges, and the poor treatment responses, VL with HIV coinfection has become a very serious challenge in East Africa today. Field experience with the use of liposomal amphotericin B in combination with miltefosine, followed by secondary prophylaxis and antiretroviral drugs, looks promising. However, this needs to be confirmed through clinical trials. Better diagnostic and follow-up methods for relapse and prediction of relapse should also be looked for. Basic research to understand the immunological interaction of the two infections may ultimately help to improve the management of the coinfection.
Collapse
Affiliation(s)
- Ermias Diro
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Internal Medicine, University of Gondar, Gondar, Ethiopia
- * E-mail:
| | - Lutgarde Lynen
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Koert Ritmeijer
- Public Health Department, Médecins Sans Frontières, Amsterdam, the Netherlands
| | - Marleen Boelaert
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Asrat Hailu
- School of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Johan van Griensven
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
26
|
Mohapatra S. Drug resistance in leishmaniasis: Newer developments. Trop Parasitol 2014; 4:4-9. [PMID: 24754020 PMCID: PMC3992802 DOI: 10.4103/2229-5070.129142] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 03/20/2014] [Indexed: 11/06/2022] Open
Abstract
Leishmaniasis is a vector borne protozoan disease and it remains a major public health problem world-wide. Lack of an effective vaccine and vector control program makes the chemotherapy as the primary tool for leishmaniasis. Antimonials were used as the first line of treatment for many years. Emergence of resistance against this drug has become a major concern. Literatures and studies published on anti-leishmanial drug resistance, newer drug discovery for leishmanial resistance etc., in PubMed, Medline and Google search and reviewed thoroughly. Various newer drugs have been identified but, are in limited use because of high cost, toxicity, resistance etc., Recently, many newer mechanisms of drug resistance have been identified which may boost in future designing and development of drugs.
Collapse
Affiliation(s)
- Sarita Mohapatra
- Department of Microbiology, Vardhaman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
27
|
van Griensven J. Editorial Commentary: Visceral Leishmaniasis and HIV Coinfection in Bihar, India: A Wake-up Call? Clin Infect Dis 2014; 59:556-8. [DOI: 10.1093/cid/ciu334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
28
|
van Griensven J, Carrillo E, López-Vélez R, Lynen L, Moreno J. Leishmaniasis in immunosuppressed individuals. Clin Microbiol Infect 2014; 20:286-99. [DOI: 10.1111/1469-0691.12556] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Leishmaniasis acquired by travellers to endemic regions in Europe: A EuroTravNet multi-centre study. Travel Med Infect Dis 2014; 12:167-72. [DOI: 10.1016/j.tmaid.2013.12.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/02/2013] [Accepted: 12/03/2013] [Indexed: 01/09/2023]
|