1
|
Awogbindin I, ŠimonČiČová E, Vidal V, Ash C, Tremblay ME. Neuroglial responses to bacterial, viral, and fungal neuroinfections. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:213-238. [PMID: 40148046 DOI: 10.1016/b978-0-443-19102-2.00027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Evidence regarding the host's response to peripheral pathogens in humans abound, whereas studies on the pathogenesis of central nervous system-penetrating infections are relatively scarce. However, given the spate of epidemic and pandemic neuroinfections in the 21st century, the field has experienced a renewed interest lately. This chapter discusses a timely and exciting topic on the roles of glial cells, mainly microglia and astrocytes, in neuroinvasive infections. This chapter considered fungal, viral, and bacterial neuroinfections, X-raying their neuroinvasiveness, neurotropism, and neurovirulence before focusing on specific examples notable for each category, including Escherichia coli, Cryptococcus neoformans, and SARS-CoV-2. These infections are renowned worldwide for a high case-fatality rate, leaving many survivors with life-long morbidity and others with a bleak future neurologic prognosis. Importantly, the chapter discusses possible ways microglia and astrocytes are culpable in these infections and provides approaches by which they can be manipulated for therapeutic purposes, identifying viable research gaps in the process. Additionally, it offers a synopsis of ongoing works considering microglial selective targeting to attenuate the pathology, morbidity, and mortality associated with these neuroinfections. Considering that microglia and astrocytes are first responders in the central nervous system, targeting these glial cells could be the game changer in managing existing and emerging neuroinvasive infections.
Collapse
Affiliation(s)
- Ifeoluwa Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada
| | - Eva ŠimonČiČová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Virginie Vidal
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Science and Technology Department, University of Bordeaux, Bordeaux, France
| | - Chantaille Ash
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
2
|
Shabalala SN, Luvuno M, Mabandla MV. Modulation of tenofovir by probenecid: Impact on drug, interleukin-1β, and dopamine concentration in the prefrontal cortex and cerebellum. Neuroscience 2024; 562:209-216. [PMID: 39461661 DOI: 10.1016/j.neuroscience.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The blood-brain barrier's limited permeability to tenofovir restricts its ability to clear HIV from the brain. Probenecid acting as an adjuvant increases tenofovir concentrations in plasma and the kidneys thereby enhancing its therapeutic effect. However, the probenecid effect on brain tenofovir concentration and possible adverse effects remains poorly understood. We investigated the effect of probenecid co-administered tenofovir on tenofovir brain concentration, interleukin-1β (IL-1β) and dopamine concentration in the prefrontal cortex (PFC) and the cerebellum. Ninety-six male BALB/c mice were divided into four groups viz: a control group, Tenofovir disoproxil fumarate (TDF) treated, probenecid treated, and TDF + probenecid treated. We orally administered a single dose of TDF (5 mg/kg), and probenecid (8.3 mg/kg), and sacrificed six mice per group after 1 h, 4 h, and 6 h post-treatment to collect plasma, PFC, and cerebellar tissue. Co-administered tenofovir increased tenofovir concentration, peaking at 6 h with the cerebellum having the highest concentration. This suggests that probenecid enhanced the entry of tenofovir into the brain. Tenofovir alone increased IL-1β concentration at all intervals post-administration, while probenecid alone had no impact on IL-1β concentration. Co-administered tenofovir also increased IL-1β concentration. Probenecid's limited impact on IL-1β concentration following co-administration suggests that its anti-inflammatory properties may require more than 6 h to have an effect. Furthermore, neither tenofovir nor probenecid affected dopamine concentration. In conclusion, probenecid enhances the concentration and retention of tenofovir in the brain, making it a possible pharmacokinetic enhancer. However, its anti-inflammatory effects may require a longer duration to fully manifest.
Collapse
Affiliation(s)
- Simangele Ne Shabalala
- Discipline of Human Physiology, School of Laboratory Medicine and MedicalSciences, College of Health Sciences, South Africa.
| | - M Luvuno
- Discipline of Human Physiology, School of Laboratory Medicine and MedicalSciences, College of Health Sciences, South Africa
| | - M V Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine and MedicalSciences, College of Health Sciences, South Africa
| |
Collapse
|
3
|
Kim JB, Kim SJ, So M, Kim DK, Noh HR, Kim BJ, Choi YR, Kim D, Koo H, Kim T, Woo HG, Park SM. Artificial intelligence-driven drug repositioning uncovers efavirenz as a modulator of α-synuclein propagation: Implications in Parkinson's disease. Biomed Pharmacother 2024; 174:116442. [PMID: 38513596 DOI: 10.1016/j.biopha.2024.116442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder with an unclear etiology. Despite significant research efforts, developing disease-modifying treatments for PD remains a major unmet medical need. Notably, drug repositioning is becoming an increasingly attractive direction in drug discovery, and computational approaches offer a relatively quick and resource-saving method for identifying testable hypotheses that promote drug repositioning. We used an artificial intelligence (AI)-based drug repositioning strategy to screen an extensive compound library and identify potential therapeutic agents for PD. Our AI-driven analysis revealed that efavirenz and nevirapine, approved for treating human immunodeficiency virus infection, had distinct profiles, suggesting their potential effects on PD pathophysiology. Among these, efavirenz attenuated α-synuclein (α-syn) propagation and associated neuroinflammation in the brain of preformed α-syn fibrils-injected A53T α-syn Tg mice and α-syn propagation and associated behavioral changes in the C. elegans BiFC model. Through in-depth molecular investigations, we found that efavirenz can modulate cholesterol metabolism and mitigate α-syn propagation, a key pathological feature implicated in PD progression by regulating CYP46A1. This study opens new avenues for further investigation into the mechanisms underlying PD pathology and the exploration of additional drug candidates using advanced computational methodologies.
Collapse
Affiliation(s)
- Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Soo-Jeong Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | | | - Dong-Kyu Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | - Hye Rin Noh
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Beom Jin Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea
| | - Doyoon Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Department of Physiology, Ajou University School of Medicine, Suwon, Korea
| | | | | | - Hyun Goo Woo
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Department of Physiology, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
4
|
Sun M, Manson ML, Guo T, de Lange ECM. CNS Viral Infections-What to Consider for Improving Drug Treatment: A Plea for Using Mathematical Modeling Approaches. CNS Drugs 2024; 38:349-373. [PMID: 38580795 PMCID: PMC11026214 DOI: 10.1007/s40263-024-01082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Neurotropic viruses may cause meningitis, myelitis, encephalitis, or meningoencephalitis. These inflammatory conditions of the central nervous system (CNS) may have serious and devastating consequences if not treated adequately. In this review, we first summarize how neurotropic viruses can enter the CNS by (1) crossing the blood-brain barrier or blood-cerebrospinal fluid barrier; (2) invading the nose via the olfactory route; or (3) invading the peripheral nervous system. Neurotropic viruses may then enter the intracellular space of brain cells via endocytosis and/or membrane fusion. Antiviral drugs are currently used for different viral CNS infections, even though their use and dosing regimens within the CNS, with the exception of acyclovir, are minimally supported by clinical evidence. We therefore provide considerations to optimize drug treatment(s) for these neurotropic viruses. Antiviral drugs should cross the blood-brain barrier/blood cerebrospinal fluid barrier and pass the brain cellular membrane to inhibit these viruses inside the brain cells. Some antiviral drugs may also require intracellular conversion into their active metabolite(s). This illustrates the need to better understand these mechanisms because these processes dictate drug exposure within the CNS that ultimately determine the success of antiviral drugs for CNS infections. Finally, we discuss mathematical model-based approaches for optimizing antiviral treatments. Thereby emphasizing the potential of CNS physiologically based pharmacokinetic models because direct measurement of brain intracellular exposure in living humans faces ethical restrictions. Existing physiologically based pharmacokinetic models combined with in vitro pharmacokinetic/pharmacodynamic information can be used to predict drug exposure and evaluate efficacy of antiviral drugs within the CNS, to ultimately optimize the treatments of CNS viral infections.
Collapse
Affiliation(s)
- Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tingjie Guo
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
5
|
Kaur H, Minchella P, Alvarez-Carbonell D, Purandare N, Nagampalli VK, Blankenberg D, Hulgan T, Gerschenson M, Karn J, Aras S, Kallianpur AR. Contemporary Antiretroviral Therapy Dysregulates Iron Transport and Augments Mitochondrial Dysfunction in HIV-Infected Human Microglia and Neural-Lineage Cells. Int J Mol Sci 2023; 24:12242. [PMID: 37569616 PMCID: PMC10419149 DOI: 10.3390/ijms241512242] [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: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
HIV-associated cognitive dysfunction during combination antiretroviral therapy (cART) involves mitochondrial dysfunction, but the impact of contemporary cART on chronic metabolic changes in the brain and in latent HIV infection is unclear. We interrogated mitochondrial function in a human microglia (hμglia) cell line harboring inducible HIV provirus and in SH-SY5Y cells after exposure to individual antiretroviral drugs or cART, using the MitoStress assay. cART-induced changes in protein expression, reactive oxygen species (ROS) production, mitochondrial DNA copy number, and cellular iron were also explored. Finally, we evaluated the ability of ROS scavengers or plasmid-mediated overexpression of the antioxidant iron-binding protein, Fth1, to reverse mitochondrial defects. Contemporary antiretroviral drugs, particularly bictegravir, depressed multiple facets of mitochondrial function by 20-30%, with the most pronounced effects in latently infected HIV+ hμglia and SH-SY5Y cells. Latently HIV-infected hμglia exhibited upregulated glycolysis. Increases in total and/or mitochondrial ROS, mitochondrial DNA copy number, and cellular iron accompanied mitochondrial defects in hμglia and SH-SY5Y cells. In SH-SY5Y cells, cART reduced mitochondrial iron-sulfur-cluster-containing supercomplex and subunit expression and increased Nox2 expression. Fth1 overexpression or pre-treatment with N-acetylcysteine prevented cART-induced mitochondrial dysfunction. Contemporary cART impairs mitochondrial bioenergetics in hμglia and SH-SY5Y cells, partly through cellular iron accumulation; some effects differ by HIV latency.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Paige Minchella
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48202, USA
| | - David Alvarez-Carbonell
- Department of Microbiology and Molecular Biology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Neeraja Purandare
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48202, USA
| | - Vijay K. Nagampalli
- Department of Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniel Blankenberg
- Department of Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Todd Hulgan
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96844, USA
| | - Jonathan Karn
- Department of Microbiology and Molecular Biology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Siddhesh Aras
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48202, USA
| | - Asha R. Kallianpur
- Department of Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Al Nasr IS, Koko WS, Khan TA, Gürbüz N, Özdemir I, Hamdi N. Evaluation of Ruthenium(II) N-Heterocyclic Carbene Complexes as Enzymatic Inhibitory Agents with Antioxidant, Antimicrobial, Antiparasitical and Antiproliferative Activity. Molecules 2023; 28:molecules28031359. [PMID: 36771026 PMCID: PMC9921063 DOI: 10.3390/molecules28031359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
A series of [RuCl2(p-cymene)(NHC)] complexes were obtained by reacting [RuCl2(p-cymene)]2 with in situ generated Ag-N-heterocyclic carbene (NHC) complexes. The structure of the obtained complexes was determined by the appropriate spectroscopy and elemental analysis. In addition, we evaluated the biological activities of these compounds as antienzymatic, antioxidant, antibacterial, anticancer, and antiparasitic agents. The results revealed that complexes 3b and 3d were the most potent inhibitors against AchE with IC50 values of 2.52 and 5.06 μM mL-1. Additionally, 3d proved very good antimicrobial activity against all examined microorganisms with IZ (inhibition zone) over 25 mm and MIC (minimum inhibitory concentration) < 4 µM. Additionally, the ligand 2a and its corresponding ruthenium (II) complex 3a had good cytotoxic activity against both cancer cells HCT-116 and HepG-2, with IC50 values of (7.76 and 11.76) and (4.12 and 9.21) μM mL-1, respectively. Evaluation of the antiparasitic activity of these complexes against Leishmania major promastigotes and Toxoplasma gondii showed that ruthenium complexes were more potent than the free ligand, with an IC50 values less than 1.5 μM mL-1. However, 3d was found the best one with SI (selectivity index) values greater than 5 so it seems to be the best candidate for antileishmanial drug discovery program, and much future research are recommended for mode of action and in vivo evaluation. In general, Ru-NHC complexes are the most effective against L. major promastigotes.
Collapse
Affiliation(s)
- Ibrahim S. Al Nasr
- Department of Biology, College of Science and Arts, Qassim University, Unaizah 51911, Saudi Arabia
- Department of Science Laboratories, College of Science and Arts, Qassim University, Ar Rass 51921, Saudi Arabia
| | - Waleed S. Koko
- Department of Science Laboratories, College of Science and Arts, Qassim University, Ar Rass 51921, Saudi Arabia
| | - Tariq A. Khan
- Department of Clinical Nutrition, College of Applied Health Sciences, Qassim University, Ar Rass 51921, Saudi Arabia
| | - Nevin Gürbüz
- Department of Chemistry, Faculty of Science and Art, İnönü University, Malatya 44280, Turkey
- Catalysis Research and Application Center, İnönü University, Malatya 44280, Turkey
| | - Ismail Özdemir
- Department of Chemistry, Faculty of Science and Art, İnönü University, Malatya 44280, Turkey
- Catalysis Research and Application Center, İnönü University, Malatya 44280, Turkey
| | - Naceur Hamdi
- Department of Chemistry, College of Science and Arts at ArRass, Qassim University, Ar Rass 51921, Saudi Arabia
- Correspondence: ; Tel.: +966-556394839
| |
Collapse
|
7
|
Hamdi N, Mansour L, Al-Tamimi J, Al-Hazmy SM, Gurbuz N, Özdemir I. Synthesis and Investigation of Antimicrobial, Antioxidant, Enzymatic Inhibitory, and Antiproliferative Activities of Ruthenium (II) Complexes Bearing Benzimidazole-Based N-Heterocyclic Carbene (NHC) Ligands. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2150659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Naceur Hamdi
- Department of Chemistry, College of Science and Arts at ArRass, Qassim University, ArRass, Saudi Arabia
| | - Lamjed Mansour
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Jameel Al-Tamimi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sadeq M. Al-Hazmy
- Chemistry Department, College of Science, Qassim University, Buraidah, Saudi Arabia
| | - Nevin Gurbuz
- Faculty of Science and Art, Department of Chemistry, İnönü University, Malatya, Turkey
- İnönü University, Catalysis Research and Application Center, Malatya, Turkey
| | - Ismail Özdemir
- Faculty of Science and Art, Department of Chemistry, İnönü University, Malatya, Turkey
- İnönü University, Catalysis Research and Application Center, Malatya, Turkey
| |
Collapse
|
8
|
Boubakri L, Chakchouk-Mtiba A, Naouali O, Mellouli L, Mansour L, Özdemir I, Yaser S, Sauthier M, Hamdi N. Ruthenium(II) complexes bearing benzimidazole-based N-heterocyclic carbene (NHC) ligands as potential antimicrobial, antioxidant, enzyme inhibition, and antiproliferative agents. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2060745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Lamia Boubakri
- Research Laboratory of Environmental Sciences and Technologies (LR16ES09), Higher Institute of Environmental Sciences and Technology, University of Carthage, Hammam-Lif, Tunisia
| | - Ahlem Chakchouk-Mtiba
- Laboratory of Microorganisms and Enzymatic Biotechnology and Biomolecules, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Olfa Naouali
- Laboratory of Applied Chemistry and Natural Substances Resources and Environment, Faculty of Sciences, University of Carthage, Zarzouna, Bizerta, Tunisia
| | - Lotfi Mellouli
- Laboratory of Microorganisms and Enzymatic Biotechnology and Biomolecules, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Lamjed Mansour
- Zoology Department, College of Science, King Saud University, Saudi Arabia, Riyadh, Saudi Arabia
| | - Ismail Özdemir
- Faculty of Science and Art, Department of Chemistry, İnönü University, Malatya, Turkey
- Catalysis Research and Application Center, İnönü University, Malatya, Turkey
| | - Sedat Yaser
- Faculty of Science and Art, Department of Chemistry, İnönü University, Malatya, Turkey
- Catalysis Research and Application Center, İnönü University, Malatya, Turkey
| | - Mathieu Sauthier
- Ecole Nationale Superieure de Chimie de Lille, Unité de Catalyse et Chimie du Solide, Villeneuve d’Ascq, France
| | - Naceur Hamdi
- Research Laboratory of Environmental Sciences and Technologies (LR16ES09), Higher Institute of Environmental Sciences and Technology, University of Carthage, Hammam-Lif, Tunisia
| |
Collapse
|
9
|
Gilmore JC, Zhang G, Cameron DW, Serghides L, Bendayan R. Impact of in-utero antiretroviral drug exposure on expression of membrane-associated transporters in mouse placenta and fetal brain. AIDS 2021; 35:2249-2258. [PMID: 34175869 DOI: 10.1097/qad.0000000000003009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Although antiretroviral therapy (ART) during pregnancy is effective in limiting vertical HIV transmission, adverse outcomes persist amongst uninfected children exposed to antiretroviral drugs in utero. Membrane-associated drug transporters, metabolic enzymes, and tight junction proteins play important roles in adult antiretroviral drug disposition and toxicity; however, the fetal expression of these proteins in the context of ART, and their impact on in-utero antiretroviral drug distribution remain poorly understood. This study aimed to characterize the role of these proteins in modulating in-utero antiretroviral drug exposure. METHODS Pregnant mice were exposed to an ART regimen consisting of lamivudine, abacavir, atazanavir, and ritonavir, at clinically relevant doses. Fetal brain, liver, placenta amniotic fluid, and maternal plasma were collected on gestational day 18.5 and concentration of antiretroviral drugs in fetal tissues was measured by LC/MS/MS, whereas transporter expression was assessed by qPCR. RESULTS Abacavir and lamivudine were detected in fetal brain and amniotic fluid, whereas atazanavir and ritonavir were detected in amniotic fluid only. Robust mRNA expression of key transporters was observed in adult and fetal tissues, and sex differences were identified in the expression of Abcc1 and Slc29a1 in the placenta. Antiretroviral drug exposure was associated with a reduction in relative placental Abcg2, Abcc1, and Slc29a1 expression. CONCLUSION These findings identify a novel effect of fetal sex and antiretroviral drug treatment on the expression of placental transporters in a mouse model, and characterize the penetration of lamivudine and abacavir into fetal brain, uncovering a potential role of transporters in modulating fetal exposure to antiretroviral drugs.
Collapse
Affiliation(s)
- Julian C Gilmore
- Department of Pharmaceutical Sciences, University of Toronto, Toronto
| | - Guijun Zhang
- Clinical Investigation Unit, University of Ottawa at the Ottawa Hospital/Research Institute, Ottawa
| | - D William Cameron
- Clinical Investigation Unit, University of Ottawa at the Ottawa Hospital/Research Institute, Ottawa
| | - Lena Serghides
- Department of Immunology and Institute of Medical Sciences, University of Toronto
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, University of Toronto, Toronto
| |
Collapse
|
10
|
CNS Considerations in ART Simplification Strategies. Curr HIV/AIDS Rep 2021; 18:549-557. [PMID: 34739699 DOI: 10.1007/s11904-021-00580-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE OF THE REVIEW This review summarizes current knowledge on central nervous system (CNS) considerations in ART simplification strategies. RECENT FINDINGS Antiretroviral therapies (ART) showing efficacy in plasma will usually show efficacy in the cerebrospinal fluid (CSF). ART simplification may virologically fail if the new regimen has less than two active drugs, the genetic barrier of drugs is not high, and the patient may harbour archived resistance. Dual therapies including a boosted protease inhibitor (PI) or dolutegravir (DTG) are generally effective from the CNS perspective. In cases of related neurotoxicity, switching from either efavirenz (EFV) or DTG to another equally effective drug with better CNS tolerability usually leads to complete resolution of CNS symptoms. However, improvement may be incomplete when factors other than ART that cannot be easily modified are involved.
Collapse
|
11
|
Mhambi S, Fisher D, Tchokonte MBT, Dube A. Permeation Challenges of Drugs for Treatment of Neurological Tuberculosis and HIV and the Application of Magneto-Electric Nanoparticle Drug Delivery Systems. Pharmaceutics 2021; 13:1479. [PMID: 34575555 PMCID: PMC8466684 DOI: 10.3390/pharmaceutics13091479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 12/27/2022] Open
Abstract
The anatomical structure of the brain at the blood-brain barrier (BBB) creates a limitation for the movement of drugs into the central nervous system (CNS). Drug delivery facilitated by magneto-electric nanoparticles (MENs) is a relatively new non-invasive approach for the delivery of drugs into the CNS. These nanoparticles (NPs) can create localized transient changes in the permeability of the cells of the BBB by inducing electroporation. MENs can be applied to deliver antiretrovirals and antibiotics towards the treatment of human immunodeficiency virus (HIV) and tuberculosis (TB) infections in the CNS. This review focuses on the drug permeation challenges and reviews the application of MENs for drug delivery for these diseases. We conclude that MENs are promising systems for effective CNS drug delivery and treatment for these diseases, however, further pre-clinical and clinical studies are required to achieve translation of this approach to the clinic.
Collapse
Affiliation(s)
- Sinaye Mhambi
- Discipline of Pharmaceutics, School of Pharmacy, University of the Western Cape, Cape Town 7535, South Africa;
| | - David Fisher
- Department of Medical Bioscience, University of the Western Cape, Cape Town 7535, South Africa;
| | | | - Admire Dube
- Discipline of Pharmaceutics, School of Pharmacy, University of the Western Cape, Cape Town 7535, South Africa;
| |
Collapse
|
12
|
BADE AN, GENDELMAN HE, MCMILLAN J, LIU Y. Chemical exchange saturation transfer for detection of antiretroviral drugs in brain tissue. AIDS 2021; 35:1733-1741. [PMID: 34049358 PMCID: PMC8373768 DOI: 10.1097/qad.0000000000002960] [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] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Antiretroviral drug theranostics facilitates the monitoring of biodistribution and efficacy of therapies designed to target HIV type-1 (HIV-1) reservoirs. To this end, we have now deployed intrinsic drug chemical exchange saturation transfer (CEST) contrasts to detect antiretroviral drugs within the central nervous system (CNS). DESIGN AND METHODS CEST effects for lamivudine (3TC) and emtricitabine (FTC) were measured by asymmetric magnetization transfer ratio analyses. The biodistribution of 3TC in different brain sub-regions of C57BL/6 mice treated with lipopolysaccharides was determined using MRI. CEST effects of 3TC protons were quantitated by Lorentzian fitting analysis. 3TC levels in plasma and brain regions were measured using ultraperformance liquid chromatography tandem mass spectrometry to affirm the CEST test results. RESULTS CEST effects of the hydroxyl and amino protons in 3TC and FTC linearly correlated to drug concentrations. 3TC was successfully detected in vivo in brain sub-regions by MRI. The imaging results were validated by measurements of CNS drug concentrations. CONCLUSION CEST contrasts can be used to detect antiretroviral drugs using MRI. Such detection can be used to assess spatial--temporal drug biodistribution. This is most notable within the CNS where drug biodistribution may be more limited with the final goal of better understanding antiretroviral drug-associated efficacy and potential toxicity.
Collapse
Affiliation(s)
- Aditya N. BADE
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Howard E. GENDELMAN
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - JoEllyn MCMILLAN
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yutong LIU
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
13
|
Zhao X, Wilson K, Uteshev V, He JJ. Activation of α7 nicotinic acetylcholine receptor ameliorates HIV-associated neurology and neuropathology. Brain 2021; 144:3355-3370. [PMID: 34196664 PMCID: PMC8677536 DOI: 10.1093/brain/awab251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/28/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
HIV-associated neurocognitive disorders (HAND) in the era of combination antiretroviral therapy are primarily manifested as impaired behaviours, glial activation/neuroinflammation and compromised neuronal integrity, for which there are no effective treatments currently available. In the current study, we used doxycycline-inducible astrocyte-specific HIV Tat transgenic mice (iTat), a surrogate HAND model, and determined effects of PNU-125096, a positive allosteric modulator of α7 nicotinic acetylcholine receptor (α7 nAChR) on Tat-induced behavioural impairments and neuropathologies. We showed that PNU-125096 treatment significantly improved locomotor, learning and memory deficits of iTat mice while inhibited glial activation and increased PSD-95 expression in the cortex and hippocampus of iTat mice. Using α7 nAChR knockout mice, we showed that α7 nAChR knockout eliminated the protective effects of PNU-125096 on iTat mice. In addition, we showed that inhibition of p38 phosphorylation by SB239063, a p38 MAPK-specific inhibitor exacerbated Tat neurotoxicity in iTat mice. Last, we used primary mouse cortical individual cultures and neuron-astrocytes co-cultures and in vivo staining of iTat mouse brain tissues and showed that glial activation was directly involved in the interplay among Tat neurotoxicity, α7 nAChR activation and the p38 MAPK signalling pathway. Taken together, these findings demonstrated for the first time that α7 nAChR activation led to protection against HAND and suggested that α7 nAChR modulator PNU-125096 holds significant promise for development of therapeutics for HAND.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Kelly Wilson
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Victor Uteshev
- Department of Pharmacology and Neuroscience, Graduate School of Biomedical Sciences of University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Johnny J He
- Department of Microbiology and Immunology, Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA.,Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University, North Chicago, IL 60064, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL 60064, USA
| |
Collapse
|
14
|
Letendre SL, Mills A, Hagins D, Swindells S, Felizarta F, Devente J, Bettacchi C, Lou Y, Ford S, Sutton K, Shaik JS, Crauwels H, D'Amico R, Patel P. Pharmacokinetics and antiviral activity of cabotegravir and rilpivirine in cerebrospinal fluid following long-acting injectable administration in HIV-infected adults. J Antimicrob Chemother 2021; 75:648-655. [PMID: 31873746 PMCID: PMC7021098 DOI: 10.1093/jac/dkz504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/18/2022] Open
Abstract
Background Long-acting (LA) formulations of cabotegravir, an HIV integrase inhibitor, and rilpivirine, an NNRTI, are in development as monthly or 2 monthly intramuscular (IM) injections for maintenance of virological suppression. Objectives To evaluate cabotegravir and rilpivirine CSF distribution and HIV-1 RNA suppression in plasma and CSF in HIV-infected adults participating in a substudy of the Phase 2b LATTE-2 study (NCT02120352). Methods Eighteen participants receiving cabotegravir LA 400 mg + rilpivirine LA 600 mg IM [every 4 weeks (Q4W), n = 3] or cabotegravir LA 600 mg + rilpivirine LA 900 mg IM [every 8 weeks (Q8W), n = 15] with plasma HIV-1 RNA <50 copies/mL enrolled. Paired steady-state CSF and plasma concentrations were evaluable in 16 participants obtained 7 (±3) days after an injection visit. HIV-1 RNA in CSF and plasma were assessed contemporaneously using commercial assays. Results Median total CSF concentrations in Q4W and Q8W groups, respectively, were 0.011 μg/mL and 0.013 μg/mL for cabotegravir (0.30% and 0.34% of the paired plasma concentrations) and 1.84 ng/mL and 1.67 ng/mL for rilpivirine (1.07% and 1.32% of paired plasma concentrations). Cabotegravir and rilpivirine total CSF concentrations exceeded their respective in vitro EC50 for WT HIV-1 (0.10 ng/mL and 0.27 ng/mL, respectively). All 16 participants had HIV-1 RNA <50 copies/mL in plasma and CSF, and 15 of 16 participants had HIV-1 RNA <2 copies/mL in CSF. Conclusions A dual regimen of cabotegravir LA and rilpivirine LA achieved therapeutic concentrations in the CSF resulting in effective virological control in CSF.
Collapse
Affiliation(s)
| | | | | | | | | | - Jerome Devente
- Long Beach Education and Research Consultants, Long Beach, CA, USA
| | | | - Yu Lou
- PAREXEL International, Durham, NC, USA
| | - Susan Ford
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | | | | | | | | | - Parul Patel
- ViiV Healthcare, Research Triangle Park, NC, USA
| |
Collapse
|
15
|
George JW, Mattingly JE, Roland NJ, Small CM, Lamberty BG, Fox HS, Stauch KL. Physiologically Relevant Concentrations of Dolutegravir, Emtricitabine, and Efavirenz Induce Distinct Metabolic Alterations in HeLa Epithelial and BV2 Microglial Cells. Front Immunol 2021; 12:639378. [PMID: 34093527 PMCID: PMC8173175 DOI: 10.3389/fimmu.2021.639378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Microglia, the resident brain phagocytes, likely play a key role in human immunodeficiency virus (HIV) infection of the central nervous system (CNS) and subsequent neuropathogenesis; however, the nature of the infection-induced changes that yield damaging CNS effects and the stimuli that provoke microglial activation remains elusive, especially in the current era of using antiretroviral (ARV) drugs for ARV therapy (ART). Altered microglial metabolism can modulate cellular functionality and pathogenicity in neurological disease. While HIV infection itself alters brain energy metabolism, the effect of ARV drugs, particularly those currently used in treatment, on metabolism is understudied. Dolutegravir (DTG) and emtricitabine (FTC) combination, together with tenofovir (TAF or TDF), is one of the recommended first line treatments for HIV. Despite the relatively good tolerability and safety profile of FTC, a nucleoside reverse transcriptase inhibitor, and DTG, an integrase inhibitor, adverse side effects have been reported and highlight a need to understand off-target effects of these medications. We hypothesized that similar to previous ART regimen drugs, DTG and FTC side effects involve mitochondrial dysfunction. To increase detection of ARV-induced mitochondrial effects, highly glycolytic HeLa epithelial cells were forced to rely on oxidative phosphorylation by substituting galactose for glucose in the growth media. We assessed ATP levels, resazurin oxidation-reduction (REDOX), and mitochondrial membrane potential following 24-hour exposure (to approximate effects of one dose equivalent) to DTG, FTC, and efavirenz (EFV, a known mitotoxic ARV drug). Further, since microglia support productive HIV infection, act as latent HIV cellular reservoirs, and when dysfunctional likely contribute to HIV-associated neurocognitive disorders, the experiments were repeated using BV2 microglial cells. In HeLa cells, FTC decreased mitochondrial REDOX activity, while DTG, similar to EFV, impaired both mitochondrial ATP generation and REDOX activity. In contrast to HeLa cells, DTG increased cellular ATP generation and mitochondrial REDOX activity in BV2 cells. Bioenergetic analysis revealed that DTG, FTC, and EFV elevated BV2 cell mitochondrial respiration. DTG and FTC exposure induced distinct mitochondrial functional changes in HeLa and BV2 cells. These findings suggest cell type-specific metabolic changes may contribute to the toxic side effects of these ARV drugs.
Collapse
Affiliation(s)
- Joseph W George
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jane E Mattingly
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Nashanthea J Roland
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Cassandra M Small
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Benjamin G Lamberty
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kelly L Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
16
|
Cheney L, Barbaro JM, Berman JW. Antiretroviral Drugs Impact Autophagy with Toxic Outcomes. Cells 2021; 10:909. [PMID: 33920955 PMCID: PMC8071244 DOI: 10.3390/cells10040909] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 01/18/2023] Open
Abstract
Antiretroviral drugs have dramatically improved the morbidity and mortality of people living with HIV (PLWH). While current antiretroviral therapy (ART) regimens are generally well-tolerated, risks for side effects and toxicity remain as PLWH must take life-long medications. Antiretroviral drugs impact autophagy, an intracellular proteolytic process that eliminates debris and foreign material, provides nutrients for metabolism, and performs quality control to maintain cell homeostasis. Toxicity and adverse events associated with antiretrovirals may be due, in part, to their impacts on autophagy. A more complete understanding of the effects on autophagy is essential for developing antiretroviral drugs with decreased off target effects, meaning those unrelated to viral suppression, to minimize toxicity for PLWH. This review summarizes the findings and highlights the gaps in our knowledge of the impacts of antiretroviral drugs on autophagy.
Collapse
Affiliation(s)
- Laura Cheney
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - John M. Barbaro
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA; (J.M.B.); (J.W.B.)
| | - Joan W. Berman
- Department of Pathology, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA; (J.M.B.); (J.W.B.)
- Department of Microbiology and Immunology, Montefiore Medical Center and Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| |
Collapse
|
17
|
Kelentse N, Moyo S, Mogwele M, Lechiile K, Moraka NO, Maruapula D, Seatla KK, Esele L, Molebatsi K, Leeme TB, Lawrence DS, Musonda R, Kasvosve I, Harrison TS, Jarvis JN, Gaseitsiwe S. Differences in human immunodeficiency virus-1C viral load and drug resistance mutation between plasma and cerebrospinal fluid in patients with human immunodeficiency virus-associated cryptococcal meningitis in Botswana. Medicine (Baltimore) 2020; 99:e22606. [PMID: 33031315 PMCID: PMC7544309 DOI: 10.1097/md.0000000000022606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/14/2020] [Accepted: 09/07/2020] [Indexed: 11/26/2022] Open
Abstract
To determine effects of cryptococcal meningitis (CM) on human immunodeficiency virus (HIV)-1C cerebrospinal fluid (CSF) viral escape, CSF/plasma viral discordance, and drug resistance mutation (DRM) discordance between CSF and plasma compartments, we compared CSF and plasma viral load (VL) and DRMs in individuals with HIV-associated CM in Botswana.This cross-sectional study utilized 45 paired CSF/plasma samples from participants in a CM treatment trial (2014-2016). HIV-1 VL was determined and HIV-1 protease and reverse transcriptase genotyping performed. DRMs were determined using the Stanford HIV database. CSF viral escape was defined as HIV-1 ribonucleic acid ≥0.5 log10 higher in CSF than plasma and VL discordance as CSF VL > plasma VL.HIV-1 VL was successfully measured in 39/45 pairs, with insufficient sample volume in 6; 34/39 (87.2%) participants had detectable HIV-1 in plasma and CSF, median 5.1 (interquartile range: 4.7-5.7) and 4.6 (interquartile range:3.7-4.9) log10 copies/mL, respectively (P≤.001). CSF viral escape was present in 1/34 (2.9%) and VL discordance in 6/34 (17.6%). Discordance was not associated with CD4 count, antiretroviral status, fungal burden, CSF lymphocyte percentage nor mental status. Twenty-six of 45 (57.8%) CSF/plasma pairs were successfully sequenced. HIV-1 DRM discordance was found in 3/26 (11.5%); 1 had I84IT and another had M46MI in CSF only. The third had K101E in plasma and V106 M in CSF.Our findings suggest that HIV-1 escape and DRM discordance may occur at lower rates in participants with advanced HIV-disease and CM compared to those with HIV associated neurocognitive impairment.
Collapse
Affiliation(s)
- Nametso Kelentse
- Botswana Harvard AIDS Institute Partnership
- University of Botswana, Department of Medical Laboratory Sciences, Gaborone, Botswana
| | - Sikhulile Moyo
- Botswana Harvard AIDS Institute Partnership
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, United States
| | - Mompati Mogwele
- Botswana Harvard AIDS Institute Partnership
- University of Botswana, Department of Biological Sciences, Gaborone, Botswana
| | | | - Natasha O. Moraka
- Botswana Harvard AIDS Institute Partnership
- Stellenbosch University, Department of Pathology, Stellenbosch, South Africa
| | - Dorcas Maruapula
- Botswana Harvard AIDS Institute Partnership
- University of Botswana, Department of Biological Sciences, Gaborone, Botswana
| | - Kaelo K. Seatla
- Botswana Harvard AIDS Institute Partnership
- University of Botswana, Department of Medical Laboratory Sciences, Gaborone, Botswana
| | | | - Kesaobaka Molebatsi
- Botswana Harvard AIDS Institute Partnership
- University of Botswana, Department of Statistics, Gaborone, Botswana
| | - Tshepo B. Leeme
- Botswana Harvard AIDS Institute Partnership
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - David S. Lawrence
- Botswana Harvard AIDS Institute Partnership
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, The London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Rosemary Musonda
- Botswana Harvard AIDS Institute Partnership
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, United States
| | - Ishmael Kasvosve
- University of Botswana, Department of Medical Laboratory Sciences, Gaborone, Botswana
| | - Thomas S. Harrison
- Centre for Global Health, Institute for Infection and Immunity, St. George's University of London, United Kingdom
| | - Joseph N. Jarvis
- Botswana Harvard AIDS Institute Partnership
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, The London School of Hygiene and Tropical Medicine, London, United Kingdom
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simani Gaseitsiwe
- Botswana Harvard AIDS Institute Partnership
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, United States
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW The application of immunotherapies to HIV presents a new horizon of treatment options, but little is known about what impact they may have on the central nervous system (CNS). Here we review the most promising immunotherapeutic strategies that can be used to target HIV in the CNS and focus on identifying their potential benefits while exploring the challenges that remain in their application. RECENT FINDINGS We have identified five immunotherapeutic strategies that hold potential in managing CNS disease among HIV-infected patients. These include broadly neutralizing antibodies, multi-affinity antibodies, CAR-T cell therapy, checkpoint inhibitors, and therapeutic vaccines. Each class of immunotherapy presents unique mechanisms by which CNS viremia and latency may be addressed but are faced with several challenges. CAR-T cell therapy and multi-affinity antibodies seem to hold promise, but combination therapy is likely to be most effective. However, more human trials are needed before the clinical benefits of these therapies are realized.
Collapse
Affiliation(s)
- Andrew Kapoor
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - C Sabrina Tan
- Division of Infectious Diseases, Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue E/CLS 1011, Boston, MA, 02215, USA.
| |
Collapse
|
19
|
Weichseldorfer M, Affram Y, Heredia A, Tagaya Y, Benedetti F, Zella D, Reitz M, Romerio F, Latinovic OS. Anti-HIV Activity of Standard Combined Antiretroviral Therapy in Primary Cells Is Intensified by CCR5-Targeting Drugs. AIDS Res Hum Retroviruses 2020; 36:835-841. [PMID: 32623916 DOI: 10.1089/aid.2020.0064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of combined antiretroviral therapy (cART) against HIV-1 is evidenced by reduction of plasma viremia, disease progression, viral transmission, and mortality. However, major challenges still remain in HIV-1 management, especially the emergence of resistant strains and the persistence of viral reservoirs, apparent after cART treatment interruption. Efforts are ongoing to explore the most effective means to intensify cART and successfully control residual viral replication. We anticipate that the reduction by cART of HIV-1 reservoirs could be further enhanced by combining cART with entry inhibitors and drugs that silence CCR5 expression. CCR5-targeting drugs are attractive option because of their low side effects when combined with other antiretroviral drugs. The concept that their inclusion would be effective has been supported by the reduction in two long terminal repeat unintegrated circular DNA, a marker for new infections, when CCR5-targeting drugs are added to standard antiretroviral treatment. This study is, in part, an extension of our previous study demonstrating greater preservation of human CD4+ T-cells and CD4+/CD8+ cell ratios in HIV-infected CD34+ NSG mice when CCR5-targeting drugs were included with standard cART. In this study, we treated HIV-1-infected cell cultures with cART or cART plus CCR5-targeting drugs (maraviroc and rapamycin). We found that treatment intensification with CCR5-targeting drugs led to a significant reduction of HIV-1 replication in peripheral blood ononuclear cells (PBMCs), as judged by measured viral DNA copies and p24 levels. Our data provide proof of principle for the benefit of adding CCR5-targeting drugs to traditional, standard cART to further lower viremia and subsequently reduce viral reservoirs in clinical settings, while potentially lowering side effects by reducing cART concentrations.
Collapse
Affiliation(s)
- Matthew Weichseldorfer
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Yvonne Affram
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Alonso Heredia
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Yutaka Tagaya
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Francesca Benedetti
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Davide Zella
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Marvin Reitz
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Fabio Romerio
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Olga S. Latinovic
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
20
|
Joseph SB, Kincer LP, Bowman NM, Evans C, Vinikoor MJ, Lippincott CK, Gisslén M, Spudich S, Menezes P, Robertson K, Archin N, Kashuba A, Eron JJ, Price RW, Swanstrom R. Human Immunodeficiency Virus Type 1 RNA Detected in the Central Nervous System (CNS) After Years of Suppressive Antiretroviral Therapy Can Originate from a Replicating CNS Reservoir or Clonally Expanded Cells. Clin Infect Dis 2020; 69:1345-1352. [PMID: 30561541 DOI: 10.1093/cid/ciy1066] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/12/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) populations are detected in cerebrospinal fluid (CSF) of some people on suppressive antiretroviral therapy (ART). Detailed analysis of these populations may reveal whether they are produced by central nervous system (CNS) reservoirs. METHODS We performed a study of 101 asymptomatic participants on stable ART. HIV-1 RNA concentrations were cross-sectionally measured in CSF and plasma. In participants with CSF HIV-1 RNA concentrations sufficient for analysis, viral populations were genetically and phenotypically characterized over multiple time points. RESULTS For 6% of participants (6 of 101), the concentration of HIV-1 RNA in their CSF was ≥0.5 log copies/mL above that of plasma (ie, CSF escape). We generated viral envelope sequences from CSF of 3 participants. One had a persistent CSF escape population that was macrophage-tropic, partially drug resistant, genetically diverse, and closely related to a minor macrophage-tropic lineage present in the blood prior to viral suppression and enriched for after ART. Two participants (1 suppressed and 1 not) had transient CSF escape populations that were R5 T cell-tropic with little genetic diversity. CONCLUSIONS Extensive analysis of viral populations in 1 participant revealed that CSF escape was from a persistently replicating population, likely in macrophages/microglia, present in the CNS over 3 years of ART. CSF escape in 2 other participants was likely produced by trafficking and transient expansion of infected T cells in the CNS. Our results show that CNS reservoirs can persist during ART and that CSF escape is not exclusively produced by replicating CNS reservoirs.
Collapse
Affiliation(s)
- Sarah B Joseph
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Maryland
| | - Laura P Kincer
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Maryland
| | - Natalie M Bowman
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Maryland
| | - Chris Evans
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Maryland
| | - Michael J Vinikoor
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Maryland
| | - Christopher K Lippincott
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Magnus Gisslén
- Department of Infectious Diseases, Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Serena Spudich
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
| | - Prema Menezes
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Maryland.,University of North Carolina Center for AIDS Research, University of North Carolina at Chapel Hill, San Francisco
| | - Kevin Robertson
- Department of Neurology, University of North Carolina at Chapel Hill, San Francisco
| | - Nancie Archin
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Maryland
| | - Angela Kashuba
- University of North Carolina Center for AIDS Research, University of North Carolina at Chapel Hill, San Francisco.,Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, San Francisco
| | - Joseph J Eron
- Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Maryland.,University of North Carolina Center for AIDS Research, University of North Carolina at Chapel Hill, San Francisco
| | - Richard W Price
- Department of Neurology, University of California, San Francisco
| | - Ronald Swanstrom
- University of North Carolina Center for AIDS Research, University of North Carolina at Chapel Hill, San Francisco.,Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill
| |
Collapse
|
21
|
The Paradox of HIV Blood-Brain Barrier Penetrance and Antiretroviral Drug Delivery Deficiencies. Trends Neurosci 2020; 43:695-708. [PMID: 32682564 PMCID: PMC7483662 DOI: 10.1016/j.tins.2020.06.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/04/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022]
Abstract
HIV attacks the body's immune cells, frequently compromises the integrity of the blood-brain barrier (BBB), and infects the CNS in the early stages of infection. Dysfunction of the BBB further potentiates viral replication within the CNS, which can lead to HIV-associated neuropathology. Antiretroviral therapy (ART) significantly improves HIV patient outcomes and reduces mortality rates. However, there has been limited progress in targeting latent viral reservoirs within the CNS, which may eventually lead to rebound viremia. While ART drugs are shown to be effective in attenuating HIV replication in the periphery, the protection of the brain by the BBB offers an isolated sanctuary to harbor HIV and maintains chronic and persistent replication within the CNS. In this review, we elucidate the pathology of the BBB, its ability to potentiate viral replication, as well as current therapies and insufficiencies in treating HIV-infected individuals.
Collapse
|
22
|
HIV Nef and Antiretroviral Therapy Have an Inhibitory Effect on Autophagy in Human Astrocytes that May Contribute to HIV-Associated Neurocognitive Disorders. Cells 2020; 9:cells9061426. [PMID: 32526847 PMCID: PMC7349791 DOI: 10.3390/cells9061426] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022] Open
Abstract
A significant number of people living with HIV (PLWH) develop HIV-associated neurocognitive disorders (HAND) despite highly effective antiretroviral therapy (ART). Dysregulated macroautophagy (autophagy) is implicated in HAND pathogenesis. The viral protein Nef, expressed even with suppressive ART, and certain antiretrovirals affect autophagy in non-CNS cells. Astrocytes, vital for CNS microenvironment homeostasis and neuronal health, require autophagy for their own homeostasis. We hypothesized that extracellular Nef and/or ART impact astrocyte autophagy, thus contributing to HAND. We studied in-bulk and selective autophagic flux in primary human astrocytes treated with extracellular Nef and/or a combination of tenofovir+emtricitabine+raltegravir (ART) using Western blotting, a tandem fluorescent LC3 reporter, and transmission electron microscopy/morphometry. We show that after 24 h treatment, Nef and ART decrease autophagosomes through different mechanisms. While Nef accelerates autophagosome degradation without inducing autophagosome formation, ART inhibits autophagosome formation. Combination Nef+ART further depletes autophagosomes by inducing both abnormalities. Additionally, extracellular Nef and/or ART inhibit lysosomal degradation of p62, indicating Nef and/or ART affect in-bulk and selective autophagy differently. Dysregulation of both autophagic processes is maintained after 7 days of Nef and/or ART treatment. Persistent autophagy dysregulation due to chronic Nef and/or ART exposure may ultimately result in astrocyte and neuronal dysfunction, contributing to HAND.
Collapse
|
23
|
An Elvitegravir Nanoformulation Crosses the Blood-Brain Barrier and Suppresses HIV-1 Replication in Microglia. Viruses 2020; 12:v12050564. [PMID: 32443728 PMCID: PMC7290679 DOI: 10.3390/v12050564] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Even with an efficient combination of antiretroviral therapy (ART), which significantly decreases viral load in human immunodeficiency virus type 1 (HIV-1)-positive individuals, the occurrence of HIV-1-associated neurocognitive disorders (HAND) still exists. Microglia have been shown to have a significant role in HIV-1 replication in the brain and in subsequent HAND pathogenesis. However, due to the limited ability of ART drugs to cross the blood-brain barrier (BBB) after systemic administration, in addition to efflux transporter expression on microglia, the efficacy of ART drugs for viral suppression in microglia is suboptimal. Previously, we developed novel poly (lactic-co-glycolic acid) (PLGA)-based elvitegravir nanoparticles (PLGA-EVG NPs), which showed improved BBB penetration in vitro and improved viral suppression in HIV-1-infected primary macrophages, after crossing an in vitro BBB model. Our objective in the current study was to evaluate the efficacy of our PLGA-EVG NPs in an important central nervous system (CNS) HIV-1 reservoir, i.e., microglia. In this study, we evaluated the cyto-compatibility of the PLGA-EVG NPs in microglia, using an XTT (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) assay and cellular morphology observation. We also studied the endocytosis pathway and the subcellular localization of PLGA NPs in microglia, using various endocytosis inhibitors and subcellular localization markers. We determined the ability of PLGA-EVG NPs to suppress HIV-1 replication in microglia, after crossing an in vitro BBB model. We also studied the drug levels in mouse plasma and brain tissue, using immunodeficient NOD scid gamma (NSG) mice, and performed a pilot study, to evaluate the efficacy of PLGA-EVG NPs on viral suppression in the CNS, using an HIV-1 encephalitic (HIVE) mouse model. From our results, the PLGA-EVG NPs showed ~100% biocompatibility with microglia, as compared to control cells. The internalization of PLGA NPs in microglia occurred through caveolae-/clathrin-mediated endocytosis. PLGA NPs can also escape from endo-lysosomal compartments and deliver the therapeutics to cells efficiently. More importantly, the PLGA-EVG NPs were able to show ~25% more viral suppression in HIV-1-infected human-monocyte-derived microglia-like cells after crossing the in vitro BBB compared to the EVG native drug, without altering BBB integrity. PLGA-EVG NPs also showed a ~two-fold higher level in mouse brain and a trend of decreasing CNS HIV-1 viral load in HIV-1-infected mice. Overall, these results help us to create a safe and efficient drug delivery method to target HIV-1 reservoirs in the CNS, for potential clinical use.
Collapse
|
24
|
Fosamprenavir with Ritonavir Pharmacokinetics during Pregnancy. Antimicrob Agents Chemother 2020; 64:AAC.02260-19. [PMID: 32015036 DOI: 10.1128/aac.02260-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022] Open
Abstract
The purpose of this study was to evaluate the pharmacokinetics of ritonavir-boosted fosamprenavir during pregnancy and postpartum. Amprenavir (the active moiety of fosamprenavir) and ritonavir intensive pharmacokinetic evaluations were performed at steady state during the second and third trimesters of pregnancy and postpartum. Plasma concentrations of amprenavir and ritonavir were measured using high-performance liquid chromatography. The target amprenavir area under the concentration-versus-time curve (AUC) was higher than the 10th percentile (27.7 μg · h/ml) of the median area under the curve for ritonavir-boosted fosamprenavir in adults receiving twice-daily fosamprenavir-ritonavir at 700 mg/100 mg. Twenty-nine women were included in the analysis. The amprenavir AUC from time zero to 12 h (AUC0-12) was lower (geometric mean ratio [GMR], 0.60 [confidence interval {CI}, 0.49 to 0.72] [P < 0.001]) while its apparent oral clearance was higher (GMR, 1.68 [CI, 1.38 to 2.03] [P < 0.001]) in the third trimester than postpartum. Similarly, the ritonavir AUC0-12 was lower in the second (GMR, 0.51 [CI, 0.28 to 0.91] [P = 0.09]) and third (GMR, 0.72 [CI, 0.55 to 0.95] [P = 0.005]) trimesters than postpartum, while its apparent oral clearance was higher in the second (GMR, 1.98 [CI, 1.10 to 3.56] [P = 0.06]) and third (GMR, 1.38 [CI, 1.05 to 1.82] [P = 0.009]) trimesters than postpartum. The amprenavir area under the curve exceeded the target for 6/8 (75%) women in the 2nd trimester, 18/28 (64%) in the 3rd trimester, and 19/22 (86.4%) postpartum, and the trough concentrations (C min) of amprenavir were 4- to 16-fold above the mean amprenavir-protein-adjusted 50% inhibitory concentration (IC50) of 0.146 μg/ml. Although amprenavir plasma concentrations in women receiving ritonavir-boosted fosamprenavir were lower during pregnancy than postpartum, the reduced amprenavir concentrations were still above the exposures needed for viral suppression.
Collapse
|
25
|
Gong Y, Chowdhury P, Nagesh PKB, Rahman MA, Zhi K, Yallapu MM, Kumar S. Novel elvitegravir nanoformulation for drug delivery across the blood-brain barrier to achieve HIV-1 suppression in the CNS macrophages. Sci Rep 2020; 10:3835. [PMID: 32123217 PMCID: PMC7052245 DOI: 10.1038/s41598-020-60684-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023] Open
Abstract
The use of antiretroviral therapy (ART) has remarkably decreased the morbidity associated with HIV-1 infection, however, the prevalence of HIV-1-associated neurocognitive disorders (HAND) is still increasing. The blood-brain barrier (BBB) is the major impediment for penetration of antiretroviral drugs, causing therapeutics to reach only suboptimal level to the brain. Conventional antiretroviral drug regimens are not sufficient to improve the treatment outcomes of HAND. In our recent report, we have developed a poloxamer-PLGA nanoformulation loaded with elvitegravir (EVG), a commonly used antiretroviral drug. The nanoformulated EVG is capable of elevating intracellular drug uptake and simultaneously enhance viral suppression in HIV-1-infected macrophages. In this work, we identified the clinical parameters including stability, biocompatibility, protein corona, cellular internalization pathway of EVG nanoformulation for its potential clinical translation. We further assessed the ability of this EVG nanoformulation to cross the in vitro BBB model and suppress the HIV-1 in macrophage cells. Compared with EVG native drug, our EVG nanoformulation demonstrated an improved BBB model penetration cross the in vitro BBB model and an enhanced HIV-1 suppression in HIV-1-infected human monocyte-derived macrophages after crossing the BBB model without altering the BBB model integrity. Overall, this is an innovative and optimized treatment strategy that has a potential for therapeutic interventions in reducing HAND.
Collapse
Affiliation(s)
- Yuqing Gong
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Prashanth K B Nagesh
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Mohammad A Rahman
- National Institute of Environmental Health Sciences, Durham, NC, 27703, USA
| | - Kaining Zhi
- Plough Center for Sterile Drug Delivery Solutions, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Microbiology and Immunology, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
26
|
Saxena SK, Maurya VK, Kumar S, Bhatt MLB. Modern Approaches in Nanomedicine for NeuroAIDS and CNS Drug Delivery. Nanobiomedicine (Rij) 2020. [DOI: 10.1007/978-981-32-9898-9_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
27
|
Sarma A, Das MK. Nose to brain delivery of antiretroviral drugs in the treatment of neuroAIDS. MOLECULAR BIOMEDICINE 2020; 1:15. [PMID: 34765998 PMCID: PMC7725542 DOI: 10.1186/s43556-020-00019-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
NeuroAIDS (Neuro Acquired Immunodeficiency Syndrome) or HIV (Human Immunodeficiency Virus) associated neuronal abnormality is continuing to be a significant health issue among AIDS patients even under the treatment of combined antiretroviral therapy (cART). Injury and damage to neurons of the brain are the prime causes of neuroAIDS, which happens due to the ingress of HIV by direct permeation across the blood-brain barrier (BBB) or else via peripherally infected macrophage into the central nervous system (CNS). The BBB performs as a stringent barricade for the delivery of therapeutics drugs. The intranasal route of drug administration exhibits as a non-invasive technique to bypass the BBB for the delivery of antiretroviral drugs and other active pharmaceutical ingredients inside the brain and CNS. This method is fruitful for the drugs that are unable to invade the BBB to show its action in the CNS and thus erase the demand of systemic delivery and thereby shrink systemic side effects. Drug delivery from the nose to the brain/CNS takes very less time through both olfactory and trigeminal nerves. Intranasal delivery does not require the involvement of any receptor as it occurs by an extracellular route. Nose to brain delivery also involves nasal associated lymphatic tissues (NALT) and deep cervical lymph nodes. However, very little research has been done to explore the utility of nose to brain delivery of antiretroviral drugs in the treatment of neuroAIDS. This review focuses on the potential of nasal route for the effective delivery of antiretroviral nanoformulations directly from nose to the brain.
Collapse
Affiliation(s)
- Anupam Sarma
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India.,Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026 India
| | - Malay K Das
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| |
Collapse
|
28
|
Santerre M, Wang Y, Arjona S, Allen C, Sawaya BE. Differential Contribution of HIV-1 Subtypes B and C to Neurological Disorders: Mechanisms and Possible Treatments. AIDS Rev 2019; 21:76-83. [PMID: 31332398 DOI: 10.24875/aidsrev.19000051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the introduction of combinatory antiretroviral therapy, patients infected with human immunodeficiency virus type 1 (HIV-1) can live much longer than before. However, the identification of HIV-associated neurocognitive disorder (HAND), especially HIV-associated dementia in 15-20% of patients infected with HIV-1, indicates additional complexity. These disorders turn out to be subtype dependent. Recently, many studies are ongoing trying to understand how the virus induces neuronal injury which could lead to neurological dysfunction. Most of these studies are focusing on the HIV-1 release of proteins such as Tat. However, the exact role of these proteins and their involvement in neuronal degeneration remains unidentified; this is especially true since viral proteins from different HIV-1 subtypes differ in their ability to cause neuronal damage. This review describes the role of different HIV-1 subtypes, identifies probable pathways involved in neuronal damage, the contribution of different HIV-1 subtypes to the progression of HAND, and potential treatments for HAND.
Collapse
Affiliation(s)
- Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Institute for Cancer Research and Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Ying Wang
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Institute for Cancer Research and Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Sterling Arjona
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Institute for Cancer Research and Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Charles Allen
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Institute for Cancer Research and Molecular Biology, Philadelphia, Pennsylvania, USA
| | - Bassel E Sawaya
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Imaging correlates of the blood-brain barrier disruption in HIV-associated neurocognitive disorder and therapeutic implications. AIDS 2019; 33:1843-1852. [PMID: 31274535 DOI: 10.1097/qad.0000000000002300] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE HIV-associated neurocognitive disorders (HANDs) in the context of suppressive combination antiretroviral therapy (cART) still occur. We explored the role of blood-brain barrier (BBB) disruption in the pathogenesis of HAND in the context of fully suppressive cART using dynamic contrast enhanced perfusion (DCE-P) MRI. DCE-P is a new MRI technique that measures capillary permeability as an indicator for BBB integrity. We hypothesized that virally suppressed incident HAND would be associated with an impaired BBB as determined by DCE-P. DESIGN A cross sectional study. METHODS K-trans, a metric derivative of DCE-P, was obtained from different regions of the brain in a cohort of 20 patients with HAND who were virally suppressed in both cerebrospinal fluid (CSF) and blood compared with CSF and blood markers of neuroinflammation as well as with neurometabolites derived from magnetic resonance (MR) spectroscopy. RESULTS The K-trans data showed significantly impaired BBB in HAND patients when compared with the controls in the regions of the basal ganglia and anterior frontal white matter (both P < 0.0001). CSF neopterin and CSF/serum albumin ratio correlated positively with K-trans but not with blood levels. CONCLUSION This study indicates that HAND in the context of viral suppression is associated with BBB disruption and the DCE MR derived K-trans metric is a very sensitive parameter to identify the BBB disruption. The finding of region-specific BBB disruption rather than globally and the lack of correlation with blood markers of neuroinflammation suggest that HIV and not systemic inflammation is driving the BBB disturbance and that the BBB disruption is a consequence of HIV already in the brain as opposed to HIV first causing BBB disruption then brain disease.
Collapse
|
30
|
Mdanda S, Ntshangase S, Singh SD, Naicker T, Kruger HG, Baijnath S, Govender T. Mass spectrometric investigations into the brain delivery of abacavir, stavudine and didanosine in a rodent model. Xenobiotica 2019; 50:570-579. [PMID: 31403353 DOI: 10.1080/00498254.2019.1655605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
HIV replication in the brain is unopposed due to reduced antiretroviral drug penetration into the central nervous system (CNS). Prevalence of HIV-associated neurocognitive disorder (HAND) has increased severely in patients living with HIV despite current treatments. The aims of this study were to evaluate the brain bio-distribution of alternative nucleoside reverse transcriptase inhibitors, abacavir, stavudine and didanosine in the CNS and to determine their localization patterns in the brain.Sprague-Dawley rats received 50 mg kg-1 single i.p dose of each drug. Mass spectrometric techniques were then used to investigate the pharmacokinetics and localization patterns of these drugs in the brain using LC-MS/MS and mass spectrometric imaging (MSI), respectively.Abacavir, stavudine and didanosine reached the Brain Cmax with concentration of 831.2, 1300 and 43.37 ngmL-1, respectively. Based on MSI analysis Abacavir and Stavudine were located in brain regions that are strongly implicated in the progression of HAND.Abacavir and Stavudine penetrated into CNS, reaching a Cmax that was above the IC50 for HIV (457.6 and 112.0 ngmL-1, respectively), however, it was noted ddI showed poor entry within the brain, therefore, it is recommended that this drug cannot be considered for treating CNS-HIV.
Collapse
Affiliation(s)
- Sipho Mdanda
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Sphamandla Ntshangase
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | | | - Tricia Naicker
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Sooraj Baijnath
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| | - Thavendran Govender
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa.,Biomedical Resource Unit, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| |
Collapse
|
31
|
Liu Y, Niu Y, Li L, Timani KA, He VL, Sanburns C, Xie J, He JJ. Tat expression led to increased histone 3 tri-methylation at lysine 27 and contributed to HIV latency in astrocytes through regulation of MeCP2 and Ezh2 expression. J Neurovirol 2019; 25:508-519. [PMID: 31020497 PMCID: PMC6750972 DOI: 10.1007/s13365-019-00751-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/03/2019] [Accepted: 04/04/2019] [Indexed: 02/07/2023]
Abstract
Astrocytes are susceptible to HIV infection and potential latent HIV reservoirs. Tat is one of three abundantly expressed HIV early genes in HIV-infected astrocytes and has been shown to be a major pathogenic factor for HIV/neuroAIDS. In this study, we sought to determine if and how Tat expression would affect HIV infection and latency in astrocytes. Using the glycoprotein from vesicular stomatitis virus-pseudotyped red-green HIV (RGH) reporter viruses, we showed that HIV infection was capable of establishing HIV latency in astrocytes. We also found that Tat expression decreased the generation of latent HIV-infected cells. Activation of latent HIV-infected astrocytes showed that treatment of GSK126, a selective inhibitor of methyltransferase enhancer of zeste homolog 2 (Ezh2) that is specifically responsible for tri-methylation of histone 3 lysine 27 (H3K27me3), led to activation of significantly more latent HIV-infected Tat-expressing astrocytes. Molecular analysis showed that H3K27me3, Ezh2, MeCP2, and Tat all exhibited a similar bimodal expression kinetics in the course of HIV infection and latency in astrocytes, although H3K27me3, Ezh2, and MeCP2 were expressed higher in Tat-expressing astrocytes and their expression were peaked immediately preceding Tat expression. Subsequent studies showed that Tat expression alone was sufficient to induce H3K27me3 expression, likely through its regulation of Ezh2 and MeCP2 expression. Taken together, these results showed for the first time that Tat expression induced H3K27me3 expression and contributed to HIV latency in astrocytes and suggest a new role and novel mechanism for Tat in HIV latency.
Collapse
Affiliation(s)
- Ying Liu
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| | - Yinghua Niu
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Lu Li
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Khalid A Timani
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Victor L He
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Chris Sanburns
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Jiafeng Xie
- Department of Microbiology, Immunology and Genetics, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | | |
Collapse
|
32
|
Grande F, Ioele G, Occhiuzzi MA, De Luca M, Mazzotta E, Ragno G, Garofalo A, Muzzalupo R. Reverse Transcriptase Inhibitors Nanosystems Designed for Drug Stability and Controlled Delivery. Pharmaceutics 2019; 11:E197. [PMID: 31035595 PMCID: PMC6572254 DOI: 10.3390/pharmaceutics11050197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/12/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022] Open
Abstract
An in-depth analysis of nanotechnology applications for the improvement of solubility, distribution, bioavailability and stability of reverse transcriptase inhibitors is reported. Current clinically used nucleoside and non-nucleoside agents, included in combination therapies, were examined in the present survey, as drugs belonging to these classes are the major component of highly active antiretroviral treatments. The inclusion of such agents into supramolecular vesicular systems, such as liposomes, niosomes and lipid solid NPs, overcomes several drawbacks related to the action of these drugs, including drug instability and unfavorable pharmacokinetics. Overall results reported in the literature show that the performances of these drugs could be significantly improved by inclusion into nanosystems.
Collapse
Affiliation(s)
- Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Giuseppina Ioele
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Maria Antonietta Occhiuzzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Michele De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Elisabetta Mazzotta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Gaetano Ragno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Antonio Garofalo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| | - Rita Muzzalupo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy.
| |
Collapse
|
33
|
Van den Hof M, Blokhuis C, Cohen S, Scherpbier HJ, Wit FWNM, Pistorius MCM, Kootstra NA, Teunissen CE, Mathot RAA, Pajkrt D. CNS penetration of ART in HIV-infected children. J Antimicrob Chemother 2019; 73:484-489. [PMID: 29126299 DOI: 10.1093/jac/dkx396] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/29/2017] [Indexed: 12/29/2022] Open
Abstract
Background Paediatric data on CNS penetration of antiretroviral drugs are scarce. Objectives To evaluate CNS penetration of antiretroviral drugs in HIV-infected children and explore associations with neurocognitive function. Patients and methods Antiretroviral drug levels were measured in paired CSF and blood samples of clinically stable HIV-infected children between 8 and 18 years old on long-term combined ART. Plasma drug concentrations were corrected for protein binding. We evaluated CNS penetration using CSF/plasma ratios and compared CSF concentrations with the IC50 as a surrogate marker for effectiveness. Blood-brain barrier permeability was assessed for possible confounding. Associations with neurocognitive function were explored using linear regression analysis. Results Median CSF/plasma ratios (IQR) were: lopinavir 0.059 (0.024-0.157, n = 7), efavirenz 0.681 (0.555-0.819, n = 12), tenofovir 0.021 (0.020-0.024, n = 4), lamivudine 0.464 (0.331-0.607, n = 17), emtricitabine 0.365 (0.343-0.435, n = 3), nevirapine 1.203 (n = 1), zidovudine 0.718 (0.711-1.227, n = 5) and abacavir 1.344 (0.670-2.450, n = 10). CSF concentrations were below the IC50 for tenofovir (100%), emtricitabine (100%), abacavir (50%) and zidovudine (17%). Lamivudine, lopinavir, efavirenz and nevirapine concentrations were all above the IC50. All participants were virologically suppressed in blood and CSF. CSF drug concentrations were not associated with blood-brain barrier permeability or neurocognitive function. Conclusions We showed adequate CSF concentrations of lamivudine, lopinavir, efavirenz and nevirapine, and potential suboptimal CSF concentrations of tenofovir, abacavir and emtricitabine in long-term treated HIV-infected children. None the less, the use of combined antiretroviral drugs led to adequate viral suppression.
Collapse
Affiliation(s)
- Malon Van den Hof
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte Blokhuis
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sophie Cohen
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Henriette J Scherpbier
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Ferdinand W N M Wit
- Department of Internal Medicine, Division of Infectious Diseases, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.,Department of Global Health, Academic Medical Center, University of Amsterdam, and Amsterdam Institute for Global Health and Development, Pietersbergweg 7, 1105 BM The Netherlands.,HIV Monitoring Foundation, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - M C M Pistorius
- Department of Hospital Pharmacy-Clinical Pharmacology Unit, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Academic Medical Centre, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, VU University Medical Centre and Neurocampus Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Ron A A Mathot
- Department of Hospital Pharmacy-Clinical Pharmacology Unit, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Emma Children's Hospital/Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
34
|
Bavaro DF, Calamo A, Lepore L, Fabrizio C, Saracino A, Angarano G, Monno L. Cerebrospinal fluid compartmentalization of HIV-1 and correlation with plasma viral load and blood-brain barrier damage. Infection 2019; 47:441-446. [PMID: 30649685 DOI: 10.1007/s15010-019-01268-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/06/2019] [Indexed: 01/17/2023]
Abstract
PURPOSE We aimed to evaluate HIV-1 compartmentalization between the cerebrospinal fluid (CSF) and plasma and investigate as to which extent HIV-1 strains in CSF differ from those in blood and whether a correlation with either plasma viral load (pVL) or an altered blood-brain barrier (BBB) does exist. STUDY DESIGN We retrospectively evaluated paired CSF/blood samples collected from 86 HIV+ patients. HIV-RNA quantification, pol (PR/RT), and V3 sequencing were performed. HIV coreceptor tropism (CRT) was inferred (g2p, false-positive rate 10%, FPR). Data of standard CSF analysis were also reviewed; an altered CSF/plasma albumin ratio signified BBB damage. Neurological abnormalities (NA) were recorded. RESULTS Overall, 32% of patients had a CSF/plasma HIV-RNA ratio > 1 (discordance); 3% of patients had detectable CSF HIV-RNA despite suppressed pVL (escape). Discordance was more frequent in ART-treated patients (p < 0.001) and in patients with NA (p = 0.016), but was independent of BBB damage (p = 0.65) and AIDS diagnosis (p = 0.96). Finally, CSF/plasma discordance was significantly more frequent (p < 0.0001) in patients with lower pVL values (< 10.000 copies/ml). Env divergence > 10% was found in 44% of sequences and was associated with ART (p = 0.008) and NA (p = 0.037). Overall, 24% of patients had a discordant CSF/blood CRT. A 100% nucleotide identity was observed in only 7.3% of pol sequences; notably, 10% of patients had resistance-associated mutations in CSF, but not in blood. CONCLUSIONS Our data confirm an independent replication and evolution of HIV within the CSF. A number of factors either hinder or contribute to the compartmentalization of HIV.
Collapse
Affiliation(s)
- Davide Fiore Bavaro
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy.
| | - A Calamo
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy
| | - L Lepore
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy
| | - C Fabrizio
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy
| | - A Saracino
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy
| | - G Angarano
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy
| | - L Monno
- Clinic of Infectious Diseases, University of Bari "Aldo Moro", Piazza G. Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
35
|
Srinivas N, Rosen EP, Gilliland WM, Kovarova M, Remling-Mulder L, De La Cruz G, White N, Adamson L, Schauer AP, Sykes C, Luciw P, Garcia JV, Akkina R, Kashuba ADM. Antiretroviral concentrations and surrogate measures of efficacy in the brain tissue and CSF of preclinical species. Xenobiotica 2018; 49:1192-1201. [PMID: 30346892 DOI: 10.1080/00498254.2018.1539278] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
1. Antiretroviral concentrations in cerebrospinal fluid (CSF) are used as surrogate for brain tissue, although sparse data support this. We quantified antiretrovirals in brain tissue across preclinical models, compared them to CSF, and calculated 90% inhibitory quotients (IQ90) for nonhuman primate (NHP) brain tissue. Spatial distribution of efavirenz was performed by mass-spectrometry imaging (MSI). 2. HIV or RT-SHIV-infected and uninfected animals from two humanized mouse models (hemopoietic-stem cell/RAG2-, n = 36; bone marrow-liver-thymus/BLT, n =13) and an NHP model (rhesus macaque, n =18) were dosed with six antiretrovirals. Brain tissue, CSF (NHPs), and plasma were collected at necropsy. Drug concentrations were measured by LC-MS/MS. Rapid equilibrium dialysis determined protein binding in NHP brain. 3. Brain tissue penetration of most antiretrovirals were >10-fold lower (p < 0.02) in humanized mice than NHPs. NHP CSF concentrations were >13-fold lower (p <0.02) than brain tissue with poor agreement except for efavirenz (r = 0.91, p = 0.001). Despite 97% brain tissue protein binding, efavirenz achieved IQ90>1 in all animals and 2-fold greater white versus gray matter concentration. 4. Brain tissue penetration varied across animal models for all antiretrovirals except raltegravir, and extrapolating brain tissue concentrations between models should be avoided. With the exception of efavirenz, CSF is not a surrogate for brain tissue concentrations.
Collapse
Affiliation(s)
- Nithya Srinivas
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Elias P Rosen
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - William M Gilliland
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Martina Kovarova
- b School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | | | - Gabriela De La Cruz
- b School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Nicole White
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Lourdes Adamson
- d School of Medicine , University of California at Davis , Davis , CA , USA
| | - Amanda P Schauer
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Craig Sykes
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Paul Luciw
- d School of Medicine , University of California at Davis , Davis , CA , USA
| | - J Victor Garcia
- b School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Ramesh Akkina
- c School of Medicine , Colorado State University , Fort Collins , CO , USA
| | - Angela D M Kashuba
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| |
Collapse
|
36
|
Persistent central nervous system immune activation following more than 10 years of effective HIV antiretroviral treatment. AIDS 2018; 32:2171-2178. [PMID: 30005007 DOI: 10.1097/qad.0000000000001950] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Low-grade immune activation is common in people living with HIV (PLHIV), despite long-term viral suppression by antiretroviral therapy (ART). The clinical significance of this activation remains unclear. The aim of this study was to examine residual intrathecal immune activation in relation to signs of neuronal injury and neurocognitive impairment in PLHIV who had been virally suppressed on ART for more than 10 years. DESIGN/METHODS Twenty neuroasymptomatic PLHIV on suppressive ART for a median of 13.2 years were retrospectively identified from the longitudinal prospective Gothenburg HIV cerebrospinal fluid (CSF) study. HIV-RNA, neopterin, and neurofilament light protein (NFL) levels were measured in paired plasma and CSF samples. Pretreatment samples were available for 14 patients. Cognitive function was assessed by CogState at follow-up. RESULTS CSF neopterin decreased from a median (IQR) of 17.8 (10.6-29.7) to 6.1 (4.6-8.0) nmol/l during treatment (P < 0.001). In 11 out of 20 participants (55%), CSF neopterin levels were above the upper normal reference limit (5.8 nmol/l) at follow-up. Age-adjusted CSF NFL decreased to within-normal levels from a median of (IQR) 1179 (557-2707) to 415 (292-610) ng/l (P < 0.001). No significant correlations were found between CSF neopterin and CSF NFL or neurocognitive performance. CONCLUSION Although CSF neopterin decreased significantly, more than 50% of the patients had CSF concentrations above the upper normal reference value despite more than 10 years of suppressive ART. We found no correlation between CSF neopterin, CSF NFL or neurocognitive performance at follow-up, indicating that low-grade immune activation during suppressive ART may be clinically benign.
Collapse
|
37
|
Exploring an alternative explanation for the second phase of viral decay: Infection of short-lived cells in a drug-limited compartment during HAART. PLoS One 2018; 13:e0198090. [PMID: 30016329 PMCID: PMC6049925 DOI: 10.1371/journal.pone.0198090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/14/2018] [Indexed: 11/19/2022] Open
Abstract
Most HIV-infected patients who initiate combination antiretroviral therapy experience a viral load decline in several phases. These phases are characterized by different rates of viral load decay that decrease when transitioning from one phase to the next. There is no consensus as to the origin of these phases. One hypothesis put forward is that short- and long-lived infected cells are responsible for the first and second phases of decay, respectively. However, significant differences in drug concentrations are observed in monocytes from various tissues, suggesting the first two phases of decay in viral loads could instead be attributed to short-lived cells being differently exposed to drugs. Compared to a well-exposed compartment, new cell infection can be expected in a compartment with limited drug exposure, thus leading to a slower viral load decay with potential virologic failure and drug resistance. In the current study, the latter hypothesis was investigated using a model of viral kinetics. Empirical datasets were involved in model elaboration and parameter estimation. In particular, susceptibility assay data was used for an in vitro to in vivo extrapolation based on the expected drug concentrations inside physiological compartments. Results from numerical experiments of the short-term evolution of viral loads can reproduce the first two phases of viral decay when allowing new short-lived cell infections in an unidentified drug-limited compartment. Model long-term predictions are however less consistent with clinical observations. For the hypothesis to hold, efavirenz, tenofovir and emtricitabine drug exposure in the drug-limited compartment would have to be very low compared to exposure in peripheral blood. This would lead to significant long-term viral growth and the frequent development of resistant strains, a prediction not supported by clinical observations. This suggests that the existence of a drug-limited anatomical compartment is unlikely, by itself, to explain the second phase of viral load decay.
Collapse
|
38
|
Kumar S, Maurya VK, Dandu HR, Bhatt ML, Saxena SK. Global Perspective of Novel Therapeutic Strategies for the Management of NeuroAIDS. Biomol Concepts 2018; 9:33-42. [PMID: 29742062 DOI: 10.1515/bmc-2018-0005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/06/2018] [Indexed: 11/15/2022] Open
Abstract
Among Human immunodeficiency virus (HIV) infected individuals, around two-thirds of patients present with neuroAIDS, where HIV-associated neurocognitive disorders (HAND), and HIV-associated dementia (HAD) are the most prevailing neurological complications. The neuropathology of neuroAIDS can be characterized by the presence of HIV infected macrophages and microglia in the brain, with the formation of multinucleated giant cells. Global predominant subtypes of HIV-1 clade B and C infections influence the differential effect of immune and neuronal dysfunctions, leading to clade-specific clinical variation in neuroAIDS patient cohorts. Highly active antiretroviral therapy (HAART) enhances the survival rate among AIDS patients, but due to the inability to cross the Blood-Brain-Barrier (BBB), incidence of neuroAIDS during disease progression may be envisaged. The complex structure of blood-brain-barrier, and poor pharmacokinetic profile coupled with weak bio-distribution of antiretroviral drugs, are the principle barriers for the treatment of neuroAIDS. In the combined antiretroviral therapy (cART) era, the frequency of HAD has decreased; however the incidence of asymptomatic neurocognitive impairment (ANI) and minor neurocognitive disorder (MND) remains consistent. Therefore, several effective novel nanotechnology based therapeutic approaches have been developed to improve the availability of antiretroviral drugs in the brain for the management of neuroAIDS.
Collapse
Affiliation(s)
- Swatantra Kumar
- Center for Advanced Research (CFAR)-Stem Cell/Cell Culture Unit, King George's Medical University (KGMU), Lucknow, 226003, India
| | - Vimal K Maurya
- Center for Advanced Research (CFAR)-Stem Cell/Cell Culture Unit, King George's Medical University (KGMU), Lucknow, 226003, India
| | - Himanshu R Dandu
- Center for Advanced Research (CFAR)-Stem Cell/Cell Culture Unit, King George's Medical University (KGMU), Lucknow, 226003, India
| | - Madan Lb Bhatt
- Center for Advanced Research (CFAR)-Stem Cell/Cell Culture Unit, King George's Medical University (KGMU), Lucknow, 226003, India
| | - Shailendra K Saxena
- Center for Advanced Research (CFAR)-Stem Cell/Cell Culture Unit, King George's Medical University (KGMU), Lucknow, 226003, India.,CSIR-Centre for Cellular and Molecular Biology, UppalRoad, Hyderabad, 500007, India
| |
Collapse
|
39
|
Imaz A, Niubó J, Amara A, Khoo S, Ferrer E, Tiraboschi JM, Acerete L, Garcia B, Vila A, Podzamczer D. Cerebrospinal fluid drug concentrations and viral suppression in HIV-1-infected patients receiving ritonavir-boosted atazanavir plus lamivudine dual antiretroviral therapy (Spanish HIV/AIDS Research Network, PreEC/RIS 39). J Neurovirol 2018. [PMID: 29542028 DOI: 10.1007/s13365-018-0626-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study aimed to assess cerebrospinal fluid (CSF) drug concentrations and viral suppression in HIV-1-infected patients on ritonavir-boosted atazanavir (ATV/r) plus lamivudine (3TC) dual therapy. HIV-1-infected adults with suppressed plasma HIV-1 RNA who switched to ATV/r plus 3TC were studied. Total ATV and 3TC concentrations at the end of the dosing interval (C24h), using a validated LC-MS/MS method, and HIV-1 RNA were measured in paired CSF and plasma samples 12 weeks after switching. Ten individuals were included. Median (range) age was 42.5 (33-70) years, time on ART was 39.5 (11-197) months, and time with plasma HIV-1 RNA < 40 copies/mL was 15.5 (6-46) months. At baseline, CSF HIV-1 RNA was < 40 copies/mL in all patients. Twelve weeks after switching to ATV/r plus 3TC, HIV-1 RNA remained at < 40 copies/mL in both plasma and CSF in 9/10 patients. One patient with suboptimal adherence to ART had HIV-1 RNA rebound in both plasma and CSF. The median CSF-to-plasma concentration ratios of ATV and 3TC were 0.013 and 0.417, respectively. Median ATV C24h in CSF was 10.4 (3.7-33.4) ng/mL (in vitro ATV IC50 range, 1-11 ng/mL). Median 3TC C24h in CSF was 43.4 (16.2-99.3) ng/mL (in vitro 3TC IC50 range, 0.68-20.6 ng/mL). Most patients maintained HIV-1 RNA in CSF < 40 copies/mL despite CSF ATV C24h close to or within the IC50 range in the majority. ATV PK data in CSF should be considered and rigorous patient selection is advisable to assure effective CSF viral suppression with this two-drug simplification regimen.
Collapse
Affiliation(s)
- Arkaitz Imaz
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | - Jordi Niubó
- Department of Microbiology, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alieu Amara
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Saye Khoo
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Elena Ferrer
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Juan M Tiraboschi
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Laura Acerete
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Benito Garcia
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Antonia Vila
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Daniel Podzamczer
- HIV and STI Unit, Department of Infectious Diseases, Bellvitge University Hospital-IDIBELL, Feixa Llarga S/N, L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| |
Collapse
|
40
|
Shepherd SJ, Black H, Thomson EC, Gunson RN. HIV positive patient with GBS-like syndrome. JMM Case Rep 2017; 4:e005107. [PMID: 29026634 PMCID: PMC5610709 DOI: 10.1099/jmmcr.0.005107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/21/2017] [Indexed: 12/17/2022] Open
Abstract
Introduction. Guillain-Barré Syndrome (GBS) is an acute demyelinating polyneuropathy which can occur post-infection. Criteria of diagnosis of GBS include areflexia with progressive bilateral weakness in arms and legs. GBS can lead to severe respiratory and cardiac complications. The fatality rate can be up to 5 % in patients, depending on the severity of the symptoms. HIV can cause a range of neurological disorders including, on rare occasions, GBS. GBS can occur at any stage of HIV infection, highlighting the complexity of diagnosis of GBS within HIV patients. Case presentation. A 57 year old female with lumbar back pain radiating to the legs, poor mobility and tiredness, with reports of a viral-like illness four days previously, was initially diagnosed with a lower respiratory tract infection and discharged. Seventeen days later the patient was readmitted to hospital with progressive lower and upper limb weakness, areflexia and sensory loss. She was diagnosed with GBS and was unexpectedly discovered to be HIV-positive. HIV avidity was low indicating a recently acquired HIV infection. The patient was treated with intravenous immunoglobulin for five days for the GBS and commenced antriretrovirals for HIV. The patient was discharge from hospital 53 days after admission with walking aids and regular physiotherapy follow-up. CONCLUSION . This case highlighted the need for all clinicians to be aware that patients with symptoms of GBS, regardless of clinical history should be offered an HIV test. GBS can be the first sign a patient is HIV-positive.
Collapse
Affiliation(s)
- Samantha J Shepherd
- West of Scotland Specialist Virology Centre, Level 5, New Lister Building, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
| | - Heather Black
- Infectious Diseases Unit, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK
| | - Emma C Thomson
- MRC-University of Glasgow Centre for Virus Research, Stoker Building, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Rory N Gunson
- West of Scotland Specialist Virology Centre, Level 5, New Lister Building, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
| |
Collapse
|
41
|
Development and validation of an LC-MS/MS assay for the quantification of efavirenz in different biological matrices. Bioanalysis 2016; 8:2125-34. [PMID: 27611731 DOI: 10.4155/bio-2016-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
AIM The non-nucleoside reverse transcriptase inhibitor efavirenz is one of the most prescribed antiretroviral therapeutics. Efavirenz-containing therapy has become associated with the occurrence of CNS side effects, including sleep disturbances, depression and even psychosis. RESULTS The investigation of efavirenz distribution required the development of a versatile and sensitive method. In addition to plasma, quantification was required in brain tissue and phosphate-buffered saline. The assay presented here was linear from 1.9 to 500 ng/ml. Accuracy and precision ranged between 93.7 and 99.5%, and 1.5 and 5.6%, respectively. DISCUSSION The method developed here represents a versatile, sensitive and easy-to-use assay. The assay has been applied to in vitro and in vivo samples demonstrating reliable efavirenz quantification in multiple matrices.
Collapse
|
42
|
Qin S, Chen Q, Wu H, Liu C, Hu J, Zhang D, Xu C. Effects of naringin on learning and memory dysfunction induced by gp120 in rats. Brain Res Bull 2016; 124:164-71. [PMID: 27154619 DOI: 10.1016/j.brainresbull.2016.04.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/20/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022]
Abstract
The aim of the present study was to investigate the effects of naringin on learning and memory dysfunction induced by HIV-1-enveloped protein gp120 in rats, and to identify its potential mechanisms of action. Learning and memory ability was evaluated via Morris water maze test, P2X7 receptor and P65 protein expressions in the rat hippocampus were detected by western blot analysis, and P2X7 mRNA expression in the hippocampus was measured by RT-PCR. We also recorded P2X7 agonist BzATP-activated current in the hippocampus via patch clamp technique. The results showed that naringin treatment (30mg/kg/day) markedly decreased the escape latency and target platform errors of rats treated with gp120 (50ng/day), and further, that naringin treatment significantly decreased the expression of P2X7 and P65 protein and P2X7 mRNA in the hippocampus of gp120-treated rats. In addition, naringin treatment reduced BzATP-activated current in the hippocampus of gp120-treated rats. These results altogether demonstrated that naringin can improve gp120-induced learning and memory dysfunction via mechanisms involving the inhibition of P2X7 expression in the hippocampus.
Collapse
Affiliation(s)
- Shanshan Qin
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, PR China
| | - Qiang Chen
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, PR China
| | - Hui Wu
- Second Clinical Medical College of Nanchang University, Nanchang 330006, PR China
| | - Chenglong Liu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, PR China
| | - Jing Hu
- Second Clinical Medical College of Nanchang University, Nanchang 330006, PR China
| | - Dalei Zhang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, PR China
| | - Changshui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
43
|
Heikinheimo T, Poutiainen E, Salonen O, Elovaara I, Ristola M. Three-decade neurological and neurocognitive follow-up of HIV-1-infected patients on best-available antiretroviral therapy in Finland. BMJ Open 2015; 5:e007986. [PMID: 26546135 PMCID: PMC4636633 DOI: 10.1136/bmjopen-2015-007986] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES Is it possible to live without neurocognitive or neurological symptoms after being infected with HIV for a very long time? These study patients with decades-long HIV infection in Finland were observed in this follow-up study during three time periods: 1986-1990, in 1997 and in 2013. SETTING Patients from greater Helsinki area were selected from outpatient's unit of infectious diseases. PARTICIPANTS The study included 80 HIV patients. Patients with heavy alcohol consumption, central nervous system disorder or psychiatric disease were excluded. PRIMARY AND SECONDARY OUTCOME MEASURES The patients underwent neurological and neuropsychological examinations, MRI of the brain and laboratory tests, including blood CD4 cells and plasma HIV-1 RNA. Neuropsychological examination included several measures: subtests of Wechsler Adult Intelligence Scale, Wechsler Memory Scale-Revised, list learning, Stroop and Trail-Making-B test. The Beck Depression Inventory and Fatigue Severity Scale were also carried out. The obtained data from the three time periods were compared with each other. RESULTS Owing to high mortality among the original 80 patients, eventually, 17 participated in all three examinations performed between 1986 and 2013. The time from the HIV diagnosis was 27 (23-30) years. Blood CD4 cells at the diagnosis were 610 (29-870) cells/mm(3), and the nadir CD4 168 (4-408) cells/mm(3). The time on combined antiretroviral treatment was 13 (5-17) years. 9 patients suffered from fatigue, 5 had polyneuropathy and 3 had lacunar cerebral infarcts. There was a subtle increase of brain atrophy in 2 patients. Mild depressive symptoms were common. The neuropsychological follow-up showed typical age-related cognitive changes. No HIV-associated dementia features were detected. CONCLUSIONS Polyneuropathy, fatigue and mild depression were common, but more severe neurological abnormalities were absent. These long-term surviving HIV-seropositive patients, while on best-available treatment, showed no evidence of HIV-associated neurocognitive disorder in neuropsychological and neuroradiological evaluations.
Collapse
Affiliation(s)
- T Heikinheimo
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - E Poutiainen
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
- Rehabilitation Foundation, Helsinki, Finland
| | - O Salonen
- Department of Radiology, Helsinki University Central Hospital, Helsinki, Finland
| | - I Elovaara
- Neuroimmunology Unit, Medical School, University of Tampere, Tampere, Finland
| | - M Ristola
- Department of Infectious Diseases at Aurora Hospital, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
44
|
Peterson CW, Haworth KG, Polacino P, Huang ML, Sykes C, Obenza WM, Repetto AC, Kashuba A, Bumgarner R, DeRosa SC, Woolfrey AE, Jerome KR, Mullins JI, Hu SL, Kiem HP. Lack of viral control and development of combination antiretroviral therapy escape mutations in macaques after bone marrow transplantation. AIDS 2015; 29:1597-606. [PMID: 26372270 PMCID: PMC4572605 DOI: 10.1097/qad.0000000000000702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We have previously demonstrated robust control of simian/human immunodeficiency virus (SHIV1157-ipd3N4) viremia following administration of combination antiretroviral therapy (cART) in pigtailed macaques. Here, we sought to determine the safety of hematopoietic stem cell transplantation (HSCT) in cART-suppressed and unsuppressed animals. DESIGN We compared disease progression in animals challenged with SHIV 100 days post-transplant, to controls that underwent transplant following SHIV challenge and stable cART-dependent viral suppression. METHODS SHIV viral load, cART levels, and anti-SHIV antibodies were measured longitudinally from plasma/serum from each animal. Flow cytometry was used to assess T-cell subset frequencies in peripheral blood and the gastrointestinal tract. Deep sequencing was used to identify cART resistance mutations. RESULTS In control animals, virus challenge induced transient peak viremia, viral set point, and durable suppression by cART. Subsequent HSCT was not associated with adverse events in these animals. Post-transplant animals were challenged during acute recovery following HSCT, and displayed sustained peak viremia and cART resistance. Although post-transplant animals had comparable plasma levels of antiretroviral drugs and showed no evidence of enhanced infection of myeloid subsets in the periphery, they exhibited a drastic reduction in virus-specific antibody production and decreased T-cell counts. CONCLUSIONS These results suggest that virus challenge prior to complete transplant recovery impairs viral control and may promote drug resistance. These findings may also have implications for scheduled treatment interruption studies in patients on cART during post-HSCT recovery: premature scheduled treatment interruption could similarly result in lack of viral control and cART resistance.
Collapse
Affiliation(s)
- Christopher W Peterson
- aClinical Research Division, Fred Hutchinson Cancer Research Center bWashington National Primate Research Center, Seattle cVaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington dDivision of Pharmacotherapy and Experimental Therapeutics, University of North Carolina, Chapel Hill, North Carolina eDepartment of Microbiology fDepartment of Pediatrics gDepartment of Laboratory Medicine hDepartment of Medicine iDepartment of Pharmaceutics jDepartment of Pathology, University of Washington, Seattle, Washington, USA. *Christopher W. Peterson and Kevin G. Haworth contributed equally to the writing of this article
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Calcagno A, Di Perri G, Bonora S. Pharmacokinetics and pharmacodynamics of antiretrovirals in the central nervous system. Clin Pharmacokinet 2015; 53:891-906. [PMID: 25200312 DOI: 10.1007/s40262-014-0171-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
HIV-positive patients may be effectively treated with highly active antiretroviral therapy and such a strategy is associated with striking immune recovery and viral load reduction to very low levels. Despite undeniable results, the central nervous system (CNS) is commonly affected during the course of HIV infection, with neurocognitive disorders being as prevalent as 20-50 % of treated subjects. This review discusses the pathophysiology of CNS infection by HIV and the barriers to efficacious control of such a mechanism, including the available data on compartmental drug penetration and on pharmacokinetic/pharmacodynamic relationships. In the reviewed articles, a high variability in drug transfer to the CNS is highlighted with several mechanisms as well as methodological issues potentially influencing the observed results. Nevirapine and zidovudine showed the highest cerebrospinal fluid (CSF) to plasma ratios, although target concentrations are currently unknown for the CNS. The use of the composite CSF concentration effectiveness score has been associated with better virological outcomes (lower HIV RNA) but has been inconsistently associated with neurocognitive outcomes. These findings support the CNS effectiveness of commonly used highly antiretroviral therapies. The use of antiretroviral drugs with increased CSF penetration and/or effectiveness in treating or preventing neurocognitive disorders however needs to be assessed in well-designed prospective studies.
Collapse
Affiliation(s)
- Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, c/o Ospedale Amedeo di Savoia, C.so Svizzera 164, 10159, Torino, Italy,
| | | | | |
Collapse
|
46
|
Boska MD, Dash PK, Knibbe J, Epstein AA, Akhter SP, Fields N, High R, Makarov E, Bonasera S, Gelbard HA, Poluektova LY, Gendelman HE, Gorantla S. Associations between brain microstructures, metabolites, and cognitive deficits during chronic HIV-1 infection of humanized mice. Mol Neurodegener 2014; 9:58. [PMID: 25523827 PMCID: PMC4297430 DOI: 10.1186/1750-1326-9-58] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/07/2014] [Indexed: 12/31/2022] Open
Abstract
Background Host-species specificity of the human immunodeficiency virus (HIV) limits pathobiologic, diagnostic and therapeutic research investigations to humans and non-human primates. The emergence of humanized mice as a model for viral infection of the nervous system has overcome such restrictions enabling research for HIV-associated end organ disease including behavioral, cognitive and neuropathologic deficits reflective of neuroAIDS. Chronic HIV-1 infection of NOD/scid-IL-2Rgcnull mice transplanted with human CD34+ hematopoietic stem cells (CD34-NSG) leads to persistent viremia, profound CD4+ T lymphocyte loss and infection of human monocyte-macrophages in the meninges and perivascular spaces. Murine cells are not infected with virus. Methods Changes in mouse behavior were measured, starting at 8 weeks after viral infection. These were recorded coordinate with magnetic resonance spectroscopy metabolites including N-acetylaspartate (NAA), creatine and choline. Diffusion tensor magnetic resonance imaging (DTI) was recorded against multispectral immunohistochemical staining for neuronal markers that included microtubule associated protein-2 (MAP2), neurofilament (NF) and synaptophysin (SYN); for astrocyte glial fibrillary acidic protein (GFAP); and for microglial ionized calcium binding adaptor molecule 1 (Iba-1). Oligodendrocyte numbers and integrity were measured for myelin associated glycoprotein (MAG) and myelin oligodendrocyte glycoprotein (MOG) antigens. Results Behavioral abnormalities were readily observed in HIV-1 infected mice. Longitudinal open field activity tests demonstrated lack of habituation indicating potential for memory loss and persistent anxiety in HIV-1 infected mice compared to uninfected controls. End-point NAA and creatine in the cerebral cortex increased with decreased MAG. NAA and glutamate decreased with decreased SYN and MAG. Robust inflammation reflected GFAP and Iba-1 staining intensities. DTI metrics were coordinate with deregulation of NF, Iba-1, MOG and MAG levels in the whisker barrel and MAP2, NF, MAG, MOG and SYN in the corpus callosum. Conclusions The findings are consistent with some of the clinical, biochemical and pathobiologic features of human HIV-1 nervous system infections. This model will prove useful towards investigating the mechanisms of HIV-1 induced neuropathology and in developing novel biomarkers and therapeutic strategies for disease. Electronic supplementary material The online version of this article (doi:10.1186/1750-1326-9-58) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | | |
Collapse
|
47
|
Calcagno A, Simiele M, Alberione MC, Bracchi M, Marinaro L, Ecclesia S, Di Perri G, D'Avolio A, Bonora S. Cerebrospinal fluid inhibitory quotients of antiretroviral drugs in HIV-infected patients are associated with compartmental viral control. Clin Infect Dis 2014; 60:311-7. [PMID: 25281609 DOI: 10.1093/cid/ciu773] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Despite the efficacy of highly active antiretroviral treatment (HAART), a large proportion of human immunodeficiency virus (HIV)-infected patients may develop moderate neurocognitive impairment. Antiretroviral drug passage into the central nervous system may be relevant for preventing and treating HIV-associated neurocognitive disorder; nevertheless, clear cerebrospinal fluid (CSF) pharmacodynamic targets are not known. METHODS HAART-treated adults with wild-type HIV were prospectively enrolled. CSF concentrations (measured by mass spectrophotometric methods) and inhibitory quotients (CSF concentrations divided by in vitro 50% and 95% inhibitory concentrations) were compared among different drugs and related to CSF HIV RNA levels. CSF escape was defined as CSF HIV RNA >50 copies/mL despite contemporary plasma HIV RNA below that threshold. RESULTS One hundred twenty-seven patients (91 male [71.7%], 93 white [73.2%], with a median age of 46 years [interquartile range, 40.5-54.5 years]) provided 174 paired CSF and plasma samples. Twice-daily darunavir, once-daily darunavir, and efavirenz had the highest CSF 95% inhibitory quotients (18.5, 8.2, and 6.4, respectively). Higher nadir CD4 cell count (P = .01) and plasma HIV RNA <50 copies/mL (P < .001) were independent predictors of controlled CSF HIV RNA. Optimal drug exposure (CSF detectable drugs and 95% inhibitory quotient >1) was protective for CSF escape (P = .01). CONCLUSIONS Cerebrospinal fluid 95% inhibitory quotients may be used to compare antiretroviral drug compartmental exposure; they deserve longitudinal studies to assess the adequacy of CSF drug concentrations in treated HIV-infected patients.
Collapse
Affiliation(s)
- Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Marco Simiele
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Maria Chiara Alberione
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Margherita Bracchi
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Letizia Marinaro
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Sara Ecclesia
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Giovanni Di Perri
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Antonio D'Avolio
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - Stefano Bonora
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| |
Collapse
|
48
|
Price RW, Spudich SS, Peterson J, Joseph S, Fuchs D, Zetterberg H, Gisslén M, Swanstrom R. Evolving character of chronic central nervous system HIV infection. Semin Neurol 2014; 34:7-13. [PMID: 24715483 DOI: 10.1055/s-0034-1372337] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system (CNS) begins early in systemic infection and continues throughout its untreated course. Despite a common cerebrospinal fluid inflammatory response, it is usually neurologically asymptomatic for much of this course, but can evolve in some individuals to HIV-associated dementia (HAD), a severe encephalopathy with characteristic cognitive and motor dysfunction. While widespread use of combination antiretroviral therapy (ART) has led to a marked decline in both the CNS infection and its neurologic severe consequence, HAD continues to afflict individuals presenting with advanced systemic infection in the developed world and a larger number in resource-poor settings where ART is more restricted. Additionally, milder CNS injury and dysfunction have broader prevalence, including in those treated with ART. Here we review the history and evolving nomenclature of HAD, its viral pathogenesis, clinical presentation and diagnosis, and treatment.
Collapse
Affiliation(s)
- Richard W Price
- Department of Neurology, University of California, San Francisco, California
| | - Serena S Spudich
- Department of Neurology, Yale University, New Haven, Connecticut
| | - Julia Peterson
- Department of Neurology, University of California, San Francisco, California
| | - Sarah Joseph
- UNC Center for AIDS Research and the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden and Institute of Neurology, Queen Square, London, United Kingdom
| | - Magnus Gisslén
- Department of Infectious Diseases, University of Gothenburg, Gothenburg, Sweden
| | - Ronald Swanstrom
- UNC Center for AIDS Research and the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
49
|
Affiliation(s)
- Marie F Grill
- Department of Neurology, Division of Hospital Neurology, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - Richard W Price
- Department of Neurology, School of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
50
|
Lisi L, Tramutola A, Navarra P, Dello Russo C. Antiretroviral agents increase NO production in gp120/IFNγ-stimulated cultures of rat microglia via an arginase-dependent mechanism. J Neuroimmunol 2014; 266:24-32. [DOI: 10.1016/j.jneuroim.2013.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/31/2013] [Accepted: 10/31/2013] [Indexed: 12/22/2022]
|