1
|
Faivre N, Verollet C, Dumas F. The chemokine receptor CCR5: multi-faceted hook for HIV-1. Retrovirology 2024; 21:2. [PMID: 38263120 PMCID: PMC10807162 DOI: 10.1186/s12977-024-00634-1] [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: 09/27/2023] [Accepted: 01/13/2024] [Indexed: 01/25/2024] Open
Abstract
Chemokines are cytokines whose primary role is cellular activation and stimulation of leukocyte migration. They perform their various functions by interacting with G protein-coupled cell surface receptors (GPCRs) and are involved in the regulation of many biological processes such as apoptosis, proliferation, angiogenesis, hematopoiesis or organogenesis. They contribute to the maintenance of the homeostasis of lymphocytes and coordinate the function of the immune system. However, chemokines and their receptors are sometimes hijacked by some pathogens to infect the host organism. For a given chemokine receptor, there is a wide structural, organizational and conformational diversity. In this review, we describe the evidence for structural variety reported for the chemokine receptor CCR5, how this variability can be exploited by HIV-1 to infect its target cells and what therapeutic solutions are currently being developed to overcome this problem.
Collapse
Affiliation(s)
- Natacha Faivre
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Fabrice Dumas
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France.
| |
Collapse
|
2
|
Zhao L, Lai Y. Monoclonal CCR5 Antibody: A Promising Therapy for HIV. Curr HIV Res 2023; 21:91-98. [PMID: 36927434 DOI: 10.2174/1570162x21666230316110830] [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: 10/05/2022] [Revised: 01/07/2023] [Accepted: 01/20/2023] [Indexed: 03/18/2023]
Abstract
HIV is one of the world's most devastating viral infections and has claimed tens of millions of lives worldwide since it was first identified in the 1980s. There is no cure for HIV infection. However, with tremendous progress in HIV diagnosis, prevention, and treatment, HIV has become a manageable chronic health disease. CCR5 is an important coreceptor used by HIV to infect target cells, and genetic deficiency of the chemokine receptor CCR5 confers a significant degree of protection against HIV infection. In addition, since CCR5 deficiency does not appear to cause any adverse health effects, targeting this coreceptor is a promising strategy for the treatment and prevention of HIV. Monoclonal antibodies are frequently used as therapeutics for many diseases and therefore are being used as a potential therapy for HIV-1 infection. This review reports on CCR5 antibody research in detail and describes the role and advantages of CCR5 antibodies in HIV prevention or treatment, introduces several main CCR5 antibodies, and discusses the future strategy of antibody-conjugated nanoparticles including the potential challenges. CCR5 antibodies may be a novel therapy for treating HIV infection effectively and could overcome the limitations of the currently available options.
Collapse
Affiliation(s)
- Li Zhao
- Acupunture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yu Lai
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Viel KR. Response to 'Reduced Cell Surface Levels of C-C Chemokine Receptor 5 and Immunosuppression in Long Coronavirus Disease 2019 Syndrome'. Clin Infect Dis 2022; 75:1485-1486. [PMID: 35819236 PMCID: PMC9384431 DOI: 10.1093/cid/ciac389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Kevin R Viel
- Department of Genomics, Histonis Incorporated, Manchester, New Hampshire, USA
| |
Collapse
|
4
|
Gaylis NB, Ritter A, Kelly SA, Pourhassan NZ, Tiwary M, Sacha JB, Hansen SG, Recknor C, Yang OO. Reduced Cell Surface Levels of C-C Chemokine Receptor 5 and Immunosuppression in Long Coronavirus Disease 2019 Syndrome. Clin Infect Dis 2022; 75:1232-1234. [PMID: 35452519 PMCID: PMC9383814 DOI: 10.1093/cid/ciac226] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Indexed: 11/12/2022] Open
Abstract
In an exploratory trial treating "long COVID" with the CCR5-binding antibody leronlimab, we observed significantly increased blood cell surface CCR5 in treated symptomatic responders but not in nonresponders or placebo-treated participants. These findings suggest an unexpected mechanism of abnormal immune downmodulation in some persons that is normalized by leronlimab. Clinical Trials Registration. NCT04678830.
Collapse
Affiliation(s)
- Norman B Gaylis
- Arthritis & Rheumatic Disease Specialties, Aventura, Florida, USA
| | - Angela Ritter
- Center for Advanced Research & Education, Gainesville, Georgia, USA
| | | | | | - Meenakshi Tiwary
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, USA.,Department of Microbiology, Immunology, and Molecular Genetics, University of California-Los Angeles, Los Angeles, California, USA
| |
Collapse
|
5
|
Mohamed H, Gurrola T, Berman R, Collins M, Sariyer IK, Nonnemacher MR, Wigdahl B. Targeting CCR5 as a Component of an HIV-1 Therapeutic Strategy. Front Immunol 2022; 12:816515. [PMID: 35126374 PMCID: PMC8811197 DOI: 10.3389/fimmu.2021.816515] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Globally, human immunodeficiency virus type 1 (HIV-1) infection is a major health burden for which successful therapeutic options are still being investigated. Challenges facing current drugs that are part of the established life-long antiretroviral therapy (ART) include toxicity, development of drug resistant HIV-1 strains, the cost of treatment, and the inability to eradicate the provirus from infected cells. For these reasons, novel anti-HIV-1 therapeutics that can prevent or eliminate disease progression including the onset of the acquired immunodeficiency syndrome (AIDS) are needed. While development of HIV-1 vaccination has also been challenging, recent advancements demonstrate that infection of HIV-1-susceptible cells can be prevented in individuals living with HIV-1, by targeting C-C chemokine receptor type 5 (CCR5). CCR5 serves many functions in the human immune response and is a co-receptor utilized by HIV-1 for entry into immune cells. Therapeutics targeting CCR5 generally involve gene editing techniques including CRISPR, CCR5 blockade using antibodies or antagonists, or combinations of both. Here we review the efficacy of these approaches and discuss the potential of their use in the clinic as novel ART-independent therapies for HIV-1 infection.
Collapse
Affiliation(s)
- Hager Mohamed
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Theodore Gurrola
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Rachel Berman
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Mackenzie Collins
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ilker K. Sariyer
- Department of Microbiology, Immunology, and Inflammation, Center for Neurovirology and Gene Editing, School of Medicine, Temple University, Philadelphia, PA, United States
| | - Michael R. Nonnemacher
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States
- *Correspondence: Brian Wigdahl,
| |
Collapse
|
6
|
Immunotherapy with Cell-Based Biological Drugs to Cure HIV-1 Infection. Cells 2021; 11:cells11010077. [PMID: 35011639 PMCID: PMC8750418 DOI: 10.3390/cells11010077] [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: 11/25/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 11/17/2022] Open
Abstract
Since its discovery 35 years ago, there have been no therapeutic interventions shown to enable full HIV-1 remission. Combined antiretroviral therapy (cART) has achieved the sustained control of HIV-1 replication, however, the life-long treatment does not eradicate long-lived latently infected reservoirs and can result in multiple side effects including the development of multidrug-resistant escape mutants. Antibody-based treatments have emerged as alternative approaches for a HIV-1 cure. Here, we will review clinical advances in coreceptor-targeting antibodies, with respect to anti-CCR5 antibodies in particular, which are currently being generated to target the early stages of infection. Among the Env-specific antibodies widely accepted as relevant in cure strategies, the potential role of those targeting CD4-induced (CD4i) epitopes of the CD4-binding site (CD4bs) in eliminating HIV-1 infected cells has gained increasing interest and will be presented. Together, with approaches targeting the HIV-1 replication cycle, we will discuss the strategies aimed at boosting and modulating specific HIV-1 immune responses, highlighting the harnessing of TLR agonists for their dual role as latency reverting agents (LRAs) and immune-modulatory compounds. The synergistic combinations of different approaches have shown promising results to ultimately enable a HIV-1 cure.
Collapse
|
7
|
Yang B, Fulcher JA, Ahn J, Berro M, Goodman-Meza D, Dhody K, Sacha JB, Naeim A, Yang OO. Clinical Characteristics and Outcomes of Coronavirus Disease 2019 Patients Who Received Compassionate-Use Leronlimab. Clin Infect Dis 2021; 73:e4082-e4089. [PMID: 33079180 PMCID: PMC7665416 DOI: 10.1093/cid/ciaa1583] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Leronlimab, a monoclonal antibody blocker of C-C chemokine receptor type 5 originally developed to treat human immunodeficiency virus infection, was administered as an open-label compassionate-use therapeutic for coronavirus disease 2019 (COVID-19). METHODS Twenty-three hospitalized severe/critical COVID-19 patients received 700 mg leronlimab subcutaneously, repeated after 7 days in 17 of 23 patients still hospitalized. Eighteen of 23 received other experimental treatments, including convalescent plasma, hydroxychloroquine, steroids, and/or tocilizumab. Five of 23 received leronlimab after blinded, placebo-controlled trials of remdesivir, sarilumab, selinexor, or tocilizumab. Outcomes and results were extracted from medical records. RESULTS Mean age was 69.5 ± 14.9 years; 20 had significant comorbidities. At baseline, 22 were receiving supplemental oxygen (3 high flow, 7 mechanical ventilation). Blood showed markedly elevated inflammatory markers (ferritin, D-dimer, C-reactive protein) and an elevated neutrophil-to-lymphocyte ratio. By day 30 after initial dosing, 17 were recovered, 2 were still hospitalized, and 4 had died. Of the 7 intubated at baseline, 4 were fully recovered off oxygen, 2 were still hospitalized, and 1 had died. CONCLUSIONS Leronlimab appeared safe and well tolerated. The high recovery rate suggested benefit, and those with lower inflammatory markers had better outcomes. Some, but not all, patients appeared to have dramatic clinical responses, indicating that unknown factors may determine responsiveness to leronlimab. Routine inflammatory and cell prognostic markers did not markedly change immediately after treatment, although interleukin-6 tended to fall. In some persons, C-reactive protein clearly dropped only after the second leronlimab dose, suggesting that a higher loading dose might be more effective. Future controlled trials will be informative.
Collapse
Affiliation(s)
- Bryant Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Jennifer A Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Jenny Ahn
- Clinical and Translational Science Institute Office of Clinical Research, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Marlene Berro
- Clinical and Translational Science Institute Office of Clinical Research, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - David Goodman-Meza
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Kush Dhody
- Amarex Clinical Research, LLC, Germantown, Maryland, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Arash Naeim
- Clinical and Translational Science Institute Office of Clinical Research, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
| | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles California, USA
| |
Collapse
|
8
|
Chang XL, Wu HL, Webb GM, Tiwary M, Hughes C, Reed JS, Hwang J, Waytashek C, Boyle C, Pessoa C, Sylwester AW, Morrow D, Belica K, Fischer M, Kelly S, Pourhassan N, Bochart RM, Smedley J, Recknor CP, Hansen SG, Sacha JB. CCR5 Receptor Occupancy Analysis Reveals Increased Peripheral Blood CCR5+CD4+ T Cells Following Treatment With the Anti-CCR5 Antibody Leronlimab. Front Immunol 2021; 12:794638. [PMID: 34868084 PMCID: PMC8640501 DOI: 10.3389/fimmu.2021.794638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
CCR5 plays a central role in infectious disease, host defense, and cancer progression, thereby making it an ideal target for therapeutic development. Notably, CCR5 is the major HIV entry co-receptor, where its surface density correlates with HIV plasma viremia. The level of CCR5 receptor occupancy (RO) achieved by a CCR5-targeting therapeutic is therefore a critical predictor of its efficacy. However, current methods to measure CCR5 RO lack sensitivity, resulting in high background and overcalculation. Here, we report on two independent, flow cytometric methods of calculating CCR5 RO using the anti-CCR5 antibody, Leronlimab. We show that both methods led to comparable CCR5 RO values, with low background on untreated CCR5+CD4+ T cells and sensitive measurements of occupancy on both blood and tissue-resident CD4+ T cells that correlated longitudinally with plasma concentrations in Leronlimab-treated macaques. Using these assays, we found that Leronlimab stabilized cell surface CCR5, leading to an increase in the levels of circulating and tissue-resident CCR5+CD4+ T cells in vivo in Leronlimab-treated macaques. Weekly Leronlimab treatment in a chronically SIV-infected macaque led to increased CCR5+CD4+ T cells levels and fully suppressed plasma viremia, both concomitant with full CCR5 RO on peripheral blood CD4+ T cells, demonstrating that CCR5+CD4+ T cells were protected from viral replication by Leronlimab binding. Finally, we extended these results to Leronlimab-treated humans and found that weekly 700 mg Leronlimab led to complete CCR5 RO on peripheral blood CD4+ T cells and a statistically significant increase in CCR5+CD4+ T cells in peripheral blood. Collectively, these results establish two RO calculation methods for longitudinal monitoring of anti-CCR5 therapeutic antibody blockade efficacy in both macaques and humans, demonstrate that CCR5+CD4+ T cell levels temporarily increase with Leronlimab treatment, and facilitate future detailed investigations into the immunological impacts of CCR5 inhibition in multiple pathophysiological processes.
Collapse
Affiliation(s)
- Xiao L. Chang
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Helen L. Wu
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Gabriela M. Webb
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Meenakshi Tiwary
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Colette Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Jason S. Reed
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Joseph Hwang
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Courtney Waytashek
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Carla Boyle
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Cleiton Pessoa
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Andrew W. Sylwester
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - David Morrow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Karina Belica
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Miranda Fischer
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, United States
| | | | | | - Rachele M. Bochart
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, United States
| | | | - Scott G. Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Jonah B. Sacha
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
9
|
Tammam SN, El Safy S, Ramadan S, Arjune S, Krakor E, Mathur S. Repurpose but also (nano)-reformulate! The potential role of nanomedicine in the battle against SARS-CoV2. J Control Release 2021; 337:258-284. [PMID: 34293319 PMCID: PMC8289726 DOI: 10.1016/j.jconrel.2021.07.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
The coronavirus disease-19 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has taken the world by surprise. To date, a worldwide approved treatment remains lacking and hence in the context of rapid viral spread and the growing need for rapid action, drug repurposing has emerged as one of the frontline strategies in the battle against SARS-CoV2. Repurposed drugs currently being evaluated against COVID-19 either tackle the replication and spread of SARS-CoV2 or they aim at controlling hyper-inflammation and the rampaged immune response in severe disease. In both cases, the target for such drugs resides in the lungs, at least during the period where treatment could still provide substantial clinical benefit to the patient. Yet, most of these drugs are administered systemically, questioning the percentage of administered drug that actually reaches the lung and as a consequence, the distribution of the remainder of the dose to off target sites. Inhalation therapy should allow higher concentrations of the drug in the lungs and lower concentrations systemically, hence providing a stronger, more localized action, with reduced adverse effects. Therefore, the nano-reformulation of the repurposed drugs for inhalation is a promising approach for targeted drug delivery to lungs. In this review, we critically analyze, what nanomedicine could and ought to do in the battle against SARS-CoV2. We start by a brief description of SARS-CoV2 structure and pathogenicity and move on to discuss the current limitations of repurposed antiviral and immune-modulating drugs that are being clinically investigated against COVID-19. This account focuses on how nanomedicine could address limitations of current therapeutics, enhancing the efficacy, specificity and safety of such drugs. With the appearance of new variants of SARS-CoV2 and the potential implication on the efficacy of vaccines and diagnostics, the presence of an effective therapeutic solution is inevitable and could be potentially achieved via nano-reformulation. The presence of an inhaled nano-platform capable of delivering antiviral or immunomodulatory drugs should be available as part of the repertoire in the fight against current and future outbreaks.
Collapse
Affiliation(s)
- Salma N Tammam
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo (GUC), 11835 Cairo, Egypt.
| | - Sara El Safy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo (GUC), 11835 Cairo, Egypt
| | - Shahenda Ramadan
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo (GUC), 11835 Cairo, Egypt
| | - Sita Arjune
- Institute of Biochemistry, Department of Chemistry, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Eva Krakor
- Institute of Inorganic Chemistry, Department of Chemistry, , University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| | - Sanjay Mathur
- Institute of Inorganic Chemistry, Department of Chemistry, , University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| |
Collapse
|
10
|
Chang XL, Webb GM, Wu HL, Greene JM, Abdulhaqq S, Bateman KB, Reed JS, Pessoa C, Weber WC, Maier N, Chew GM, Gilbride RM, Gao L, Agnor R, Giobbi T, Torgerson J, Siess D, Burnett N, Fischer M, Shiel O, Moats C, Patterson B, Dhody K, Kelly S, Pourhassan N, Magnani DM, Smedley J, Bimber BN, Haigwood NL, Hansen SG, Brown TR, Ndhlovu LC, Sacha JB. Antibody-based CCR5 blockade protects Macaques from mucosal SHIV transmission. Nat Commun 2021; 12:3343. [PMID: 34099693 PMCID: PMC8184841 DOI: 10.1038/s41467-021-23697-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/11/2021] [Indexed: 11/29/2022] Open
Abstract
In the absence of a prophylactic vaccine, the use of antiretroviral therapy (ART) as pre-exposure prophylaxis (PrEP) to prevent HIV acquisition by uninfected individuals is a promising approach to slowing the epidemic, but its efficacy is hampered by incomplete patient adherence and ART-resistant variants. Here, we report that competitive inhibition of HIV Env-CCR5 binding via the CCR5-specific antibody Leronlimab protects rhesus macaques against infection following repeated intrarectal challenges of CCR5-tropic SHIVSF162P3. Injection of Leronlimab weekly at 10 mg/kg provides significant but partial protection, while biweekly 50 mg/kg provides complete protection from SHIV acquisition. Tissue biopsies from protected macaques post challenge show complete CCR5 receptor occupancy and an absence of viral nucleic acids. After Leronlimab washout, protected macaques remain aviremic, and adoptive transfer of hematologic cells into naïve macaques does not transmit viral infection. These data identify CCR5 blockade with Leronlimab as a promising approach to HIV prophylaxis and support initiation of clinical trials.
Collapse
Affiliation(s)
- Xiao L Chang
- Vaccine & Gene Therapy Institute, Portland, OR, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Gabriela M Webb
- Vaccine & Gene Therapy Institute, Portland, OR, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Helen L Wu
- Vaccine & Gene Therapy Institute, Portland, OR, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | - Jason S Reed
- Vaccine & Gene Therapy Institute, Portland, OR, USA
| | | | | | | | | | | | - Lina Gao
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Rebecca Agnor
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Travis Giobbi
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey Torgerson
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Don Siess
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Nicole Burnett
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Miranda Fischer
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Oriene Shiel
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Cassandra Moats
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | - Kush Dhody
- Amarex Clinical Research LLC, Germantown, MD, USA
| | | | | | - Diogo M Magnani
- MassBiologics of the University of Massachusetts Medical School, Boston, MA, USA
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Benjamin N Bimber
- Vaccine & Gene Therapy Institute, Portland, OR, USA
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | - Lishomwa C Ndhlovu
- Department of Medicine, Division of Infectious Disease, Weill Cornell Medicine, New York, NY, USA.
| | - Jonah B Sacha
- Vaccine & Gene Therapy Institute, Portland, OR, USA.
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
11
|
Patterson BK, Seethamraju H, Dhody K, Corley MJ, Kazempour K, Lalezari J, Pang APS, Sugai C, Mahyari E, Francisco EB, Pise A, Rodrigues H, Wu HL, Webb GM, Park BS, Kelly S, Pourhassan N, Lelic A, Kdouh L, Herrera M, Hall E, Bimber BN, Plassmeyer M, Gupta R, Alpan O, O'Halloran JA, Mudd PA, Akalin E, Ndhlovu LC, Sacha JB. CCR5 inhibition in critical COVID-19 patients decreases inflammatory cytokines, increases CD8 T-cells, and decreases SARS-CoV2 RNA in plasma by day 14. Int J Infect Dis 2021; 103:25-32. [PMID: 33186704 PMCID: PMC7654230 DOI: 10.1016/j.ijid.2020.10.101] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now a global pandemic. Emerging results indicate a dysregulated immune response. Given the role of CCR5 in immune cell migration and inflammation, we investigated the impact of CCR5 blockade via the CCR5-specific antibody leronlimab on clinical, immunological, and virological parameters in severe COVID-19 patients. METHODS In March 2020, 10 terminally ill, critical COVID-19 patients received two doses of leronlimab via individual emergency use indication. We analyzed changes in clinical presentation, immune cell populations, inflammation, as well as SARS-CoV-2 plasma viremia before and 14 days after treatment. RESULTS Over the 14-day study period, six patients survived, two were extubated, and one discharged. We observed complete CCR5 receptor occupancy in all donors by day 7. Compared with the baseline, we observed a concomitant statistically significant reduction in plasma IL-6, restoration of the CD4/CD8 ratio, and resolution of SARS-CoV2 plasma viremia (pVL). Furthermore, the increase in the CD8 percentage was inversely correlated with the reduction in pVL (r = -0.77, p = 0.0013). CONCLUSIONS Our study design precludes clinical efficacy inferences but the results implicate CCR5 as a therapeutic target for COVID-19 and they form the basis for ongoing randomized clinical trials.
Collapse
Affiliation(s)
| | | | - Kush Dhody
- Amarex Clinical Research LLC, Germantown, MD, USA
| | - Michael J Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Alina P S Pang
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Eisa Mahyari
- Vaccine & Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | - Helen L Wu
- Vaccine & Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Gabriela M Webb
- Vaccine & Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Byung S Park
- Vaccine & Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | | | | | - Benjamin N Bimber
- Vaccine & Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | - Raavi Gupta
- State University of New York-University Hospital of Brooklyn, NY, USA
| | | | - Jane A O'Halloran
- Division of Infectious Diseases, Department of Internal Medicine, USA
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jonah B Sacha
- Vaccine & Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
12
|
Abstract
Even after more than 30 years since its discovery, there is no cure for HIV-1 infection. Combination antiretroviral therapy (cART) is currently the only HIV-1 infection management option in clinics. Despite its success in suppressing viral replication and converting HIV-1 from a lethal infection to a chronic and manageable disease, cART treatment is life long and long-term use can result in major drawbacks such as high cost, multiple side effects, and an increase in the development of multidrug-resistant escape mutants. Recently, antibody-based anti-HIV-1 treatment has emerged as a potential alternative therapeutic modality for HIV-1 treatment and cure strategies. These antibody-based anti-HIV-1 treatments comprising either receptor-targeting antibodies or broad neutralizing antibodies (bNAbs) are currently being developed and evaluated in clinical trials. These antibodies have demonstrated potent antiviral effects against multiple strains of HIV-1, and shown promise for prevention, maintenance, and prolonged remission of HIV-1 infection. This review gives an update on the current status of these antibody-based treatments for HIV-1, discusses their mechanism of action and the challenges in developing them, providing insight for their development as novel clinical therapies against HIV-1 infection.
Collapse
Affiliation(s)
- Wanwisa Promsote
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan E DeMouth
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cassandra G Almasri
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
13
|
Kichloo A, Albosta M, Kumar A, Aljadah M, Mohamed M, El-Amir Z, Wani F, Jamal S, Singh J, Kichloo A. Emerging therapeutics in the management of COVID-19. World J Virol 2021; 10:1-29. [PMID: 33585175 PMCID: PMC7852573 DOI: 10.5501/wjv.v10.i1.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (coronavirus disease 2019, COVID-19) pandemic has placed a tremendous burden on healthcare systems globally. Therapeutics for treatment of the virus are extremely inconsistent due to the lack of time evaluating drug efficacy in clinical trials. Currently, there is a deficiency of published literature that comprehensively discusses all therapeutics being considered for the treatment of COVID-19. A review of the literature was performed for articles related to therapeutics and clinical trials in the context of the current COVID-19 pandemic. We used PubMed, Google Scholar, and Clinicaltrials.gov to search for articles relative to the topic of interest. We used the following keywords: "COVID-19", "therapeutics", "clinical trials", "treatment", "FDA", "ICU", "mortality", and "management". In addition, searches through the references of retrieved articles was also performed. In this paper, we have elaborated on the therapeutic strategies that have been hypothesized or trialed to-date, the mechanism of action of each therapeutic, the clinical trials finished or in-process that support the use of each therapeutic, and the adverse effects associated with each therapeutic. Currently, there is no treatment that has been proven to provide significant benefit in reducing morbidity and mortality. There are many clinical trials for numerous different therapeutic agents currently underway. By looking back and measuring successful strategies from previous pandemics in addition to carrying out ongoing research, we provide ourselves with the greatest opportunity to find treatments that are beneficial.
Collapse
Affiliation(s)
- Asim Kichloo
- Department of Internal Medicine, Samaritan Medical Center, Watertown, NY 13601, United States
| | - Michael Albosta
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48602, United States
| | - Akshay Kumar
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Michael Aljadah
- Deparment of Internal Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Mohamed Mohamed
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48602, United States
| | - Zain El-Amir
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48602, United States
| | - Farah Wani
- Department of Family Medicine, Samaritan Medical Center, Watertown, NY 13601, United States
| | - Shakeel Jamal
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48602, United States
| | - Jagmeet Singh
- Department of Transplant Nephrology, Geisinger Commonwealth School of Medicine, Sayre, PA 18510, United States
| | - Akif Kichloo
- Department of Anesthesiology and Critical Care, Saraswathi Institue of Medical Sciences, Uttar Pradesh 245304, India
| |
Collapse
|
14
|
Agresti N, Lalezari JP, Amodeo PP, Mody K, Mosher SF, Seethamraju H, Kelly SA, Pourhassan NZ, Sudduth CD, Bovinet C, ElSharkawi AE, Patterson BK, Stephen R, Sacha JB, Wu HL, Gross SA, Dhody K. Disruption of CCR5 signaling to treat COVID-19-associated cytokine storm: Case series of four critically ill patients treated with leronlimab. J Transl Autoimmun 2021; 4:100083. [PMID: 33521616 PMCID: PMC7823045 DOI: 10.1016/j.jtauto.2021.100083] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/25/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is associated with considerable morbidity and mortality. The number of confirmed cases of infection with SARS-CoV-2, the virus causing COVID-19 continues to escalate with over 70 million confirmed cases and over 1.6 million confirmed deaths. Severe-to-critical COVID-19 is associated with a dysregulated host immune response to the virus, which is thought to lead to pathogenic immune dysregulation and end-organ damage. Presently few effective treatment options are available to treat COVID-19. Leronlimab is a humanized IgG4, kappa monoclonal antibody that blocks C–C chemokine receptor type 5 (CCR5). It has been shown that in patients with severe COVID-19 treatment with leronlimab reduces elevated plasma IL-6 and chemokine ligand 5 (CCL5), and normalized CD4/CD8 ratios. We administered leronlimab to 4 critically ill COVID-19 patients in intensive care. All 4 of these patients improved clinically as measured by vasopressor support, and discontinuation of hemodialysis and mechanical ventilation. Following administration of leronlimab there was a statistically significant decrease in IL-6 observed in patient A (p=0.034) from day 0–7 and patient D (p=0.027) from day 0–14. This corresponds to restoration of the immune function as measured by CD4+/CD8+ T cell ratio. Although two of the patients went on to survive the other two subsequently died of surgical complications after an initial recovery from SARS-CoV-2 infection. Leronlimab is a monoclonal antibody in clinical trials to treat the cytokine storm. Critically ill patients received leronlimab through compassionate use and had remarkable recoveries measured objectively. The CCR5 receptor is important in recruiting inflammatory cells mainly T cells and macrophages. Leronlimab disrupted this signal and may have been responsible for restoration of the immune system, improved survival and decrease in IL-6.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- ALT, alanine aminotransferase
- ARDS, acute respiratory distress syndrome
- AST, aspartate aminotransferase
- Acute respiratory distress syndrome (ARDS)
- BID, bis in die (twice a day)
- CCL2, chemokine C–C motif ligand 2
- CCL3, chemokine C–C motif ligand 3
- CCL4, chemokine C–C motif ligand 4
- CCL5, chemokine C–C motif ligand 5
- CCR1, C–C chemokine receptor type 1
- CCR5, C–C chemokine receptor type 5
- CDC, Centers for Disease Control
- CK, creatine kinase
- COPD, chronic obstructive pulmonary disease
- COVID-19, coronavirus disease 2019
- CRP, C-reactive protein
- CXCL10, chemokine C-X-C motif ligand 10
- CXCL2, chemokine C-X-C motif ligand 2
- Coronavirus disease 2019 (COVID-19)
- DPP4, dipeptidyl peptidase-4
- DVT, deep vein thrombosis
- EDTA, ethylenediaminetetraacetic acid
- FDA, Food and Drug Administration
- Fi02, fraction of inspired oxygen, IgG4
- Hydroxychloroquine, HLH
- Leronlimab (PRO 140)
- Middle East respiratory syndrome coronavirus, MIG
- National Early Warning Score, NK
- RO, receptor occupancy
- RT–PCR, reverse transcriptase polymerase chain reaction
- SARS-CoV, severe acute respiratory syndrome coronavirus
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- T-reg RO, regulatory T cells – receptor occupancy
- TGF- α, transforming growth factor alpha
- TNF-α, tumor necrosis factor alpha
- TNF-β, tumor necrosis factor beta
- Tregs, regulatory T cells
- VEGF-A, vascular endothelial growth factor A
- WBC, white blood cell
- WHO, World Health Organization
- eIND, emergency investigational new drug application
- hemophagocytic lymphohistiocytosis, HTN
- hypertension, ICU
- immunoglobulin G4, HCQ
- intensive care unit, IL-1β
- interferon gamma, IL-6
- interferon gamma-inducible protein (IP) 10 or CXCL10, LOA
- interleukin 1 beta, IFN-ƴ
- interleukin 6, IP-10
- letter of authorization, MCP
- macrophage Inflammatory Proteins 1-alpha, MIP-1β
- macrophage Inflammatory Proteins 1-beta, N/A
- macrophage colony stimulating factor, MDC (CCL22)
- macrophage colony-stimulating factor encoded by the CCL22 gene, MERS-CoV
- monocyte chemoattractant protein, M-CSF
- monokine induced by IFN-γ (interferon gamma), MIP-1α
- natural killer, OSA
- not applicable, NEWS2
- obstructive sleep apnea, PDGF-AA
- per os (taken by mouth), RANTES
- platelet-derived growth factor AA, PDGF-AA/BB
- platelet-derived growth factor AA/BB, PEEP
- positive end-expiratory pressure, PNA
- pulmonary nodular amyloidosis, po
- regulated on activation, normal T expressed and secreted (also known as CCL5)
Collapse
Affiliation(s)
- Nicholas Agresti
- Southeast Georgia Health System, 2415 Parkwood Drive, Brunswick, GA, 31520, USA
| | | | - Phillip P Amodeo
- Southeast Georgia Health System, 2415 Parkwood Drive, Brunswick, GA, 31520, USA
| | - Kabir Mody
- Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 3222, USA
| | - Steven F Mosher
- Southeast Georgia Health System, 2415 Parkwood Drive, Brunswick, GA, 31520, USA
| | - Harish Seethamraju
- Montefiore Medical Center, Albert Einstein University, 1695A Eastchester Rd, Bronx, NY, 10467, USA
| | - Scott A Kelly
- CytoDyn, 1111 Main Street, Suite 660 Vancouver, WA, 98660, USA
| | | | - C David Sudduth
- Southeast Georgia Health System, 2415 Parkwood Drive, Brunswick, GA, 31520, USA
| | - Christopher Bovinet
- Spine Center of Southeast Georgia, 1111 Glynco Pkwy Ste 300, Brunswick, GA, 31525, USA
| | - Ahmed E ElSharkawi
- Southeast Georgia Health System, 2415 Parkwood Drive, Brunswick, GA, 31520, USA
| | | | - Reejis Stephen
- Southeast Georgia Health System, 2415 Parkwood Drive, Brunswick, GA, 31520, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, 505 N.W. 185th Avenue, Beaverton, OR, 97006, USA
| | - Helen L Wu
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, 505 N.W. 185th Avenue, Beaverton, OR, 97006, USA
| | - Seth A Gross
- NYU Langone Gastroenterology Associates, 240 East 38th Street, 23rd Floor New York, NY, 10016, USA
| | - Kush Dhody
- Amarex Clinical Research, 20201 Century Blvd, Germantown, MD, 20874, USA
| |
Collapse
|
15
|
Borcherding N, Jethava Y, Vikas P. Repurposing Anti-Cancer Drugs for COVID-19 Treatment. Drug Des Devel Ther 2020; 14:5045-5058. [PMID: 33239864 PMCID: PMC7680713 DOI: 10.2147/dddt.s282252] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic has caused catastrophic damage to human life across the globe along with social and financial hardships. According to the Johns Hopkins University Coronavirus Resource Center, more than 41.3 million people worldwide have been infected, and more than 1,133,000 people have died as of October 22, 2020. At present, there is no available vaccine and a scarcity of efficacious therapies. However, there is tremendous ongoing effort towards identifying effective drugs and developing novel vaccines. Early data from Adaptive COVID-19 Treatment Trials (ACTT) sponsored by the National Institute of Allergy and Infectious Diseases (NIAID) and compassionate use study have shown promise for remdesivir, leading to emergency authorization by the Food and Drug Administration (FDA) for treatment of hospitalized COVID-19 patients. However, several randomized studies have now shown no benefit or increased adverse events associated with remdesivir treatment. Drug development is a time-intensive process and requires extensive safety and efficacy evaluations. In contrast, drug repurposing is a time-saving and cost-effective drug discovery strategy geared towards using existing drugs instead of de novo drug discovery. Treatments for cancer and COVID-19 often have similar goals of controlling inflammation, inhibiting cell division, and modulating the host microenvironment to control the disease. In this review, we focus on anti-cancer drugs that can potentially be repurposed for COVID-19 and are currently being tested in clinical trials.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA, USA
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA, USA
| | - Yogesh Jethava
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, IA, USA
| | - Praveen Vikas
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, IA, USA
| |
Collapse
|
16
|
Kufel WD. Antibody-based strategies in HIV therapy. Int J Antimicrob Agents 2020; 56:106186. [PMID: 33045349 PMCID: PMC7546180 DOI: 10.1016/j.ijantimicag.2020.106186] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/22/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Antibody therapies offer a unique mechanism of action, decreased resistance and improved safety. Ibalizumab is a CD4 post-attachment inhibitor and leronlimab is a CCR5 inhibitor. Ibalizumab is approved for multi-drug-resistant human immunodeficiency virus (MDR HIV) treatment in combination with antiretroviral therapy. Leronlimab is being studied for treatment of MDR HIV and as maintenance monotherapy.
Antibody-based strategies have been introduced for a number of disease states, but represent a novel approach in the management of human immunodeficiency virus (HIV). Ibalizumab and leronlimab are monoclonal antibodies with unique mechanisms as a CD4-directed post-attachment inhibitor and a C-C chemokine receptor type 5-directed inhibitor, respectively. These antibody-based strategies are generally well tolerated, have a favourable pharmacokinetic profile allowing for less-frequent dosing, and have a high barrier to resistance. Ibalizumab is currently approved by the US Food and Drug Administration (US FDA) for management of multi-drug-resistant (MDR) HIV infection in patients who are failing their current regimens. Clinical data demonstrated impressive antiretroviral activity with ibalizumab among a complex HIV population in combination with an optimized background regimen, where limited therapeutic options exist. To date, leronlimab has not been granted approval by the US FDA, but has been designated fast-track status. Leronlimab is being studied as a maintenance monotherapy agent in virologically suppressed patients, as well as for treatment of MDR HIV infection in patients who are failing their current regimens. Currently available data in both of these potential areas appear promising for leronlimab. The mechanism of action, pharmacokinetic profile, efficacy and safety of these novel antibody-based strategies represent an advance in the management of HIV. Future studies and post-marketing experience will further determine longer-term clinical efficacy, safety and resistance data for ibalizumab and leronlimab.
Collapse
Affiliation(s)
- Wesley D Kufel
- Binghamton University School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA; State University of New York Upstate Medical University, Syracuse, NY, USA; State University of New York Upstate University Hospital, Syracuse, NY, USA.
| |
Collapse
|
17
|
Qi B, Fang Q, Liu S, Hou W, Li J, Huang Y, Shi J. Advances of CCR5 antagonists: From small molecules to macromolecules. Eur J Med Chem 2020; 208:112819. [PMID: 32947226 DOI: 10.1016/j.ejmech.2020.112819] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 02/08/2023]
Abstract
C-C chemokine receptor 5(CCR5) is a cell membrane protein from G protein-coupled receptors (GPCR) family, which is an important modulator for leukocyte activation and mobilization. In the 1980s, several reports suggest that lack of the HIV-1 co-receptor, the chemokine receptor CCR5, offers protection against HIV infection. Later, it was shown that CCR5 was confirmed to be the most common co-receptor for the HIV-1 virus R5 strain. In recent years, many studies have shown that CCR5 is closely related to the development of various cancers and inflammations to facilitate the discovery of CCR5 antagonists. There are many types of CCR5 antagonists, mainly including chemokine derivatives, non-peptide small molecule compounds, monoclonal antibodies, and peptide compounds. This review focus on the recent research processes and pharmacological effects of CCR5 antagonists such as Maraviroc, TAK-779 and PRO 140. After focusing on the therapeutic effect of CCR5 antagonists on AIDS, it also discusses the therapeutic prospect of CCR5 in other diseases such as inflammation and tumor.
Collapse
Affiliation(s)
- Baowen Qi
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, China; College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Qiang Fang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Shiyuan Liu
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Wenli Hou
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, China
| | - Jian Li
- Department of Pharmacy, West China Hospital Sichuan University, Chengdu, 610041, China.
| | - Yingchun Huang
- Beijing Key Laboratory of Biomass Waste Resource Utilization, College of Biochemical Engineering, Beijing Union University, Beijing, 100023, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
18
|
Patterson BK, Seethamraju H, Dhody K, Corley MJ, Kazempour K, Lalezari JP, Pang AP, Sugai C, Francisco EB, Pise A, Rodrigues H, Ryou M, Wu HL, Webb GM, Park BS, Kelly S, Pourhassan N, Lelic A, Kdouh L, Herrera M, Hall E, Aklin E, Ndhlovu L, Sacha JB. Disruption of the CCL5/RANTES-CCR5 Pathway Restores Immune Homeostasis and Reduces Plasma Viral Load in Critical COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.05.02.20084673. [PMID: 32511656 PMCID: PMC7277012 DOI: 10.1101/2020.05.02.20084673] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is now pandemic with nearly three million cases reported to date. Although the majority of COVID-19 patients experience only mild or moderate symptoms, a subset will progress to severe disease with pneumonia and acute respiratory distress syndrome (ARDS) requiring mechanical ventilation. Emerging results indicate a dysregulated immune response characterized by runaway inflammation, including cytokine release syndrome (CRS), as the major driver of pathology in severe COVID-19. With no treatments currently approved for COVID-19, therapeutics to prevent or treat the excessive inflammation in severe disease caused by SARS-CoV-2 infection are urgently needed. Here, in 10 terminally-ill, critical COVID-19 patients we report profound elevation of plasma IL-6 and CCL5 (RANTES), decreased CD8+ T cell levels, and SARS-CoV-2 plasma viremia. Following compassionate care treatment with the CCR5 blocking antibody leronlimab, we observed complete CCR5 receptor occupancy on macrophage and T cells, rapid reduction of plasma IL-6, restoration of the CD4/CD8 ratio, and a significant decrease in SARS-CoV-2 plasma viremia. Consistent with reduction of plasma IL-6, single-cell RNA-sequencing revealed declines in transcriptomic myeloid cell clusters expressing IL-6 and interferon-related genes. These results demonstrate a novel approach to resolving unchecked inflammation, restoring immunologic deficiencies, and reducing SARS-CoV-2 plasma viral load via disruption of the CCL5-CCR5 axis, and support randomized clinical trials to assess clinical efficacy of leronlimab-mediated inhibition of CCR5 for COVID-19.
Collapse
|
19
|
Abstract
HIV is one of the most devastating viral infections the world has ever encountered. Ever since HIV was first identified in the 1980s, it has claimed millions of lives worldwide. There has been tremendous research and development in the diagnosis, prevention and treatment of HIV. Small molecules have been shown to reduce the virus to nondetectable level in human plasma, however, there are reservoirs of latent virus that reemerge if antiretroviral therapy is stopped. There is no vaccine to prevent or cure HIV. A significant amount of research has been reported in the literature regarding antibodies for CCR5, a HIV entry host receptor. This report describes the role of CCR5 antibody in HIV prevention/treatment and how antibody-conjugated nanoparticles could be a future strategy with the potential to effectively eradicate the virus from the human system.
Collapse
|
20
|
Flexner C. Modern Human Immunodeficiency Virus Therapy: Progress and Prospects. Clin Pharmacol Ther 2019; 105:61-70. [PMID: 30411787 DOI: 10.1002/cpt.1284] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/31/2018] [Indexed: 01/05/2023]
Abstract
Safe and effective lifelong treatment to control human immunodeficiency virus (HIV) infection is one of the greatest scientific and public health achievements of the past century. The majority of infected individuals able to maintain a daily oral regimen now have a normal or near-normal life expectancy. More than 30 approved drugs and dozens of formulations have produced thousands of possible drug combinations used clinically in the past, but today most patients receive only a handful of high priority and rigorously tested regimens. Unique features of antiretroviral therapy include the need for lifelong treatment to control virus replication and the possibility of rapid emergence of permanent drug resistance if these agents are not properly used. Although three-drug combination oral regimens have radically altered the course of this epidemic, the future will include long-acting injectable and implantable drugs and devices to treat and prevent infection.
Collapse
Affiliation(s)
- Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, Departments of Medicine, Pharmacology and Molecular Sciences, and International Health, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Wang CY, Wong WW, Tsai HC, Chen YH, Kuo BS, Lynn S, Blazkova J, Clarridge KE, Su HW, Lin CY, Tseng FC, Lai A, Yang FH, Lin CH, Tseng W, Lin HY, Finstad CL, Wong-Staal F, Hanson CV, Chun TW, Liao MJ. Effect of Anti-CD4 Antibody UB-421 on HIV-1 Rebound after Treatment Interruption. N Engl J Med 2019; 380:1535-1545. [PMID: 30995373 DOI: 10.1056/nejmoa1802264] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Administration of a single broadly neutralizing human immunodeficiency virus (HIV)-specific antibody to HIV-infected persons leads to the development of antibody-resistant virus in the absence of antiretroviral therapy (ART). It is possible that monotherapy with UB-421, an antibody that blocks the virus-binding site on human CD4+ T cells, could induce sustained virologic suppression without induction of resistance in HIV-infected persons after analytic treatment interruption. METHODS We conducted a nonrandomized, open-label, phase 2 clinical study evaluating the safety, pharmacokinetics, and antiviral activity of UB-421 monotherapy in HIV-infected persons undergoing analytic treatment interruption. All the participants had undetectable plasma viremia (<20 copies of HIV RNA per milliliter) at the screening visit. After discontinuation of ART, participants received eight intravenous infusions of UB-421, at a dose of either 10 mg per kilogram of body weight every week (Cohort 1) or 25 mg per kilogram every 2 weeks (Cohort 2). The primary outcome was the time to viral rebound (≥400 copies per milliliter). RESULTS A total of 29 participants were enrolled, 14 in Cohort 1 and 15 in Cohort 2. Administration of UB-421 maintained virologic suppression (<20 copies per milliliter) in all the participants (94.5% of measurements at study visits 2 through 9) during analytic treatment interruption, with intermittent viral blips (range, 21 to 142 copies per milliliter) observed in 8 participants (28%). No study participants had plasma viral rebound to more than 400 copies per milliliter. CD4+ T-cell counts remained stable throughout the duration of the study. Rash, mostly of grade 1, was a common and transient adverse event; one participant discontinued the study drug owing to a rash. A decrease in the population of CD4+ regulatory T cells was observed during UB-421 monotherapy. CONCLUSIONS UB-421 maintained virologic suppression (during the 8 to 16 weeks of study) in participants in the absence of ART. One participant discontinued therapy owing to a rash. (Funded by United Biomedical and others; ClinicalTrials.gov number, NCT02369146.).
Collapse
Affiliation(s)
- Chang-Yi Wang
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Wing-Wai Wong
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Hung-Chin Tsai
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Yen-Hsu Chen
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Be-Sheng Kuo
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Shugene Lynn
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Jana Blazkova
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Katherine E Clarridge
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Hsiao-Wen Su
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Chia-Ying Lin
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Fan-Chen Tseng
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Annie Lai
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Fu-Hung Yang
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Chen-Han Lin
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - William Tseng
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Hsiao-Yi Lin
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Connie L Finstad
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Flossie Wong-Staal
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Carl V Hanson
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Tae-Wook Chun
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| | - Mei-June Liao
- From United Biomedical, Hauppauge, NY (C.-Y.W., B.-S.K., C.L.F., F.W.-S.); United Biomedical Asia (C.-Y.W., S.L.) and United BioPharma (C.-Y.W., B.-S.K., H.-W.S., C.-Y.L., F.-C.T., A.L., F.-H.Y., C.-H.L., W.T., M.-J.L.), Hsinchu, Taipei Veterans General Hospital, Taipei (W.-W.W., H.-Y.L.), and Kaohsiung Veterans General Hospital (H.-C.T.) and Kaohsiung Medical University Hospital (Y.-H.C.), Kaohsiung - all in Taiwan; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.B., K.E.C., T.-W.C.); and the California Department of Public Health, Richmond (C.V.H.)
| |
Collapse
|
22
|
Venanzi Rullo E, Ceccarelli M, Condorelli F, Facciolà A, Visalli G, D'Aleo F, Paolucci I, Cacopardo B, Pinzone MR, Di Rosa M, Nunnari G, Pellicanò GF. Investigational drugs in HIV: Pros and cons of entry and fusion inhibitors (Review). Mol Med Rep 2019; 19:1987-1995. [PMID: 30628713 DOI: 10.3892/mmr.2019.9840] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 11/29/2018] [Indexed: 11/06/2022] Open
Abstract
Despite the profound changes and improvements reached in the field of HIV treatment, tolerability and adherence to highly active antiretroviral therapy remains a challenge. Furthermore, multi-experienced patients could take advantage of drugs with different mechanisms of action to combat the spread of resistance to actual therapy. For these reasons identification of new HIV drugs is crucial. Among all the molecules that at present are under investigation, entry and fusion inhibitors pose an interesting class owing to their peculiar characteristics, including prevention of entry of the virus into the human cells. In this study, we reviewed articles, clinical trials, and conference communications about all the drugs under investigation belonging to the class of entry and fusion inhibitors that are at least in phase I clinical trials.
Collapse
Affiliation(s)
- Emmanuele Venanzi Rullo
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, I-90124 Messina, Italy
| | - Manuela Ceccarelli
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, I-90124 Messina, Italy
| | - Fabrizio Condorelli
- Department of Pharmacological Sciences, University of Eastern Piedmont 'A. Avogadro', I-13100 Novara, Italy
| | - Alessio Facciolà
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, I-90124 Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, I-90124 Messina, Italy
| | - Francesco D'Aleo
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, I-90124 Messina, Italy
| | - Ivana Paolucci
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, I-90124 Messina, Italy
| | - Bruno Cacopardo
- Department of Clinical and Experimental Medicine, University of Catania, I-95123 Catania, Italy
| | - Marilia Rita Pinzone
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michele Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, University of Catania, I-95123 Catania, Italy
| | - Giuseppe Nunnari
- Department of Clinical and Experimental Medicine, Unit of Infectious Diseases, University of Messina, I-90124 Messina, Italy
| | - Giovanni F Pellicanò
- Department of Human Pathology of the Adult and the Developmental Age 'G. Barresi', Unit of Infectious Diseases, University of Messina, I-98122 Messina, Italy
| |
Collapse
|
23
|
Creating demand for long-acting formulations for the treatment and prevention of HIV, tuberculosis, and viral hepatitis. Curr Opin HIV AIDS 2019; 14:13-20. [PMID: 30394948 DOI: 10.1097/coh.0000000000000510] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Long-acting parenteral drug delivery is an established and widely accepted solution to the problem of poor adherence when daily oral medications are used to treat or prevent chronic medical conditions. Poor adherence to oral formulations remains a major barrier to successfully treating or preventing HIV, tuberculosis (TB), and viral hepatitis. The uptake of long-acting formulations developed for these infections is uncertain, despite their promise. This review addresses the current state of development of long-acting and extended-release approaches to HIV, TB, and viral hepatitis in the context of creating market demand for such products. RECENT FINDINGS Two nanoformulated long-acting injectable antiretroviral compounds, cabotegravir and rilpivirine, recently completed Phase 2 clinical trials demonstrating safety, tolerability, and antiretroviral activity, and should be available in high income countries following completion of ongoing Phase 3 trials. Long-acting polymer implants of the antiretroviral nucleosides tenofovir alafenamide and 4'-ethynyl-2-fluoro-2'-deoxyadenosine are being tested in animals and should soon enter human studies; tenofovir alafenamide also has activity against hepatitis B virus. Long-acting versions of several broadly neutralizing monoclonal antibodies are in advanced clinical trials for HIV prevention and treatment. Long-acting formulations for TB are in preclinical development. There is no evidence that comparable formulations for viral hepatitis are being developed at present. SUMMARY Long-acting and extended release formulations are promising approaches to the treatment and prevention of common infectious diseases, but their availability is limited at this time. These products hold great promise for the global control of important human infections. Based on experience with other diseases, it is likely that their use will become more widespread if they are cost competitive with generic oral formulations.
Collapse
|
24
|
Falkenhagen A, Joshi S. Genetic Strategies for HIV Treatment and Prevention. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:514-533. [PMID: 30388625 PMCID: PMC6205348 DOI: 10.1016/j.omtn.2018.09.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 08/28/2018] [Accepted: 09/02/2018] [Indexed: 01/02/2023]
Abstract
Conventional HIV gene therapy approaches are based on engineering HIV target cells that are non-permissive to viral replication. However, expansion of gene-modified HIV target cells has been limited in patients. Alternative genetic strategies focus on generating gene-modified producer cells that secrete antiviral proteins (AVPs). The secreted AVPs interfere with HIV entry, and, therefore, they extend the protection against infection to unmodified HIV target cells. Since any cell type can potentially secrete AVPs, hematopoietic and non-hematopoietic cell lineages can function as producer cells. Secretion of AVPs from non-hematopoietic cells opens the possibility of using a genetic approach for HIV prevention. Another strategy aims at modifying cytotoxic T cells to selectively target and eliminate infected cells. This review provides an overview of the different genetic approaches for HIV treatment and prevention.
Collapse
Affiliation(s)
- Alexander Falkenhagen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sadhna Joshi
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
25
|
Welles HC, Jennewein MF, Mason RD, Narpala S, Wang L, Cheng C, Zhang Y, Todd JP, Lifson JD, Balazs AB, Alter G, McDermott AB, Mascola JR, Roederer M. Vectored delivery of anti-SIV envelope targeting mAb via AAV8 protects rhesus macaques from repeated limiting dose intrarectal swarm SIVsmE660 challenge. PLoS Pathog 2018; 14:e1007395. [PMID: 30517201 PMCID: PMC6296672 DOI: 10.1371/journal.ppat.1007395] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 12/17/2018] [Accepted: 10/10/2018] [Indexed: 01/17/2023] Open
Abstract
Gene based delivery of immunoglobulins promises to safely and durably provide protective immunity to individuals at risk of acquiring infectious diseases such as HIV. We used a rhesus macaque animal model to optimize delivery of naturally-arising, autologous anti-SIV neutralizing antibodies expressed by Adeno-Associated Virus 8 (AAV8) vectors. Vectored transgene expression was confirmed by quantitation of target antibody abundance in serum and mucosal surfaces. We tested the expression achieved at varying doses and numbers of injections. Expression of the transgene reached a saturation at about 2 x 1012 AAV8 genome copies (gc) per needle-injection, a physical limitation that may not scale clinically into human trials. In contrast, expression increased proportionately with the number of injections. In terms of anti-drug immunity, anti-vector antibody responses were universally strong, while those directed against the natural transgene mAb were detected in only 20% of animals. An anti-transgene antibody response was invariably associated with loss of detectable plasma expression of the antibody. Despite having atypical glycosylation profiles, transgenes derived from AAV-directed muscle cell expression retained full functional activity, including mucosal accumulation, in vitro neutralization, and protection against repeated limiting dose SIVsmE660 swarm challenge. Our findings demonstrate feasibility of a gene therapy-based passive immunization strategy against infectious disease, and illustrate the potential for the nonhuman primate model to inform clinical AAV-based approaches to passive immunization. Antibodies are the humoral component of an immune response against an invading pathogen or vaccine immunogen. For challenging vaccine targets, as an alternative to active vaccination to induce the immune system to generate antibodies, current research is exploring the delivery of these proteins to populations at high risk of infection as prophylactics against infectious diseases, like HIV, RSV, and Ebola, amongst others. Passive vaccination via purified protein will require periodic reinjection to retain protective levels in subjects, adding a barrier to large scale coverage. Alternatively, delivery of antibodies using gene therapy may provide a one-time passive vaccination alternative. This strategy comes with its own hurdles, including anti-vector immunity, anti-drug immunity, physical limitations of vector uptake and the need to confirm antibody functionality. To date, many passive vaccinations strategies remain untested in humans. Non-human primate models of infection are frequently useful for predicting the success of vaccine candidates or concepts. Here, we characterize and optimize a rhesus macaque model for the delivery of anti-viral antibodies via the gene therapy vector adeno-associated virus. Lastly, we demonstrate the ability of the mAbs to protect against viral challenge. Our work demonstrates the feasibility and utility of vectored delivery of antibody transgenes in rhesus macaques. We hope this model of antibody delivery may be applied to various disease models in non-human primates and will inform clinical trial design of passive vaccination against infectious diseases.
Collapse
Affiliation(s)
- Hugh C. Welles
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Institute for Biomedical Sciences, The George Washington University, Washington, DC, United States of America
- * E-mail: (HCW); (MR)
| | - Madeleine F. Jennewein
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Rosemarie D. Mason
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sandeep Narpala
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lingshu Wang
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheng Cheng
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yi Zhang
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John-Paul Todd
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory of Cancer Research, Frederick, Maryland, United States of America
| | - Alejandro B. Balazs
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Adrian B. McDermott
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John R. Mascola
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mario Roederer
- Immunotechnology Section, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (HCW); (MR)
| |
Collapse
|
26
|
Abstract
Antiretroviral drugs have revolutionized the treatment and prevention of HIV infection; however, adherence is critical for sustained efficacy. Current HIV treatment consists of three-drug regimens, and current HIV pre-exposure prophylaxis (PrEP) consists of a two-drug regimen; both generally require adherence to once-daily dosing. Long-acting formulations are useful in the treatment and prevention of other conditions (e.g., contraceptives, antipsychotics) and help promote adherence. Newer long-acting formulations of approved and investigational antiretroviral drugs in existing and newer mechanistic classes are under study for HIV treatment and prevention, including some phase III trials. Although long-acting antiretroviral drugs hold promise, some clinical challenges exist, including managing side effects, drug-drug interactions, pregnancy, and long-lasting drug concentrations that could lead to the development of drug resistance. This review aims to summarize currently available information on long-acting antiretroviral drugs for HIV treatment and prevention.
Collapse
Affiliation(s)
- Roy M Gulick
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Charles Flexner
- Divisions of Clinical Pharmacology and Infectious Diseases, Long Acting/Extended Release Antiretroviral Resource Program, School of Medicine and Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21287-5554, USA;
| |
Collapse
|
27
|
Abstract
Infectious disease represent the most significant threat to human health. Significant geologic cataclysmic events have caused the extinction of countless species, but these “Wrath of God” events predate the emergence of Homo sapiens. Pandemic infections have accompanied the rise of human civilization frequently re-occurring leaving a lasting imprint on human history punctuated by profound loss of life. Emerging infections become endemic and are here to stay marking their presence with an annual death toll. Each decade brings a new onslaught of emerging infectious agents. We are surprised again and again but are never prepared. The long-term consequences often remain unrecognized and are always inconvenient including cancer, cardiovascular disease and immune associated diseases that threaten our health. Reliance on clusters of clinical symptoms in the face of diverse and non-descriptive viral infection symptoms is a foolhardy form of crisis management. Viral success is based on rapid replication resulting in large numbers. Single-stranded RNA viruses with their high replication error rate represent a paradigm for resilience.
Collapse
|
28
|
Falkenhagen A, Joshi S. HIV Entry and Its Inhibition by Bifunctional Antiviral Proteins. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 13:347-364. [PMID: 30340139 PMCID: PMC6197789 DOI: 10.1016/j.omtn.2018.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
HIV entry is a highly specific and time-sensitive process that can be divided into receptor binding, coreceptor binding, and membrane fusion. Bifunctional antiviral proteins (bAVPs) exploit the multi-step nature of the HIV entry process by binding to two different extracellular targets. They are generated by expressing a fusion protein containing two entry inhibitors with a flexible linker. The resulting fusion proteins exhibit exceptional neutralization potency and broad cross-clade inhibition. In this review, we summarize the HIV entry process and provide an overview of the design, antiviral potency, and methods of delivery of bAVPs. Additionally, we discuss the advantages and limitations of bAVPs for HIV prevention and treatment.
Collapse
Affiliation(s)
- Alexander Falkenhagen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Sadhna Joshi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E2, Canada.
| |
Collapse
|
29
|
|
30
|
Scott‐Taylor TH, Axinia S, Amin S, Pettengell R. Immunoglobulin G; structure and functional implications of different subclass modifications in initiation and resolution of allergy. Immun Inflamm Dis 2018; 6:13-33. [PMID: 29164823 PMCID: PMC5818455 DOI: 10.1002/iid3.192] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/14/2017] [Accepted: 07/23/2017] [Indexed: 12/24/2022] Open
Abstract
IgE and not IgG is usually associated with allergy. IgE lodged on mast cells in skin or gut and basophils in the blood allows for the prolonged duration of allergy through the persistent expression of high affinity IgE receptors. However, many allergic reactions are not dependent on IgE and are generated in the absence of allergen specific and even total IgE. Instead, IgG plasma cells are involved in induction of, and for much of the pathogenesis of, allergic diseases. The pattern of IgG producing plasma cells in atopic children and the tendency for direct or further class switching to IgE are the principle factors responsible for long-lasting sensitization of mast cells in allergic children. Indirect class switching from IgG producing plasma cells has been shown to be the predominant pathway for production of IgE while a Th2 microenvironment, genetic predisposition, and the concentration and nature of allergens together act on IgG plasma cells in the atopic tendency to undergo further immunoglobulin gene recombination. The seminal involvement of IgG in allergy is further indicated by the principal role of IgG4 in the natural resolution of allergy and as the favourable immunological response to immunotherapy. This paper will look at allergy through the role of different antibodies than IgE and give current knowledge of the nature and role of IgG antibodies in the start, maintenance and resolution of allergy.
Collapse
Affiliation(s)
| | - Stefan‐Claudiu Axinia
- School of Life SciencesLondon Metropolitan University166‐220 Holloway RoadLondon, N7 8DB
| | - Sumeya Amin
- School of Life SciencesLondon Metropolitan University166‐220 Holloway RoadLondon, N7 8DB
| | - Ruth Pettengell
- Department of HaematologySt George's University of LondonCranmer TerraceLondon SW17 0RE
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Poor adherence to oral antiretroviral treatment in a subpopulation of persons with HIV-1 infection interferes with the potential success of the drug regimens in treating the infection. Here, we review long-acting antiretroviral strategies currently in clinical development that could prove useful for the treatment of HIV-1 infection in individuals not succeeding with short-acting oral regimens. RECENT FINDINGS Pharmaceutical nanotechnology has succeeded in creating two novel long-acting injectable antiretroviral compounds, carbotegravir and rilpivirine, which have completed early clinical trials demonstrating the safety, tolerability and prolonged antiretroviral activity. 4'-Ethynyl-2-fluoro-2'-deoxyadenosine (EFdA; MK8591) is a novel nucleoside reverse transcriptase inhibitor in early clinical development as a long-acting orally administered drug and in a long-acting polymer implant. Broadly neutralizing and cell-entry inhibitor monoclonal antibodies have demonstrated potent antiviral activity in early human trials; however, there is substantial baseline resistance. In addition, monotherapy leads to rapid resistance in those with baseline susceptibility. SUMMARY Long-acting antiretroviral chemical compounds and monoclonal antibodies have demonstrated potent anti-HIV activity in the early-stage clinical investigations, and are actively being studied in advanced clinical trials for treatment and prevention. Strategies to manage toxicities and waning drug levels of chemical compounds, as well as primary and secondary resistance to current monoclonal antibodies are important considerations.
Collapse
|
32
|
Iacob SA, Iacob DG. Ibalizumab Targeting CD4 Receptors, An Emerging Molecule in HIV Therapy. Front Microbiol 2017; 8:2323. [PMID: 29230203 PMCID: PMC5711820 DOI: 10.3389/fmicb.2017.02323] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/10/2017] [Indexed: 11/13/2022] Open
Abstract
The HIV infection is responsible for the most devastating global pandemic of the last century. More than 39 million people have died of HIV/AIDS since 1981. The development of the antiretroviral (ARV) treatment begins with the discovery of zidovudine a nucleoside reverse transcriptase inhibitor. This breakthrough was followed by other ARV drug classes and representatives. Presently, HIV treatment employs 27 ARV representatives belonging to five different classes. Despite the proven benefits of ARV treatment and its long-term control of the HIV infection, there is an increasing concern about the numerous adverse effects and resistance to current ARV drugs. Therefore, the new HIV treatment strategies focus on the development of new ARV agents with a high genetic barrier to resistance and low toxicity. Monoclonal antibodies (MAbs) belong to a new drug class with encouraging results in the treatment of cancer, autoimmune disorders and most recently against HIV infection. The advantages of using MAbs for HIV treatment are related to their antiviral effect, lack of toxicity, good resistance profile, additional synergy with other ARV drug classes and ability to restore CD4 T-cell responses. The current article is a short summary of ibalizumab, an anti-CD4 monoclonal antibody that interferes with HIV viral entry. Current studies on ibalizumab have underlined its antiviral potential, minimal adverse effects, and lack of crossed resistance with other ARV agents thus supporting its further therapeutic use in multidrug resistant HIV-infected patients.
Collapse
Affiliation(s)
- Simona A Iacob
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Diana G Iacob
- National Institute for Infectious Diseases "Prof.dr. Matei Bals", Bucharest, Romania
| |
Collapse
|
33
|
Gravatt LAH, Leibrand CR, Patel S, McRae M. New Drugs in the Pipeline for the Treatment of HIV: a Review. Curr Infect Dis Rep 2017; 19:42. [DOI: 10.1007/s11908-017-0601-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
34
|
Discovery and Characterization of a Novel CD4-Binding Adnectin with Potent Anti-HIV Activity. Antimicrob Agents Chemother 2017; 61:AAC.00508-17. [PMID: 28584151 DOI: 10.1128/aac.00508-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/30/2017] [Indexed: 02/08/2023] Open
Abstract
A novel fibronectin-based protein (Adnectin) HIV-1 inhibitor was generated using in vitro selection. This inhibitor binds to human CD4 with a high affinity (3.9 nM) and inhibits viral entry at a step after CD4 engagement and preceding membrane fusion. The progenitor sequence of this novel inhibitor was selected from a library of trillions of Adnectin variants using mRNA display and then further optimized for improved antiviral and physical properties. The final optimized inhibitor exhibited full potency against a panel of 124 envelope (gp160) proteins spanning 11 subtypes, indicating broad-spectrum activity. Resistance profiling studies showed that this inhibitor required 30 passages (151 days) in culture to acquire sufficient resistance to result in viral titer breakthrough. Resistance mapped to the loss of multiple potential N-linked glycosylation sites in gp120, suggesting that inhibition is due to steric hindrance of CD4-binding-induced conformational changes.
Collapse
|
35
|
Opportunities for therapeutic antibodies directed at G-protein-coupled receptors. Nat Rev Drug Discov 2017; 16:787-810. [PMID: 28706220 DOI: 10.1038/nrd.2017.91] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) are activated by a diverse range of ligands, from large proteins and proteases to small peptides, metabolites, neurotransmitters and ions. They are expressed on all cells in the body and have key roles in physiology and homeostasis. As such, GPCRs are one of the most important target classes for therapeutic drug discovery. The development of drugs targeting GPCRs has therapeutic value across a wide range of diseases, including cancer, immune and inflammatory disorders as well as neurological and metabolic diseases. The progress made by targeting GPCRs with antibody-based therapeutics, as well as technical hurdles to overcome, are presented and discussed in this Review. Antibody therapeutics targeting C-C chemokine receptor type 4 (CCR4), CCR5 and calcitonin gene-related peptide (CGRP) are used as illustrative clinical case studies.
Collapse
|
36
|
Hua CK, Ackerman ME. Engineering broadly neutralizing antibodies for HIV prevention and therapy. Adv Drug Deliv Rev 2016; 103:157-173. [PMID: 26827912 DOI: 10.1016/j.addr.2016.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/15/2023]
Abstract
A combination of advances spanning from isolation to delivery of potent HIV-specific antibodies has begun to revolutionize understandings of antibody-mediated antiviral activity. As a result, the set of broadly neutralizing and highly protective antibodies has grown in number, diversity, potency, and breadth of viral recognition and neutralization. These antibodies are now being further enhanced by rational engineering of their anti-HIV activities and coupled to cutting edge gene delivery and strategies to optimize their pharmacokinetics and biodistribution. As a result, the prospects for clinical use of HIV-specific antibodies to treat, clear, and prevent HIV infection are gaining momentum. Here we discuss the diverse methods whereby antibodies are being optimized for neutralization potency and breadth, biodistribution, pharmacokinetics, and effector function with the aim of revolutionizing HIV treatment and prevention options.
Collapse
|
37
|
Monoclonal antibodies to host cellular receptors for the treatment and prevention of HIV-1 infection. Curr Opin HIV AIDS 2016; 10:144-50. [PMID: 25700204 DOI: 10.1097/coh.0000000000000146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Clinically relevant monoclonal antibodies (mAb) to host cellular receptors have been generated to both the CD4 receptor and the CCR5 coreceptor, cell surface proteins critical for HIV-1 entry. Ibalizumab is a novel humanized mAb that binds to a conformational epitope on CD4 and blocks entry of HIV-1. It has broad and potent antiviral activity in vitro and in vivo. PRO 140 is a humanized mAb that binds to the CCR5 coreceptor and inhibits CCR5-tropic HIV-1 by interfering with viral entry. Antiviral activity has been demonstrated both in vitro against R5 viruses and in vivo in HIV-1-infected individuals harboring CCR5-tropic virus. RECENT FINDINGS Both antibodies have been administered intravenously in early-phase clinical trials, and current emphasis is on the development of formulations that can be administered subcutaneously. Most recently, bispecific antibodies combining either ibalizumab or PRO 140 with anti-Env broadly neutralizing antibodies have been constructed with vastly improved in-vitro neutralizing profiles, and may offer substantial advantages in the clinic. SUMMARY mAb to host cellular receptors particularly when combined with broadly neutralizing antibodies in novel conformations may offer advances in both the treatment and prevention of HIV-1 infection.
Collapse
|
38
|
Protein/peptide-based entry/fusion inhibitors as anti-HIV therapies: challenges and future direction. Rev Med Virol 2015; 26:4-20. [DOI: 10.1002/rmv.1853] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/02/2015] [Accepted: 07/15/2015] [Indexed: 11/07/2022]
|
39
|
Abstract
ABSTRACT HIV resistance against currently approved entry inhibitors, the chemokine receptor-5 (CCR5) antagonist maraviroc and the fusion inhibitor enfuvirtide (T-20), manifests in a complex manner that is distinct from the resistance patterns against other classes of antiretroviral drugs. Several attachment and fusion inhibitors are currently under various stages of development. Whereas CCR5 co-receptor antagonists have been widely studied until now, because patients who lack CCR5 are healthy and protected to some extent from HIV-infection, CXCR4-antagonist development has been slower, due to limited antiviral activity and potential toxicity given that CXCR4 may have essential cellular functions. Novel fusion inhibitor development is focusing on orally available small-molecule inhibitors that might replace T-20, which needs to be administered by subcutaneous injection.
Collapse
Affiliation(s)
- Victor G Kramer
- McGill AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Mark A Wainberg
- McGill AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
40
|
Abstract
Whereas active immunity refers to the process of exposing the individual to an antigen to generate an adaptive immune response, passive immunity refers to the transfer of antibodies from one individual to another. Passive immunity provides immediate but short-lived protection, lasting several weeks up to 3 or 4 months. Passive immunity can occur naturally, when maternal antibodies are transferred to the fetus through the placenta or from breast milk to the gut of the infant. It can also be produced artificially, when antibody preparations derived from sera or secretions of immunized donors or, more recently, different antibody producing platforms are transferred via systemic or mucosal route to nonimmune individuals. Passive immunization has recently become an attractive approach because of the emergence of new and drug-resistant microorganisms, diseases that are unresponsive to drug therapy and individuals with an impaired immune system who are unable to respond to conventional vaccines. This chapter addresses the contributions of natural and artificial acquired passive immunity in understanding the concept of passive immunization. We will mainly focus on administration of antibodies for protection against various infectious agents entering through mucosal surfaces.
Collapse
|
41
|
HIV Treatment. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2014. [DOI: 10.1007/s40506-014-0023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
42
|
Li L, Tian JH, Yang K, Zhang P, Jia WQ. Humanized PA14 (a monoclonal CCR5 antibody) for treatment of people with HIV infection. Cochrane Database Syst Rev 2014; 2014:CD008439. [PMID: 25063928 PMCID: PMC7173721 DOI: 10.1002/14651858.cd008439.pub3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND PRO 140 (a humanized form of the PA14 antibody, a monoclonal CCR5 antibody) inhibits CCR5-tropic (R5) type 1 human immunodeficiency virus (HIV). This may be an effective new treatment with the potential to address the limitations of currently available therapies for HIV-infected patients. OBJECTIVES We aimed to assess the efficacy, safety, clinical disease progression and immunologic (CD4 count/percentage) and virologic (plasma HIV RNA viral load) markers of PRO 140 for HIV-infected patients in randomized controlled trials (RCTs) and quasi-randomized controlled trials (quasi-RCTs). SEARCH METHODS We searched databases including The Cochrane Central Register of Controlled Trials (The Cochrane Library 2014, Issue 4), MEDLINE (PubMed, January 1966 to April 2014), EMBASE (January 1978 to April 2014) and ISI Web of Knowledge (January 1966 to April 2014), online trials registries and other sources. We also screened the reference lists of related literature and eligible studies, and presentations from major HIV/AIDS (human immunodeficiency virus/acquired immunodeficiency syndrome) conferences. SELECTION CRITERIA We included RCTs and quasi-RCTs comparing PRO 140 with placebo or other antiretroviral drugs, or different doses of PRO 140 for individuals infected with HIV. DATA COLLECTION AND ANALYSIS Two reviewers (L Li and JH Tian) independently screened all retrieved citations and selected eligible studies. Two authors (P Zhang and WQ Jia) independently extracted data. Any disagreements when selecting studies and extracting data were adjudicated by the review mentor (KH Yang). We used Review Manager (RevMan) software for statistical analysis based on an intention-to-treat analysis. We examined heterogeneity using the Chi(2) statistic. We regarded I(2) estimates greater than 50% as moderate or high levels of heterogeneity. According to the level of heterogeneity, we used either a fixed or random-effects model.If significant heterogeneity existed and the reasons could not be found, we reported the results qualitatively. MAIN RESULTS We included three trials comparing PRO 140 with placebo in adult patients with HIV infection. Our review indicates that PRO 140 may offer significant dose-dependent HIV-1 RNA suppression with tolerable side effects. PRO 140 2 mg/kg, 5 mg/kg, 10 mg/kg, 162 mg weekly, 324 mg biweekly, and 324 mg weekly showed statistically significant differences in the changes of HIV-1 RNA levels. HIV-1 RNA levels were reduced by intravenous (IV) infusion of PRO 140 2 mg/kg or 5 mg/kg on day 10, 5 mg/kg or 10 mg/kg on day 12, 162 mg weekly, 324 mg biweekly, or 324 mg weekly on day 22. PRO 140 2 mg/kg, 5 mg/kg, 10 mg/kg, 162 mg weekly, 324 mg biweekly, and 324 mg weekly demonstrated greater antiviral response. PRO 140 324 mg weekly, 5 mg/kg, and 10 mg/kg showed more patients with ≦ 400 copies/mL HIV-1 RNA. Only PRO 140 5 mg/kg showed greater change in CD4(+) cell count on day eight. Headache, lymphadenopathy, diarrhoea, fatigue, hypertension, nasal congestion and pruritus were reported to be the most frequent adverse events. AUTHORS' CONCLUSIONS Limited evidence from three small trials suggests that PRO 140 might demonstrate potent, short-term, dose-dependent, highly significant antiviral activity. However, as the evidence is insufficient, recommendations cannot yet be made. Larger, longer-term, double-blind RCTs are required to provide conclusive evidence.
Collapse
Affiliation(s)
- Lun Li
- Lanzhou UniversityEvidence‐Based Medicine Center, School of Basic Medical SciencesNo. 199, Donggang West RoadLanzhou CityGansuChina730000
| | - Jin Hui Tian
- Lanzhou UniversityEvidence‐Based Medicine Center, School of Basic Medical SciencesNo. 199, Donggang West RoadLanzhou CityGansuChina730000
| | - KeHu Yang
- Lanzhou UniversityKey Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu ProvinceNo. 199, Donggang West RoadLanzhou CityGansuChina730000
| | - Peng Zhang
- Nanyang Central HospitalNanyangHenanChina473000
| | - Wen Qin Jia
- Lanzhou UniversityEvidence‐Based Medicine Center, School of Basic Medical SciencesNo. 199, Donggang West RoadLanzhou CityGansuChina730000
| | | |
Collapse
|
43
|
Odongo FCA. Parenteral antiretroviral formulations are still urgently needed: a case report and commentary. Int J STD AIDS 2014; 26:436-40. [PMID: 24890687 DOI: 10.1177/0956462414538242] [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] [Received: 03/03/2014] [Accepted: 05/05/2014] [Indexed: 11/17/2022]
Abstract
This case report highlights a challenging clinical dilemma to administer antiretroviral therapy in a critically-ill human immunodeficiency virus-infected patient who presented with multiple opportunistic infections and a non-functional gastrointestinal tract. The need for parenteral antiretroviral drug options is discussed and investigational drugs are briefly reviewed.
Collapse
|
44
|
Shimoni M, Herschhorn A, Britan-Rosich Y, Kotler M, Benhar I, Hizi A. The isolation of novel phage display-derived human recombinant antibodies against CCR5, the major co-receptor of HIV. Viral Immunol 2014; 26:277-90. [PMID: 23941674 DOI: 10.1089/vim.2012.0029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Selecting for antibodies against specific cell-surface proteins is a difficult task due to many unrelated proteins that are expressed on the cell surface. Here, we describe a method to screen antibody-presenting phage libraries against native cell-surface proteins. We applied this method to isolate antibodies that selectively recognize CCR5, which is the major co-receptor for HIV entry (consequently, playing a pivotal role in HIV transmission and pathogenesis). We employed a phage screening strategy by using cells that co-express GFP and CCR5, along with an excess of control cells that do not express these proteins (and are otherwise identical to the CCR5-expressing cells). These control cells are intended to remove most of the phages that bind the cells nonspecifically; thus leading to an enrichment of the phages presenting anti-CCR5-specific antibodies. Subsequently, the CCR5-presenting cells were quantitatively sorted by flow cytometry, and the bound phages were eluted, amplified, and used for further successive selection rounds. Several different clones of human single-chain Fv antibodies that interact with CCR5-expressing cells were identified. The most specific monoclonal antibody was converted to a full-length IgG and bound the second extracellular loop of CCR5. The experimental approach presented herein for screening for CCR5-specific antibodies can be applicable to screen antibody-presenting phage libraries against any cell-surface expressed protein of interest.
Collapse
Affiliation(s)
- Moria Shimoni
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
45
|
Panos G, Watson DC. Effect of HIV-1 subtype and tropism on treatment with chemokine coreceptor entry inhibitors; overview of viral entry inhibition. Crit Rev Microbiol 2014; 41:473-87. [DOI: 10.3109/1040841x.2013.867829] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
46
|
Flanagan CA. Receptor Conformation and Constitutive Activity in CCR5 Chemokine Receptor Function and HIV Infection. ADVANCES IN PHARMACOLOGY 2014; 70:215-63. [DOI: 10.1016/b978-0-12-417197-8.00008-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
47
|
A vaccine against CCR5 protects a subset of macaques upon intravaginal challenge with simian immunodeficiency virus SIVmac251. J Virol 2013; 88:2011-24. [PMID: 24307581 DOI: 10.1128/jvi.02447-13] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
As an alternative to targeting human immunodeficiency virus (HIV), we have developed vaccines targeting CCR5, a self-protein critically involved in HIV replication and pathogenesis. By displaying peptides derived from CCR5 at high density on the surface of virus-like particles, we can efficiently induce high-titer IgG antibodies against this self-molecule. Here, we investigated whether prophylactic immunization of rhesus macaques with a particle-based vaccine targeting two regions of macaque CCR5 could prevent or suppress vaginal infection with highly virulent SIVmac251. Twelve macaques were vaccinated with a bacteriophage Qß-based vaccine targeting macaque CCR5 (Qß.CCR5). Six control animals were immunized with the Qß platform alone. All animals immunized with Qß.CCR5 developed high-titer anti-CCR5 antibody responses. Macaques were vaginally challenged with a high dose of SIVmac251. The mean peak viral RNA levels in the vaccinated groups were 30-fold lower than in the control group (10(6.8) versus 10(8.3) copies/ml plasma). Three of the 12 vaccinated macaques dramatically suppressed simian immunodeficiency virus (SIV) replication: peak viral loads were low (10(3) to 10(4) RNA copies/ml), and SIV RNA became undetectable from 6 weeks onward. No viral RNA or DNA could be detected in colon and lymph node biopsy specimens collected 13 months after challenge. In vivo depletion of CD8(+) cells failed to induce a viral rebound. However, once anti-CCR5 antibody responses had waned, the 3 animals became infected after intravaginal and/or intravenous rechallenge. In conclusion, vaccination against CCR5 was associated with dramatic suppression of virus replication in a subset (25%) of macaques. These data support further research of vaccination against CCR5 to combat HIV infection.
Collapse
|
48
|
Hashimoto C, Nomura W, Narumi T, Fujino M, Nakahara T, Yamamoto N, Murakami T, Tamamura H. CXCR4-derived synthetic peptides inducing anti-HIV-1 antibodies. Bioorg Med Chem 2013; 21:6878-85. [PMID: 24119449 DOI: 10.1016/j.bmc.2013.09.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 01/17/2023]
Abstract
Despite almost 30 years since the identification of the human immunodeficiency virus type I (HIV-1), development of effective AIDS vaccines has been hindered by the high mutability of HIV-1. The HIV-1 co-receptors CCR5 and CXCR4 are genetically stable, but viral proteins may mutate rapidly during the course of infection. CXCR4 is a seven transmembrane G protein-coupled receptor, possessing an N-terminal region (NT) and three extracellular loops (ECL1-3). Previous studies have shown that the CXCR4-ED-derived peptides inhibit the entry of HIV-1 by interacting with gp120, an HIV-1 envelope glycoprotein. In the present study, antigenicity of CXCR4-derived peptides has been investigated and the anti-HIV-1 effects of induced antisera have been assessed. It was found that CXCR4-ED-derived antigen molecules immunize mice, showing that the linear peptides have higher antigenicity than the cyclic peptides. The L1- and L2-induced antisera inhibited the HIV-1 entry significantly, while anti-N1 antibodies have no inhibitory activity. This study produced promising examples for the design of AIDS vaccines which target the human protein and can overcome mutability of HIV-1.
Collapse
Affiliation(s)
- Chie Hashimoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo 101-0062, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Santa-Marta M, de Brito PM, Godinho-Santos A, Goncalves J. Host Factors and HIV-1 Replication: Clinical Evidence and Potential Therapeutic Approaches. Front Immunol 2013; 4:343. [PMID: 24167505 PMCID: PMC3807056 DOI: 10.3389/fimmu.2013.00343] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 10/06/2013] [Indexed: 12/17/2022] Open
Abstract
HIV and human defense mechanisms have co-evolved to counteract each other. In the process of infection, HIV takes advantage of cellular machinery and blocks the action of the host restriction factors (RF). A small subset of HIV+ individuals control HIV infection and progression to AIDS in the absence of treatment. These individuals known as long-term non-progressors (LNTPs) exhibit genetic and immunological characteristics that confer upon them an efficient resistance to infection and/or disease progression. The identification of some of these host factors led to the development of therapeutic approaches that attempted to mimic the natural control of HIV infection. Some of these approaches are currently being tested in clinical trials. While there are many genes which carry mutations and polymorphisms associated with non-progression, this review will be specifically focused on HIV host RF including both the main chemokine receptors and chemokines as well as intracellular RF including, APOBEC, TRIM, tetherin, and SAMHD1. The understanding of molecular profiles and mechanisms present in LTNPs should provide new insights to control HIV infection and contribute to the development of novel therapies against AIDS.
Collapse
Affiliation(s)
- Mariana Santa-Marta
- URIA-Centro de Patogénese Molecular, Faculdade de Farmácia, Universidade de Lisboa , Lisboa , Portugal ; Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa , Lisboa , Portugal
| | | | | | | |
Collapse
|
50
|
Johnson HR, Lenhoff AM. Characterization and suitability of therapeutic antibody dense phases for subcutaneous delivery. Mol Pharm 2013; 10:3582-91. [PMID: 24011376 DOI: 10.1021/mp400006g] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Subcutaneous antibody dosing formulations comprising solid suspensions have the potential to reduce dosage viscosity and injection volume. Gel beads of three therapeutic antibodies were prepared to determine the feasibility of such formulations. The beads were formed directly from aqueous solution within 0.1-4 days upon addition of biocompatible precipitating agents under conditions compatible with the use of stabilizing excipients. The phase behavior of antibody gel beads and their mechanical characteristics were measured. Gel beads were characterized by reduced elastic moduli of 0.4-1.0 MPa, as measured by atomic force microscopy, and completely redissolved within 10-20 min under physiologic conditions, in vitro. Crystalline particles could also be prepared in some cases and were found to have reduced elastic moduli 3 orders of magnitude greater than those for the gel beads. Both crystalline and gel particles had protein concentrations of 100-180 mg/mL within the dense phase. Protein stored within the dense phase was recoverable after 40 days of incubation at room temperature or 4 °C.
Collapse
Affiliation(s)
- Harvey R Johnson
- Center for Molecular and Engineering Thermodynamics, Department of Chemical and Biomolecular Engineering, University of Delaware , Newark, Delaware 19716, United States
| | | |
Collapse
|