1
|
Lu L, Li J, Jiang X, Bai R. CXCR4/CXCL12 axis: "old" pathway as "novel" target for anti-inflammatory drug discovery. Med Res Rev 2024; 44:1189-1220. [PMID: 38178560 DOI: 10.1002/med.22011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/25/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024]
Abstract
Inflammation is the body's defense response to exogenous or endogenous stimuli, involving complex regulatory mechanisms. Discovering anti-inflammatory drugs with both effectiveness and long-term use safety is still the direction of researchers' efforts. The inflammatory pathway was initially identified to be involved in tumor metastasis and HIV infection. However, research in recent years has proved that the CXC chemokine receptor type 4 (CXCR4)/CXC motif chemokine ligand 12 (CXCL12) axis plays a critical role in the upstream of the inflammatory pathway due to its chemotaxis to inflammatory cells. Blocking the chemotaxis of inflammatory cells by CXCL12 at the inflammatory site may block and alleviate the inflammatory response. Therefore, developing CXCR4 antagonists has become a novel strategy for anti-inflammatory therapy. This review aimed to systematically summarize and analyze the mechanisms of action of the CXCR4/CXCL12 axis in more than 20 inflammatory diseases, highlighting its crucial role in inflammation. Additionally, the anti-inflammatory activities of CXCR4 antagonists were discussed. The findings might help generate new perspectives for developing anti-inflammatory drugs targeting the CXCR4/CXCL12 axis.
Collapse
Affiliation(s)
- Liuxin Lu
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junjie Li
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoying Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Curreli F, Ahmed S, Benedict Victor SM, Iusupov IR, Spiridonov EA, Belov DS, Altieri A, Kurkin AV, Debnath AK. Design, synthesis, and antiviral activity of a series of CD4-mimetic small-molecule HIV-1 entry inhibitors. Bioorg Med Chem 2021; 32:116000. [PMID: 33461144 DOI: 10.1016/j.bmc.2021.116000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/31/2020] [Indexed: 11/28/2022]
Abstract
We presented our continuing stride to optimize the second-generation NBD entry antagonist targeted to the Phe43 cavity of HIV-1 gp120. We have synthesized thirty-eight new and novel analogs of NBD-14136, earlier designed based on a CH2OH "positional switch" hypothesis, and derived a comprehensive SAR. The antiviral data confirmed that the linear alcohol towards the "N" (C4) of the thiazole ring yielded more active inhibitors than those towards the "S" (C5) of the thiazole ring. The best inhibitor, NBD-14273 (compound 13), showed both improved antiviral activity and selectivity index (SI) against HIV-1HXB2 compared to NBD-14136. We also tested NBD-14273 against a large panel of 50 HIV-1 Env-pseudotyped viruses representing clinical isolates of diverse subtypes. The overall mean data indicate that antiviral potency against these isolates improved by ~3-fold, and SI also improved ~3-fold compared to NBD-14136. This new and novel inhibitor is expected to pave the way for further optimization to a more potent and clinically relevant inhibitor against HIV-1.
Collapse
Affiliation(s)
- Francesca Curreli
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA
| | - Shahad Ahmed
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA
| | - Sofia M Benedict Victor
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA
| | - Ildar R Iusupov
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Evgeny A Spiridonov
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Dmitry S Belov
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Asim K Debnath
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA.
| |
Collapse
|
3
|
Akand EH, Maher SJ, Murray JM. Mutational networks of escape from transmitted HIV-1 infection. PLoS One 2020; 15:e0243391. [PMID: 33284837 PMCID: PMC7721145 DOI: 10.1371/journal.pone.0243391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
Human immunodeficiency virus (HIV) is subject to immune selective pressure soon after it establishes infection at the founder stage. As an individual progresses from the founder to chronic stage of infection, immune pressure forces a history of mutations that are embedded in envelope sequences. Determining this pathway of coevolving mutations can assist in understanding what is different with the founder virus and the essential pathways it takes to maintain infection. We have combined operations research and bioinformatics methods to extract key networks of mutations that differentiate founder and chronic stages for 156 subtype B and 107 subtype C envelope (gp160) sequences. The chronic networks for both subtypes revealed strikingly different hub-and-spoke topologies compared to the less structured transmission networks. This suggests that the hub nodes are impacted by the immune response and the resulting loss of fitness is compensated by mutations at the spoke positions. The major hubs in the chronic C network occur at positions 12, 137 (within the N136 glycan), and 822, and at position 306 for subtype B. While both founder networks had a more heterogeneous connected network structure, interestingly founder B subnetworks around positions 640 and 837 preferentially contained CD4 and coreceptor binding domains. Finally, we observed a differential effect of glycosylation between founder and chronic subtype B where the latter had mutational pathways significantly driven by N-glycosylation. Our study provides insights into the mutational pathways HIV takes to evade the immune response, and presents features more likely to establish founder infection, valuable for effective vaccine design.
Collapse
Affiliation(s)
- Elma H. Akand
- School of Mathematics and Statistics, UNSW Sydney, Kensington, NSW, Australia
| | - Stephen J. Maher
- College of Engineering, Mathematical and Physical Sciences, University of Exeter, Exeter, United Kingdom
| | - John M. Murray
- School of Mathematics and Statistics, UNSW Sydney, Kensington, NSW, Australia
| |
Collapse
|
4
|
Vitale RM, Thellung S, Tinto F, Solari A, Gatti M, Nuzzo G, Ioannou E, Roussis V, Ciavatta ML, Manzo E, Florio T, Amodeo P. Identification of the hydantoin alkaloids parazoanthines as novel CXCR4 antagonists by computational and in vitro functional characterization. Bioorg Chem 2020; 105:104337. [PMID: 33113408 DOI: 10.1016/j.bioorg.2020.104337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/19/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022]
Abstract
CXCR4 chemokine receptor represents an attractive pharmacological target due to its key role in cancer metastasis and inflammatory diseases. Starting from our previously-developed pharmacophoric model, we applied a combined computational and experimental approach that led to the identification of the hydantoin alkaloids parazoanthines, isolated from the Mediterranean Sea anemone Parazoanthus axinellae, as novel CXCR4 antagonists. Parazoanthine analogues were then synthesized to evaluate the contribution of functional groups to the overall activity. Within the panel of synthesized natural and non-natural parazoanthines, parazoanthine-B was identified as the most potent CXCR4 antagonist with an IC50 value of 9.3 nM, even though all the investigated compounds were able to antagonize in vitro the down-stream effects of CXC12, albeit with variable potency and efficacy. The results of our study strongly support this class of small molecules as potent CXCR4 antagonists in tumoral pathologies characterized by an overexpression of this receptor. Furthermore, their structure-activity relationships allowed the optimization of our pharmacophoric model, useful for large-scale in silico screening.
Collapse
Affiliation(s)
- Rosa Maria Vitale
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078, Pozzuoli, (NA), Italy
| | - Stefano Thellung
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Francesco Tinto
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078, Pozzuoli, (NA), Italy
| | - Agnese Solari
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Monica Gatti
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Genoveffa Nuzzo
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078, Pozzuoli, (NA), Italy
| | - Efstathia Ioannou
- Department of Pharmacognosy and Chemistry of Natural Products, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens 15771, Greece
| | - Vassilios Roussis
- Department of Pharmacognosy and Chemistry of Natural Products, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens 15771, Greece
| | - Maria Letizia Ciavatta
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078, Pozzuoli, (NA), Italy
| | - Emiliano Manzo
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078, Pozzuoli, (NA), Italy.
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; IRCCS Policlinico San Martino, 16132 Genova, Italy.
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry, National Research Council (ICB-CNR), Via Campi Flegrei 34, 80078, Pozzuoli, (NA), Italy.
| |
Collapse
|
5
|
Kardani K, Basimi P, Fekri M, Bolhassani A. Antiviral therapy for the sexually transmitted viruses: recent updates on vaccine development. Expert Rev Clin Pharmacol 2020; 13:1001-1046. [PMID: 32838584 DOI: 10.1080/17512433.2020.1814743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The sexually transmitted infections (STIs) caused by viruses including human T cell leukemia virus type-1 (HTLV-1), human immunodeficiency virus-1 (HIV-1), human simplex virus-2 (HSV-2), hepatitis C virus (HCV), hepatitis B virus (HBV), and human papillomavirus (HPV) are major public health issues. These infections can cause cancer or result in long-term health problems. Due to high prevalence of STIs, a safe and effective vaccine is required to overcome these fatal viruses. AREAS COVERED This review includes a comprehensive overview of the literatures relevant to vaccine development against the sexually transmitted viruses (STVs) using PubMed and Sciencedirect electronic search engines. Herein, we discuss the efforts directed toward development of effective vaccines using different laboratory animal models including mice, guinea pig or non-human primates in preclinical trials, and human in clinical trials with different phases. EXPERT OPINION There is no effective FDA approved vaccine against the sexually transmitted viruses (STVs) except for HBV and HPV as prophylactic vaccines. Many attempts are underway to develop vaccines against these viruses. There are several approaches for improving prophylactic or therapeutic vaccines such as heterologous prime/boost immunization, delivery system, administration route, adjuvants, etc. In this line, further studies can be helpful for understanding the immunobiology of STVs in human. Moreover, development of more relevant animal models is a worthy goal to induce effective immune responses in humans.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | - Parya Basimi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | - Mehrshad Fekri
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran , Tehran, Iran
| |
Collapse
|
6
|
A novel selective histone deacetylase I inhibitor CC-4a activates latent HIV-1 through NF-κB pathway. Life Sci 2020; 267:118427. [PMID: 32941894 DOI: 10.1016/j.lfs.2020.118427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 11/23/2022]
Abstract
AIMS The fact that HIV-1 inside human bodies can perform reverse transcription and integrate resultant DNA into host chromosome remains a challenge in AIDS treatment. "Shock and kill" strategy was proposed to achieve the functional cure, which requested latency reactivating agents (LRAs) to reactivate latent HIV-1 and then extirpate viruses and infected cells with antiviral agents and the immune system. However, there are no feasible LRAs clinically applied. Herein, we examined a synthesized HDAC I inhibitor, CC-4a, in reactivating latent HIV-1 and investigated its mechanisms. MATERIALS AND METHODS Two HIV-1 infected cell models and human PBMCs were used in this study. Flow cytometry, ELISA, luciferase, and RT-PCR assay were used to analyze the expression of viral protein and mRNA. The mechanisms were explored by using cytoplasmic nuclear protein isolation and western blotting assays. KEY FINDINGS CC-4a could successfully reactivate latent HIV-1 at the protein and gene levels with low cytotoxicity. Intriguingly, CC-4a showed the ability to induce apoptosis in HIV-1 infected cell models. CC-4a exerted a synergistic activation effect with prostratin without triggering global T cell activation and inflammatory factor storm. It was further found that CC-4a down-regulated the expressions of CCR5 and CD4. Moreover, CC-4a together with antiviral drugs was proved to antagonize HIV-1 without mutual interference. Finally, the enhanced histone acetylation and activated NF-κB pathway were detected in CC-4a mechanisms. SIGNIFICANCE The results suggested that CC-4a activated latent HIV-1 and showed promising clinical applications, demonstrating that CC-4a played a role in HIV-1 eradication in "shock and kill" strategy.
Collapse
|
7
|
Discovery of novel aminopiperidinyl amide CXCR4 modulators through virtual screening and rational drug design. Eur J Med Chem 2020; 201:112479. [PMID: 32534343 DOI: 10.1016/j.ejmech.2020.112479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/29/2022]
Abstract
The C-X-C chemokine receptor type 4 (CXCR4) is a potential therapeutic target for HIV infection, metastatic cancer, and inflammatory autoimmune diseases. In this study, we screened the ZINC chemical database for novel CXCR4 modulators through a series of in silico guided processes. After evaluating the screened compounds for their binding affinities to CXCR4 and inhibitory activities against the chemoattractant CXCL12, we identified a hit compound (ZINC 72372983) showing 100 nM affinity and 69% chemotaxis inhibition at the same concentration (100 nM). To increase the potency of our hit compound, we explored the protein-ligand interactions at an atomic level using molecular dynamics simulation which enabled us to design and synthesize a novel compound (Z7R) with nanomolar affinity (IC50 = 1.25 nM) and improved chemotaxis inhibition (78.5%). Z7R displays promising anti-inflammatory activity (50%) in a mouse edema model by blocking CXCR4-expressed leukocytes, being supported by our immunohistochemistry study.
Collapse
|
8
|
Design, synthesis and biological evaluation of novel 5-((substituted quinolin-3-yl/1-naphthyl) methylene)-3-substituted imidazolidin-2,4-dione as HIV-1 fusion inhibitors. Bioorg Chem 2020; 99:103782. [PMID: 32229348 DOI: 10.1016/j.bioorg.2020.103782] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/19/2020] [Accepted: 03/19/2020] [Indexed: 01/16/2023]
Abstract
A series of novel 5-(substituted quinolin-3-yl or 1-naphthyl)methylene)-3-substituted imidazolidin-2,4-dione 9-26 was designed and synthesized. The prepared compounds were identified using 1H NMR, 13C NMR as well as elemental analyses. The inhibitory activity of 9-26 on HIV-1IIIB replication in MT-2 cells was evaluated. Some derivatives showed good to excellent anti-HIV activities as compounds 13, 18, 19, 20, 22 and 23. They showed EC50 of 0.148, 0.460, 0.332, 0.50, 0.271 and 0.420 μM respectively being more potent than compound I (EC50 = 0.70 μM) and II ( EC50 = 2.40 μM) as standards. The inhibitory activity of 9-26 on infected primary HIV-1 domain, 92US657 (clade B, R5) was investigated. All the tested compounds consistently inhibited infection of this virus with EC50 from 0.520 to 11.857 μM. Results from SAR studies showed that substitution on ring A with 6/7/8-methyl group resulted in significant increase in the inhibitory activity against HIV-1IIIB infection (5- >300 times) compared to the unsubstituted analog 9. The cytotoxicity of these compounds on MT-2 cells was tested and their CC50 values ranged from 11 to 85 μM with selectivity indexes ranged from 0.53 to 166. The docking study revealed nice fitting of the new compounds into the hydrophobic pocket of HIV-1 gp41 and higher affinity than NB-64. Compound 13, the most active in preventing HIV-1IIIB infection, adopted a similar orientation to compound IV. Molecular docking analysis of the new compounds revealed hydrogen bonding interactions between the imidazolidine-2,4-dione ring and LYS574 which were missed in the weakly active derivatives.
Collapse
|
9
|
Tan S, Li W, Li Z, Li Y, Luo J, Yu L, Yang J, Qiu M, Cheng H, Xu W, Jiang S, Lu L, Liu S, Ma W. A Novel CXCR4 Targeting Protein SDF-1/54 as an HIV-1 Entry Inhibitor. Viruses 2019; 11:v11090874. [PMID: 31540474 PMCID: PMC6783869 DOI: 10.3390/v11090874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 02/06/2023] Open
Abstract
CXC chemokine receptor 4 (CXCR4) is a co-receptor for HIV-1 entry into target cells. Its natural ligand, the chemokine SDF-1, inhibits viral entry mediated by this receptor. However, the broad expression pattern of CXCR4 and its critical roles in various physiological and pathological processes indicate that the direct application of SDF-1 as an entry inhibitor might have severe consequences. Previously, we constructed an effective SDF-1 mutant, SDF-1/54, by deleting the α-helix of the C-terminal functional region of SDF-1. Of note, SDF-1/54 shows remarkable decreased chemotoxic ability, but maintains a similar binding affinity to CXCR4, suggesting SDF-1/54 might better serve as a CXCR4 inhibitor. Here, we found that SDF-1/54 exhibited potent antiviral activity against various X4 HIV-1 strains, including the infectious clone HIV-1 NL4-3, laboratory-adapted strain HIV-1 IIIB, clinical isolates and even drug-resistant strains. By using time-of-addition assay, non-infectious and infectious cell–cell fusion assay and CXCR4 internalization assay, we demonstrated SDF-1/54 is an HIV-1 entry inhibitor. A combination of SDF-1/54 with several antiretroviral drugs exhibited potent synergistic anti-HIV-1 activity. Moreover, SDF-1/54 was stable and its anti-HIV-1 activity was not significantly affected by the presence of seminal fluid, vaginal fluid simulant and human serum albumin. SDF-1/54 showed limited in vitro cytotoxicity to lymphocytes and vaginal epithelial cells. Based on these findings, SDF-1/54 could have a therapeutic potential as an HIV-1 entry inhibitor.
Collapse
Affiliation(s)
- Suiyi Tan
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenjuan Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhaofeng Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yujing Li
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jiangyan Luo
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Liangzhentian Yu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jie Yang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Mengjie Qiu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongyan Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai 200032, China.
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Weifeng Ma
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
10
|
Mediouni S, Jablonski JA, Tsuda S, Richard A, Kessing C, Andrade MV, Biswas A, Even Y, Tellinghuisen T, Choe H, Cameron M, Stevenson M, Valente ST. Potent suppression of HIV-1 cell attachment by Kudzu root extract. Retrovirology 2018; 15:64. [PMID: 30236131 PMCID: PMC6149077 DOI: 10.1186/s12977-018-0446-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/10/2018] [Indexed: 01/02/2023] Open
Abstract
There is a constant need to improve antiretrovirals against HIV since therapy is limited by cost, side effects and the emergence of drug resistance. Kudzu is a climbing vine from which the root extract (Pueraria lobata), rich in isoflavones and saponins, has long been used in traditional Chinese medicine for a variety of purposes, from weight loss to alcoholism prevention. Here we show that Kudzu root extract significantly inhibits HIV-1 entry into cell lines, primary human CD4+T lymphocytes and macrophages, without cell-associated toxicity. Specifically, Kudzu inhibits the initial attachment of the viral particle to the cell surface, a mechanism that depends on the envelope glycoprotein gp120 but is independent from the HIV-1 cell receptor CD4 and co-receptors CXCR4/CCR5. This activity seems selective to lentiviruses since Kudzu inhibits HIV-2 and simian immunodeficiency virus, but does not interfere with Hepatitis C, Influenza, Zika Brazil and adenovirus infection. Importantly, depending on the dose, Kudzu can act synergistically or additively with the current antiretroviral cocktails against HIV-1 and can block viruses resistant to the fusion inhibitor Enfuvirtide. Together our results highlight Kudzu's root extract value as a supplement to current antiretroviral therapy against HIV.
Collapse
Affiliation(s)
- S Mediouni
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - J A Jablonski
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - S Tsuda
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - A Richard
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - C Kessing
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - M V Andrade
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Biswas
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - Y Even
- The Botanist's Beach Farm, Jupiter, FL, USA
| | - T Tellinghuisen
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA.,Roche, Basel, Switzerland
| | - H Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA
| | - M Cameron
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL, USA
| | - M Stevenson
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - S T Valente
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, 3C1, Jupiter, FL, 33458, USA.
| |
Collapse
|
11
|
Curreli F, Belov DS, Kwon YD, Ramesh R, Furimsky AM, O'Loughlin K, Byrge PC, Iyer LV, Mirsalis JC, Kurkin AV, Altieri A, Debnath AK. Structure-based lead optimization to improve antiviral potency and ADMET properties of phenyl-1H-pyrrole-carboxamide entry inhibitors targeted to HIV-1 gp120. Eur J Med Chem 2018; 154:367-391. [PMID: 29860061 PMCID: PMC5993640 DOI: 10.1016/j.ejmech.2018.04.062] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/24/2018] [Accepted: 04/29/2018] [Indexed: 11/20/2022]
Abstract
We are continuing our concerted effort to optimize our first lead entry antagonist, NBD-11021, which targets the Phe43 cavity of the HIV-1 envelope glycoprotein gp120, to improve antiviral potency and ADMET properties. In this report, we present a structure-based approach that helped us to generate working hypotheses to modify further a recently reported advanced lead entry antagonist, NBD-14107, which showed significant improvement in antiviral potency when tested in a single-cycle assay against a large panel of Env-pseudotyped viruses. We report here the synthesis of twenty-nine new compounds and evaluation of their antiviral activity in a single-cycle and multi-cycle assay to derive a comprehensive structure-activity relationship (SAR). We have selected three inhibitors with the high selectivity index for testing against a large panel of 55 Env-pseudotyped viruses representing a diverse set of clinical isolates of different subtypes. The antiviral activity of one of these potent inhibitors, 55 (NBD-14189), against some clinical isolates was as low as 63 nM. We determined the sensitivity of CD4-binding site mutated-pseudoviruses to these inhibitors to confirm that they target HIV-1 gp120. Furthermore, we assessed their ADMET properties and compared them to the clinical candidate attachment inhibitor, BMS-626529. The ADMET data indicate that some of these new inhibitors have comparable ADMET properties to BMS-626529 and can be optimized further to potential clinical candidates.
Collapse
Affiliation(s)
- Francesca Curreli
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA
| | - Dmitry S Belov
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Young Do Kwon
- Structural Biology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ranjith Ramesh
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA
| | - Anna M Furimsky
- SRI International, Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Kathleen O'Loughlin
- SRI International, Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Patricia C Byrge
- SRI International, Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Lalitha V Iyer
- SRI International, Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Jon C Mirsalis
- SRI International, Biosciences Division, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Park, Moscow State University, Leninskie Gory Bld. 75, 77-101b, 119992 Moscow, Russia
| | - Asim K Debnath
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, NY 10065, USA.
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Alternative approaches to conventional drug-based cancer treatments have seen T cell therapies deployed more widely over the last decade. This is largely due to their ability to target and kill specific cell types based on receptor recognition. Introduction of recombinant T cell receptors (TCRs) using viral vectors and HLA-independent T cell therapies using chimeric antigen receptors (CARs) are discussed. This article reviews the tools used for genome editing, with particular emphasis on the applications of site-specific DNA nuclease mediated editing for T cell therapies. RECENT FINDINGS Genetic engineering of T cells using TCRs and CARs with redirected antigen-targeting specificity has resulted in clinical success of several immunotherapies. In conjunction, the application of genome editing technologies has resulted in the generation of HLA-independent universal T cells for allogeneic transplantation, improved T cell sustainability through knockout of the checkpoint inhibitor, programmed cell death protein-1 (PD-1), and has shown efficacy as an antiviral therapy through direct targeting of viral genomic sequences and entry receptors. SUMMARY The combined use of engineered antigen-targeting moieties and innovative genome editing technologies have recently shown success in a small number of clinical trials targeting HIV and hematological malignancies and are now being incorporated into existing strategies for other immunotherapies.
Collapse
Affiliation(s)
- Juliette M. K. M. Delhove
- Molecular Immunology Unit, UCL Great Ormond Street Institute of Child Health, University College London (UCL), 30 Guilford Street, London, WC1N 1EH UK
| | - Waseem Qasim
- Molecular Immunology Unit, UCL Great Ormond Street Institute of Child Health, University College London (UCL), 30 Guilford Street, London, WC1N 1EH UK
| |
Collapse
|
13
|
Curreli F, Kwon YD, Belov DS, Ramesh RR, Kurkin AV, Altieri A, Kwong PD, Debnath AK. Synthesis, Antiviral Potency, in Vitro ADMET, and X-ray Structure of Potent CD4 Mimics as Entry Inhibitors That Target the Phe43 Cavity of HIV-1 gp120. J Med Chem 2017; 60:3124-3153. [PMID: 28266845 DOI: 10.1021/acs.jmedchem.7b00179] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In our attempt to optimize the lead HIV-1 entry antagonist, NBD-11021, we present in this study the rational design and synthesis of 60 new analogues and determination of their antiviral activity in a single-cycle and a multicycle infection assay to derive a comprehensive structure-activity relationship (SAR). Two of these compounds, NBD-14088 and NBD-14107, showed significant improvement in antiviral activity compared to the lead entry antagonist in a single-cycle assay against a large panel of Env-pseudotyped viruses. The X-ray structure of a similar compound, NBD-14010, confirmed the binding mode of the newly designed compounds. The in vitro ADMET profiles of these compounds are comparable to that of the most potent attachment inhibitor BMS-626529, a prodrug of which is currently undergoing phase III clinical trials. The systematic study presented here is expected to pave the way for improving the potency, toxicity, and ADMET profile of this series of compounds with the potential to be moved to the early preclinical development.
Collapse
Affiliation(s)
- Francesca Curreli
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center , 310 E 67th Street, New York, New York 10065, United States
| | - Young Do Kwon
- Structural Biology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Dmitry S Belov
- EDASA Scientific, Scientific Park, Moscow State University , Leninskie Gory, Bld. 75, 77-101b; 119992 Moscow, Russia
| | - Ranjith R Ramesh
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center , 310 E 67th Street, New York, New York 10065, United States
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Park, Moscow State University , Leninskie Gory, Bld. 75, 77-101b; 119992 Moscow, Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Park, Moscow State University , Leninskie Gory, Bld. 75, 77-101b; 119992 Moscow, Russia
| | - Peter D Kwong
- Structural Biology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Asim K Debnath
- Laboratory of Molecular Modeling and Drug Design, Lindsey F. Kimball Research Institute, New York Blood Center , 310 E 67th Street, New York, New York 10065, United States
| |
Collapse
|
14
|
Riley JL, Montaner LJ. Cell-Mediated Immunity to Target the Persistent Human Immunodeficiency Virus Reservoir. J Infect Dis 2017; 215:S160-S171. [PMID: 28520969 PMCID: PMC5853458 DOI: 10.1093/infdis/jix002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Effective clearance of virally infected cells requires the sequential activity of innate and adaptive immunity effectors. In human immunodeficiency virus (HIV) infection, naturally induced cell-mediated immune responses rarely eradicate infection. However, optimized immune responses could potentially be leveraged in HIV cure efforts if epitope escape and lack of sustained effector memory responses were to be addressed. Here we review leading HIV cure strategies that harness cell-mediated control against HIV in stably suppressed antiretroviral-treated subjects. We focus on strategies that may maximize target recognition and eradication by the sequential activation of a reconstituted immune system, together with delivery of optimal T-cell responses that can eliminate the reservoir and serve as means to maintain control of HIV spread in the absence of antiretroviral therapy (ART). As evidenced by the evolution of ART, we argue that a combination of immune-based strategies will be a superior path to cell-mediated HIV control and eradication. Available data from several human pilot trials already identify target strategies that may maximize antiviral pressure by joining innate and engineered T cell responses toward testing for sustained HIV remission and/or cure.
Collapse
Affiliation(s)
- James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, and
| | - Luis J Montaner
- HIV-1 Immunopathogenesis Laboratory, Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Singh P, Mirdha BR, Srinivasan A, Rukmangadachar LA, Singh S, Sharma P, Hariprasad G, Gururao H, Luthra K. Identification of invasion proteins of Cryptosporidium parvum. World J Microbiol Biotechnol 2016; 31:1923-34. [PMID: 26492887 DOI: 10.1007/s11274-015-1936-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/27/2015] [Indexed: 11/27/2022]
Abstract
Host cell interactions and invasion by Cryptosporidium is a complex process mediated by zoites ligand-host cell receptors. Knowledge of proteins involved in this process will enable entry level inhibitors to be tried as therapeutic agents. In the present study, invasion proteins of Cryptosporidium parvum were studied in vitro. Cryptosporidium sporozoites membrane proteins were isolated and Cy5 dye labelled. They were then allowed to interact with the intact host cells. The interacting proteins were identified using 2-dimensional gel electrophoresis followed by mass spectrometry analysis. Sixty-one proteins were identified including twenty-seven previously reported invasion proteins. The newly identified proteins such as serine/threonine protein kinase, PI4 kinase, Hsp105 and coiled coil may have their roles in the parasitic invasion process. Thus, a new approach was used in the study to identify the probable proteins involved in invasion and/or host-parasite interactions. The advantage of this method is that it takes only a months' time instead of decades to identify these proteins involved in invasion process.
Collapse
|
16
|
Becerra JC, Bildstein LS, Gach JS. Recent Insights into the HIV/AIDS Pandemic. MICROBIAL CELL (GRAZ, AUSTRIA) 2016; 3:451-475. [PMID: 28357381 PMCID: PMC5354571 DOI: 10.15698/mic2016.09.529] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/27/2016] [Indexed: 12/21/2022]
Abstract
Etiology, transmission and protection: Transmission of HIV, the causative agent of AIDS, occurs predominantly through bodily fluids. Factors that significantly alter the risk of HIV transmission include male circumcision, condom use, high viral load, and the presence of other sexually transmitted diseases. Pathology/Symptomatology: HIV infects preferentially CD4+ T lymphocytes, and Monocytes. Because of their central role in regulating the immune response, depletion of CD4+ T cells renders the infected individual incapable of adequately responding to microorganisms otherwise inconsequential. Epidemiology, incidence and prevalence: New HIV infections affect predominantly young heterosexual women and homosexual men. While the mortality rates of AIDS related causes have decreased globally in recent years due to the use of highly active antiretroviral therapy (HAART) treatment, a vaccine remains an elusive goal. Treatment and curability: For those afflicted HIV infection remains a serious illness. Nonetheless, the use of advanced therapeutics have transformed a dire scenario into a chronic condition with near average life spans. When to apply those remedies appears to be as important as the remedies themselves. The high rate of HIV replication and the ability to generate variants are central to the viral survival strategy and major barriers to be overcome. Molecular mechanisms of infection: In this review, we assemble new details on the molecular events from the attachment of the virus, to the assembly and release of the viral progeny. Yet, much remains to be learned as understanding of the molecular mechanisms used in viral replication and the measures engaged in the evasion of immune surveillance will be important to develop effective interventions to address the global HIV pandemic.
Collapse
Affiliation(s)
- Juan C. Becerra
- Department of Medicine, Division of Infectious Diseases, University
of California, Irvine, Irvine, CA 92697, USA
| | | | - Johannes S. Gach
- Department of Medicine, Division of Infectious Diseases, University
of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
17
|
Barriga GP, Villalón-Letelier F, Márquez CL, Bignon EA, Acuña R, Ross BH, Monasterio O, Mardones GA, Vidal SE, Tischler ND. Inhibition of the Hantavirus Fusion Process by Predicted Domain III and Stem Peptides from Glycoprotein Gc. PLoS Negl Trop Dis 2016; 10:e0004799. [PMID: 27414047 PMCID: PMC4945073 DOI: 10.1371/journal.pntd.0004799] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 06/02/2016] [Indexed: 12/17/2022] Open
Abstract
Hantaviruses can cause hantavirus pulmonary syndrome or hemorrhagic fever with renal syndrome in humans. To enter cells, hantaviruses fuse their envelope membrane with host cell membranes. Previously, we have shown that the Gc envelope glycoprotein is the viral fusion protein sharing characteristics with class II fusion proteins. The ectodomain of class II fusion proteins is composed of three domains connected by a stem region to a transmembrane anchor in the viral envelope. These fusion proteins can be inhibited through exogenous fusion protein fragments spanning domain III (DIII) and the stem region. Such fragments are thought to interact with the core of the fusion protein trimer during the transition from its pre-fusion to its post-fusion conformation. Based on our previous homology model structure for Gc from Andes hantavirus (ANDV), here we predicted and generated recombinant DIII and stem peptides to test whether these fragments inhibit hantavirus membrane fusion and cell entry. Recombinant ANDV DIII was soluble, presented disulfide bridges and beta-sheet secondary structure, supporting the in silico model. Using DIII and the C-terminal part of the stem region, the infection of cells by ANDV was blocked up to 60% when fusion of ANDV occurred within the endosomal route, and up to 95% when fusion occurred with the plasma membrane. Furthermore, the fragments impaired ANDV glycoprotein-mediated cell-cell fusion, and cross-inhibited the fusion mediated by the glycoproteins from Puumala virus (PUUV). The Gc fragments interfered in ANDV cell entry by preventing membrane hemifusion and pore formation, retaining Gc in a non-resistant homotrimer stage, as described for DIII and stem peptide inhibitors of class II fusion proteins. Collectively, our results demonstrate that hantavirus Gc shares not only structural, but also mechanistic similarity with class II viral fusion proteins, and will hopefully help in developing novel therapeutic strategies against hantaviruses. The infection of cells by enveloped viruses involves the fusion of membranes between viruses and cells. This process is mediated by viral fusion proteins that have been grouped into at least three structural classes. Membrane-enveloped hantaviruses are worldwide spread pathogens that can cause human disease with mortality rates reaching up to 50%, however, neither a therapeutic drug nor preventive measures are currently available. Here we show that the entrance of Andes hantavirus into target cells can be blocked by fragments derived from the Gc fusion protein that are analogous to inhibitory fragments of class II fusion proteins. The Gc fragments acted directly over the viral fusion process, preventing its late stages. Together, our data demonstrate that the hantavirus Gc protein shares not only structural, but also mechanistic similarity with class II fusion proteins, suggesting its evolution from a common or related ancestral fusion protein. Furthermore, the results outline novel approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Gonzalo P. Barriga
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | | | - Chantal L. Márquez
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Eduardo A. Bignon
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Rodrigo Acuña
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Breyan H. Ross
- Laboratory of Structural Cell Biology, Department of Physiology, and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Octavio Monasterio
- Laboratorio de Biología Estructural y Molecular, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gonzalo A. Mardones
- Laboratory of Structural Cell Biology, Department of Physiology, and Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Simon E. Vidal
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
| | - Nicole D. Tischler
- Molecular Virology Laboratory, Fundación Ciencia & Vida, Santiago, Chile
- * E-mail:
| |
Collapse
|
18
|
Li X, Kuang Y, Huang X, Zou L, Huang L, Yang T, Li W, Yang Y. Preparation and characterization of a new monoclonal antibody against CXCR4 using lentivirus vector. Int Immunopharmacol 2016; 36:100-105. [DOI: 10.1016/j.intimp.2016.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 11/27/2022]
|
19
|
Garg H, Lee RT, Maurer-Stroh S, Joshi A. HIV-1 adaptation to low levels of CCR5 results in V3 and V2 loop changes that increase envelope pathogenicity, CCR5 affinity and decrease susceptibility to Maraviroc. Virology 2016; 493:86-99. [DOI: 10.1016/j.virol.2016.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/08/2016] [Accepted: 03/11/2016] [Indexed: 11/26/2022]
|
20
|
New Approach for Inhibition of HIV Entry: Modifying CD4 Binding Sites by Thiolated Pyrimidine Derivatives. Pathol Oncol Res 2016; 22:617-23. [PMID: 26860867 DOI: 10.1007/s12253-016-0044-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/27/2016] [Indexed: 10/22/2022]
Abstract
Thiolated pyrimidine derivatives have been synthetized and their antiretroviral effect against human immunodeficiency virus type 1 (HIV-1IIIB) and HIV-1 chimeric pseudovirions have been quantitatively determined in cell-based viral infectivity assays including syncytium inhibition assay as well as a single-cycle viral infection assay on HeLaCD4-LTR/ß-gal cells. Pseudotype virions prepared bearing HIV-1 envelope preference for CCR5 coreceptor, CXCR4 coreceptor or for both, respectively, with a HIV-1 core containing luciferase reporter gene were able to infect susceptible cells but are replication defective so unable to replicate in the cells . Data indicate that thiolated pyrimidine derivatives inhibited effectively virally induced cell fusion in vitro as well as infectivity of primary HIV-1IIIB strain and HIV-1 pseudovirions using chemokine receptors CCR5 or CXCR4 or both for virus entry a dose dependent manner. Inhibition was selective, depended on the pseudovirus coreceptor preference. Our results suggest that some of these sulfur containing pyrimidines interact with redoxactive -SH groups required for successful HIV entry, including a redox active disulfide in the CD4 molecule as well as -SH groups in HIV viral envelope gp120. This mode of action is unique representing a new class of potential HIV entry inhibitors.
Collapse
|
21
|
Identification of novel 2-(1H-indol-1-yl)-benzohydrazides CXCR4 ligands impairing breast cancer growth and motility. Future Med Chem 2016; 8:93-106. [DOI: 10.4155/fmc.15.176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Stromal-derived-factor-1 (SDF-1) and the G-protein-coupled receptor CXCR4 are involved in several physiological and pathological processes including breast cancer spread and progression. Several CXCR4 antagonists have currently reached advanced development stages as potential therapeutic agents for different diseases. Results: A small series of novel CXCR4 ligands, based on a 2-(1H-indol-1-yl)-benzohydrazide scaffold, has been designed and synthesized. The interaction with CXCR4-active site was predicted by molecular docking and confirmed by whole cell-based [125I]-SDF-1 ligand competition binding assays. One of the synthesized compounds was particularly active in blocking SDF-1-induced breast cancer cell motility, proliferation and downstream signaling activation in different breast cancer cell models and coculture systems. Conclusion: The newly synthesized compounds represent suitable leads for the development of innovative therapeutic agents targeting CXCR4.
Collapse
|
22
|
Prigozhin DM, Modis Y. Flunarizine arrests hepatitis C virus membrane fusion. Hepatology 2016; 63:14-6. [PMID: 26389725 PMCID: PMC4688161 DOI: 10.1002/hep.28224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/13/2015] [Indexed: 12/07/2022]
Affiliation(s)
- Daniil M. Prigozhin
- Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Yorgo Modis
- Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| |
Collapse
|
23
|
|
24
|
Tryptophan dendrimers that inhibit HIV replication, prevent virus entry and bind to the HIV envelope glycoproteins gp120 and gp41. Eur J Med Chem 2015; 106:34-43. [DOI: 10.1016/j.ejmech.2015.10.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 11/17/2022]
|
25
|
Emileh A, Duffy C, Holmes AP, Rosemary Bastian A, Aneja R, Tuzer F, Rajagopal S, Li H, Abrams CF, Chaiken IM. Covalent conjugation of a peptide triazole to HIV-1 gp120 enables intramolecular binding site occupancy. Biochemistry 2014; 53:3403-14. [PMID: 24801282 PMCID: PMC4045323 DOI: 10.1021/bi500136f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
The HIV-1 gp120 glycoprotein is the
main viral surface protein
responsible for initiation of the entry process and, as such, can
be targeted for the development of entry inhibitors. We previously
identified a class of broadly active peptide triazole (PT) dual antagonists
that inhibit gp120 interactions at both its target receptor and coreceptor
binding sites, induce shedding of gp120 from virus particles prior
to host–cell encounter, and consequently can prevent viral
entry and infection. However, our understanding of the conformational
alterations in gp120 by which PT elicits its dual receptor antagonism
and virus inactivation functions is limited. Here, we used a recently
developed computational model of the PT–gp120 complex as a
blueprint to design a covalently conjugated PT–gp120 recombinant
protein. Initially, a single-cysteine gp120 mutant, E275CYU-2, was expressed and characterized. This variant retains excellent
binding affinity for peptide triazoles, for sCD4 and other CD4 binding
site (CD4bs) ligands, and for a CD4-induced (CD4i) ligand that binds
the coreceptor recognition site. In parallel, we synthesized a PEGylated
and biotinylated peptide triazole variant that retained gp120 binding
activity. An N-terminally maleimido variant of this PEGylated PT,
denoted AE21, was conjugated to E275C gp120 to produce the AE21–E275C
covalent conjugate. Surface plasmon resonance interaction analysis
revealed that the PT–gp120 conjugate exhibited suppressed binding
of sCD4 and 17b to gp120, signatures of a PT-bound state of envelope
protein. Similar to the noncovalent PT–gp120 complex, the covalent
conjugate was able to bind the conformationally dependent mAb 2G12.
The results argue that the PT–gp120 conjugate is structurally
organized, with an intramolecular interaction between the PT and gp120
domains, and that this structured state embodies a conformationally
entrapped gp120 with an altered bridging sheet but intact 2G12 epitope.
The similarities of the PT–gp120 conjugate to the noncovalent
PT–gp120 complex support the orientation of binding of PT to
gp120 predicted in the molecular dynamics simulation model of the
PT–gp120 noncovalent complex. The conformationally stabilized
covalent conjugate can be used to expand the structural definition
of the PT-induced “off” state of gp120, for example,
by high-resolution structural analysis. Such structures could provide
a guide for improving the subsequent structure-based design of inhibitors
with the peptide triazole mode of action.
Collapse
Affiliation(s)
- Ali Emileh
- Chemical and Biological Engineering, Drexel University , Philadelphia, Pennsylvania 19104, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhao N, Pei SN, Parekh P, Salazar E, Zu Y. Blocking interaction of viral gp120 and CD4-expressing T cells by single-stranded DNA aptamers. Int J Biochem Cell Biol 2014; 51:10-8. [PMID: 24661998 DOI: 10.1016/j.biocel.2014.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 02/26/2014] [Accepted: 03/13/2014] [Indexed: 01/29/2023]
Abstract
To investigate the potential clinical application of aptamers to prevention of HIV infection, single-stranded DNA (ssDNA) aptamers specific for CD4 were developed using the systematic evolution of ligands by exponential enrichment approach and next generation sequencing. In contrast to RNA-based aptamers, the developed ssDNA aptamers were stable in human serum up to 12h. Cell binding assays revealed that the aptamers specifically targeted CD4-expressing cells with high binding affinity (Kd=1.59nM), a concentration within the range required for therapeutic application. Importantly, the aptamers selectively bound CD4 on human cells and disrupted the interaction of viral gp120 to CD4 receptors, which is a prerequisite step of HIV-1 infection. Functional studies showed that the aptamer polymers significantly blocked binding of viral gp120 to CD4-expressing cells by up to 70% inhibition. These findings provide a new approach to prevent HIV-1 transmission using oligonucleotide aptamers.
Collapse
Affiliation(s)
- Nianxi Zhao
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, and Cancer Pathology Laboratory, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Sung-nan Pei
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, and Cancer Pathology Laboratory, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Parag Parekh
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, and Cancer Pathology Laboratory, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Eric Salazar
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, and Cancer Pathology Laboratory, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, and Cancer Pathology Laboratory, Houston Methodist Research Institute, 6565 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Ray-Saha S, Huber T, Sakmar TP. Antibody epitopes on g protein-coupled receptors mapped with genetically encoded photoactivatable cross-linkers. Biochemistry 2014; 53:1302-10. [PMID: 24490954 PMCID: PMC3985944 DOI: 10.1021/bi401289p] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
We
developed a strategy for creating epitope maps of monoclonal
antibodies (mAbs) that bind to G protein-coupled receptors (GPCRs)
containing photo-cross-linkers. Using human CXC chemokine receptor
4 (CXCR4) as a model system, we genetically incorporated the photolabile
unnatural amino acid p-azido-l-phenylalanine
(azF) at various positions within extracellular loop 2 (EC2). We then
mapped the interactions of the azF-CXCR4 variants with mAb 12G5 using
targeted loss-of-function studies and photo-cross-linking in whole
cells in a microplate-based format. We used a novel variation of a
whole cell enzyme-linked immunosorbent assay to quantitate cross-linking
efficiency. 12G5 cross-linked primarily to residues 184, 178, and
189 in EC2 of CXCR4. Mapping of the data to the crystal structure
of CXCR4 showed a distinct mAb epitope footprint with the photo-cross-linked
residues clustered around the loss-of-function sites. We also used
the targeted photo-cross-linking approach to study the interaction
of human CC chemokine receptor 5 (CCR5) with PRO 140, a humanized
mAb that inhibits human immunodeficiency virus-1 cellular entry, and
2D7. The mAbs produced distinct cross-linking patterns on EC2 of CCR5.
PRO 140 cross-linked primarily to residues 174 and 175 at the amino-terminal
end of EC2, and 2D7 cross-linked mainly to residues 170, 176, and
184. These results were mapped to the recent crystal structure of
CCR5 in complex with maraviroc, showing cross-linked residues at the
tip of the maraviroc binding crevice formed by EC2. As a strategy
for mapping mAb epitopes on GPCRs, our targeted photo-cross-linking
method is complementary to loss-of-function mutagenesis results and
should be especially useful for studying mAbs with discontinuous epitopes.
Collapse
Affiliation(s)
- Sarmistha Ray-Saha
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University , 1230 York Avenue, New York, New York 10065, United States
| | | | | |
Collapse
|
28
|
Fofana I, Jilg N, Chung RT, Baumert TF. Entry inhibitors and future treatment of hepatitis C. Antiviral Res 2014; 104:136-42. [PMID: 24525381 DOI: 10.1016/j.antiviral.2014.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/29/2014] [Accepted: 02/02/2014] [Indexed: 12/25/2022]
Abstract
Hepatitis C virus (HCV) is a major cause of liver cirrhosis and hepatocellular carcinoma. Furthermore, HCV-induced liver disease is the leading indication for liver transplantation. The recent introduction of direct-acting antivirals (DAAs) has revolutionized HCV treatment by making possible the cure of the majority of patients. However, their efficacy and safety in difficult-to-treat patients such as patients receiving immunosuppression, those with advanced liver disease, co-morbidity and HIV/HCV-co-infection remain to be determined. Furthermore, prevention of liver graft infection remains a pressing issue. HCV entry inhibitors target the very first step of the HCV life cycle and efficiently inhibit cell-cell transmission - a key prerequisite for viral spread. Because of their unique mechanism of action on cell-cell transmission they may provide a promising and simple perspective for prevention of liver graft infection. A high genetic barrier to resistance and complementary mechanism of action compared to DAAs makes entry inhibitors attractive as a new strategy for treatment of multi-resistant or difficult-to-treat patients. Clinical studies are needed to determine the future role of entry inhibitors in the arsenal of antivirals to combat HCV infection. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication."
Collapse
Affiliation(s)
- Isabel Fofana
- Inserm U1110, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | - Nikolaus Jilg
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Raymond T Chung
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Thomas F Baumert
- Inserm U1110, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
29
|
Dürr R, Keppler O, Christ F, Crespan E, Garbelli A, Maga G, Dietrich U. Targeting Cellular Cofactors in HIV Therapy. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_45] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
30
|
Rudick JG, Laakso MM, Schloss AC, DeGrado WF. Template-constrained cyclic sulfopeptide HIV-1 entry inhibitors. Org Biomol Chem 2013; 11:7096-100. [PMID: 24065278 PMCID: PMC3889485 DOI: 10.1039/c3ob41395k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Template-constrained cyclic sulfopeptides that inhibit HIV-1 entry were rationally designed based on a loop from monoclonal antibody (mAb) 412d. A focused set of sulfopeptides was synthesized using Fmoc-Tyr(SO3DCV)-OH (DCV = 2,2-dichlorovinyl). Three cyclic sulfopeptides that inhibit entry of HIV-1 and complement the activity of known CCR5 antagonists were identified.
Collapse
Affiliation(s)
- Jonathan G. Rudick
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meg M. Laakso
- Department Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ashley C. Schloss
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William F. DeGrado
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
31
|
Stolp ZD, Stotland A, Diaz S, Hilton BJ, Burford W, Wolkowicz R. A Novel Two-Tag System for Monitoring Transport and Cleavage through the Classical Secretory Pathway - Adaptation to HIV Envelope Processing. PLoS One 2013; 8:e68835. [PMID: 23840860 PMCID: PMC3686725 DOI: 10.1371/journal.pone.0068835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 06/03/2013] [Indexed: 11/19/2022] Open
Abstract
The classical secretory pathway is essential for the transport of a host of proteins to the cell surface and/or extracellular matrix. While the pathway is well-established, many factors still remain to be elucidated. One of the most relevant biological processes that occur during transport involves the cleavage of pro-proteins by enzymes residing in the endoplasmic reticulum/Golgi/TransGolgi Network compartment. Teasing out the requirements involved in the classical secretory pathway and cleavage during transport would shed new light into mis-regulation leading to disease. Current methodologies fail to link transport and cleavage at the single cell level. Here, we describe a cell-based assay that relies on an engineered protein scaffold that can discriminate between transport to the cell surface, in the absence or presence of cleavage. Our novel two-tag system works in a robust and quantitative manner and distinguishes between cleaved and non-cleaved events based on cell surface expression of one or two epitope tags, respectively. Here, we have used the HIV-1 envelope as a substrate, which is cleaved during transport, as proof of principle. Importantly, this assay can be easily coupled to existing siRNA-based screens to identify novel regulators and effectors involved in transport and/or cleavage of cell surface proteins. In addition, unlike other in vivo based assays, the assay described here can also be easily adapted to drug discovery purposes.
Collapse
Affiliation(s)
- Zachary D. Stolp
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Aleksandr Stotland
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Samantha Diaz
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Brett J. Hilton
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Wesley Burford
- Department of Biology, San Diego State University, San Diego, California, United States of America
| | - Roland Wolkowicz
- Department of Biology, San Diego State University, San Diego, California, United States of America
| |
Collapse
|
32
|
Characterization of antiviral activity of benzamide derivative AH0109 against HIV-1 infection. Antimicrob Agents Chemother 2013; 57:3547-54. [PMID: 23669388 DOI: 10.1128/aac.00100-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In the absence of an effective vaccine against HIV-1 infection, anti-HIV-1 strategies play a major role in disease control. However, the rapid emergence of drug resistance against all currently used anti-HIV-1 molecules necessitates the development of new antiviral molecules and/or strategies against HIV-1 infection. In this study, we have identified a benzamide derivative named AH0109 that exhibits potent anti-HIV-1 activity at an 50% effective concentration of 0.7 μM in HIV-1-susceptible CD4(+) C8166 T cells. Mechanistic analysis revealed that AH0109 significantly inhibits both HIV-1 reverse transcription and viral cDNA nuclear import. Furthermore, our infection experiments indicated that AH0109 is capable of disrupting the replication of HIV-1 strains that are resistant to the routinely used anti-HIV-1 drugs zidovudine, lamivudine, nevirapine, and raltegravir. Together, these findings provide evidence for a newly identified antiviral molecule that can potentially be developed as an anti-HIV-1 agent.
Collapse
|
33
|
Identification of potent and orally bioavailable nucleotide competing reverse transcriptase inhibitors: in vitro and in vivo optimization of a series of benzofurano[3,2-d]pyrimidin-2-one derived inhibitors. Bioorg Med Chem Lett 2013; 23:3967-75. [PMID: 23673016 DOI: 10.1016/j.bmcl.2013.04.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/10/2013] [Accepted: 04/16/2013] [Indexed: 02/05/2023]
Abstract
Recently, a new class of HIV reverse transcriptase (HIV-RT) inhibitors has been reported. The novel mechanism of inhibition by this class involves competitive binding to the active site of the RT enzyme and has been termed Nucleotide-Competing Reverse Transcriptase Inhibitors (NcRTIs). In this publication we describe the optimization of a novel benzofurano[3,2-d]pyrimidin-2-one series of NcRTIs. The starting point for the current study was inhibitor 2, which had high biochemical and antiviral potency but only moderate permeability in a Caco-2 assay and high B-to-A efflux, resulting in moderate rat bioavailability and low Cmax. We present herein the results and strategies we employed to optimize both the potency as well as the permeability, metabolic stability and pharmacokinetic profile of this series. One of the key observations of the present study was the importance of shielding polar functionality, at least in the context of the current chemotype, to enhance permeability. These studies led to the identification of inhibitors 39 and 45, which display sub-nanomolar antiviral potency in a p24 ELISA assay with significantly reduced efflux ratios (ratios <1.5). These inhibitors also display excellent rat pharmacokinetic profiles with high bioavailabilities and low clearance.
Collapse
|
34
|
Haqqani AA, Tilton JC. Entry inhibitors and their use in the treatment of HIV-1 infection. Antiviral Res 2013; 98:158-70. [PMID: 23541872 DOI: 10.1016/j.antiviral.2013.03.017] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 12/20/2022]
Abstract
Entry of HIV into target cells is a complex, multi-stage process involving sequential attachment and CD4 binding, coreceptor binding, and membrane fusion. HIV entry inhibitors are a complex group of drugs with multiple mechanisms of action depending on the stage of the viral entry process they target. Two entry inhibitors are currently approved for the treatment of HIV-infected patients. Maraviroc, a CCR5 antagonist, blocks interactions between the viral envelope proteins and the CCR5 coreceptor. Enfuvirtide, a fusion inhibitor, disrupts conformational changes in gp41 that drive membrane fusion. A wide array of additional agents are in various stages of development. This review covers the entry inhibitors and their use in the treatment of HIV-infected patients.
Collapse
Affiliation(s)
- Aiman A Haqqani
- Case Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH 44106, USA
| | | |
Collapse
|
35
|
Emileh A, Tuzer F, Yeh H, Umashankara M, Moreira DRM, Lalonde JM, Bewley CA, Abrams CF, Chaiken IM. A model of peptide triazole entry inhibitor binding to HIV-1 gp120 and the mechanism of bridging sheet disruption. Biochemistry 2013; 52:2245-61. [PMID: 23470147 DOI: 10.1021/bi400166b] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Peptide triazole (PT) entry inhibitors prevent HIV-1 infection by blocking the binding of viral gp120 to both the HIV-1 receptor and the coreceptor on target cells. Here, we used all-atom explicit solvent molecular dynamics (MD) to propose a model for the encounter complex of the peptide triazoles with gp120. Saturation transfer difference nuclear magnetic resonance (STD NMR) and single-site mutagenesis experiments were performed to test the simulation results. We found that docking of the peptide to a conserved patch of residues lining the "F43 pocket" of gp120 in a bridging sheet naïve gp120 conformation of the glycoprotein led to a stable complex. This pose prevents formation of the bridging sheet minidomain, which is required for receptor-coreceptor binding, providing a mechanistic basis for dual-site antagonism of this class of inhibitors. Burial of the peptide triazole at the gp120 inner domain-outer domain interface significantly contributed to complex stability and rationalizes the significant contribution of hydrophobic triazole groups to peptide potency. Both the simulation model and STD NMR experiments suggest that the I-X-W [where X is (2S,4S)-4-(4-phenyl-1H-1,2,3-triazol-1-yl)pyrrolidine] tripartite hydrophobic motif in the peptide is the major contributor of contacts at the gp120-PT interface. Because the model predicts that the peptide Trp side chain hydrogen bonding with gp120 S375 contributes to the stability of the PT-gp120 complex, we tested this prediction through analysis of peptide binding to gp120 mutant S375A. The results showed that a peptide triazole KR21 inhibits S375A with 20-fold less potency than WT, consistent with predictions of the model. Overall, the PT-gp120 model provides a starting point for both the rational design of higher-affinity peptide triazoles and the development of structure-minimized entry inhibitors that can trap gp120 into an inactive conformation and prevent infection.
Collapse
Affiliation(s)
- Ali Emileh
- Chemical and Biological Engineering, Drexel University, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
James CA, DeRoy P, Duplessis M, Edwards PJ, Halmos T, Minville J, Morency L, Morin S, Simoneau B, Tremblay M, Bethell R, Cordingley M, Duan J, Lamorte L, Pelletier A, Rajotte D, Salois P, Tremblay S, Sturino CF. Nucleotide competing reverse transcriptase inhibitors: discovery of a series of non-basic benzofurano[3,2-d]pyrimidin-2-one derived inhibitors. Bioorg Med Chem Lett 2013; 23:2781-6. [PMID: 23545107 DOI: 10.1016/j.bmcl.2013.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/24/2013] [Accepted: 02/01/2013] [Indexed: 10/27/2022]
Abstract
A HTS screen led to the identification of a benzofurano[3,2-d]pyrimidin-2-one core structure which upon further optimization resulted in 1 as a potent HIV-1 nucleotide competing reverse transcriptase inhibitor (NcRTI). Investigation of the SAR at N-1 allowed significant improvements in potency and when combined with the incorporation of heterocycles at C-8 resulted in potent analogues not requiring a basic amine to achieve antiviral activity. Additional modifications at N-1 resulted in 33 which demonstrated excellent antiviral potency and improved physicochemical properties.
Collapse
Affiliation(s)
- Clint A James
- Boehringer Ingelheim (Canada) Ltd, Research and Development, 2100 Cunard Street, Laval, Québec, Canada H7S 2G5.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Peterson CW, Younan P, Jerome KR, Kiem HP. Combinatorial anti-HIV gene therapy: using a multipronged approach to reach beyond HAART. Gene Ther 2013; 20:695-702. [PMID: 23364313 DOI: 10.1038/gt.2012.98] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/19/2012] [Accepted: 11/22/2012] [Indexed: 12/11/2022]
Abstract
The 'Berlin Patient', who maintains suppressed levels of HIV viremia in the absence of antiretroviral therapy, continues to be a standard bearer in HIV eradication research. However, the unique circumstances surrounding his functional cure are not applicable to most HIV(+) patients. To achieve a functional or sterilizing cure in a greater number of infected individuals worldwide, combinatorial treatments, targeting multiple stages of the viral life cycle, will be essential. Several anti-HIV gene therapy approaches have been explored recently, including disruption of the C-C chemokine receptor 5 (CCR5) and CXC chemokine receptor 4 (CXCR4) coreceptor loci in CD4(+) T cells and CD34(+) hematopoietic stem cells. However, less is known about the efficacy of these strategies in patients and more relevant HIV model systems such as non-human primates (NHPs). Combinatorial approaches, including genetic disruption of integrated provirus, functional enhancement of endogenous restriction factors and/or the use of pharmacological adjuvants, could amplify the anti-HIV effects of CCR5/CXCR4 gene disruption. Importantly, delivering gene disruption molecules to genetic sites of interest will likely require optimization on a cell type-by-cell type basis. In this review, we highlight the most promising gene therapy approaches to combat HIV infection, methods to deliver these therapies to hematopoietic cells and emphasize the need to target viral replication pre- and post-entry to mount a suitably robust defense against spreading infection.
Collapse
Affiliation(s)
- C W Peterson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
38
|
Pierson TC, Kielian M. Flaviviruses: braking the entering. Curr Opin Virol 2013; 3:3-12. [PMID: 23352692 DOI: 10.1016/j.coviro.2012.12.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/05/2012] [Accepted: 12/05/2012] [Indexed: 12/12/2022]
Abstract
Flaviviruses are small spherical virus particles covered by a dense icosahedral array of envelope (E) proteins that mediate virus attachment to cells and the fusion of viral and cellular membranes. Our understanding of the mechanism by which flavivirus E proteins orchestrate entry into cells has been advanced by studies of E structure and arrangement on the virion at different steps of the virus entry/membrane fusion process. When combined with an increasingly clear (albeit still incomplete) view of the cell biology of virus entry, these advances suggest new antiviral strategies. Indeed, inhibitors that target cellular and viral processes involved in entry show promise as powerful tools to study this critical step of the viral lifecycle, and with luck, may ultimately lead to therapeutic advances.
Collapse
Affiliation(s)
- Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, United States.
| | | |
Collapse
|
39
|
Debnath B, Xu S, Grande F, Garofalo A, Neamati N. Small molecule inhibitors of CXCR4. Am J Cancer Res 2013; 3:47-75. [PMID: 23382786 PMCID: PMC3563081 DOI: 10.7150/thno.5376] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/04/2012] [Indexed: 12/18/2022] Open
Abstract
CXCR4 is a G-protein-coupled receptor involved in a number of physiological processes in the hematopoietic and immune systems. The SDF-1/CXCR4 axis is significantly associated with several diseases, such as HIV, cancer, WHIM syndrome, rheumatoid arthritis, pulmonary fibrosis and lupus. For example, CXCR4 is one of the major co-receptors for HIV entry into target cells, while in cancer it plays an important role in tumor cell metastasis. Several promising CXCR4 antagonists have been developed to block SDF-1/CXCR4 interactions that are currently under different stages of development. The first in class CXCR4 antagonist, plerixafor, was approved by the FDA in 2008 for the mobilization of hematopoietic stem cells and several other drugs are currently in clinical trials for cancer, HIV, and WHIM syndrome. While the long-term safety data for the first generation CXCR4 antagonists are not yet available, several new compounds are under preclinical development in an attempt to provide safer and more efficient treatment options for HIV and cancer patients.
Collapse
|
40
|
Alonzo F, Kozhaya L, Rawlings SA, Reyes-Robles T, DuMont AL, Myszka DG, Landau NR, Unutmaz D, Torres VJ. CCR5 is a receptor for Staphylococcus aureus leukotoxin ED. Nature 2012; 493:51-5. [PMID: 23235831 PMCID: PMC3536884 DOI: 10.1038/nature11724] [Citation(s) in RCA: 228] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 10/26/2012] [Indexed: 12/14/2022]
Abstract
Pore-forming toxins are critical virulence factors for many bacterial pathogens and are central to Staphylococcus aureus-mediated killing of host cells. S. aureus encodes pore-forming bi-component leukotoxins that are toxic towards neutrophils, but also specifically target other immune cells. Despite decades since the first description of staphylococcal leukocidal activity, the host factors responsible for the selectivity of leukotoxins towards different immune cells remain unknown. Here we identify the human immunodeficiency virus (HIV) co-receptor CCR5 as a cellular determinant required for cytotoxic targeting of subsets of myeloid cells and T lymphocytes by the S. aureus leukotoxin ED (LukED). We further demonstrate that LukED-dependent cell killing is blocked by CCR5 receptor antagonists, including the HIV drug maraviroc. Remarkably, CCR5-deficient mice are largely resistant to lethal S. aureus infection, highlighting the importance of CCR5 targeting in S. aureus pathogenesis. Thus, depletion of CCR5(+) leukocytes by LukED suggests a new immune evasion mechanism of S. aureus that can be therapeutically targeted.
Collapse
Affiliation(s)
- Francis Alonzo
- Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | | | | | | | | | |
Collapse
|