1
|
Ji L, Deng S, Xu JB, Li X, De Jonghe S, Schols D, Gao F. Synthesis of Thioloformate-Containing Lathyrane Diterpene Derivates via a [3,3] Sigmatropic Rearrangement and Their Anti-HIV Activity. JOURNAL OF NATURAL PRODUCTS 2025; 88:1091-1097. [PMID: 40132128 DOI: 10.1021/acs.jnatprod.5c00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
The [3,3] sigmatropic rearrangement of a lathyrane diterpene with various allylic thionoformates was carried out, affording C17-thioloformate-containing lathyrane derivatives (3a-3h) for the first time. The reaction features a mild, rapid, and easy operation. All newly synthesized derivatives were evaluated for potential antiviral activity against HIV-1 and HIV-2. The incorporation of an appropriate thionoformate into the lathyrane diterpene framework enhances their anti-HIV activity. The derivative 3d, featuring an O-(p-tolyl) carbonothionate substitution, exhibited the most potent anti-HIV activity, with an EC50 value of 11.3 μM against HIV-1 NL 4.3 and an EC50 value of 6.6 μM against HIV-2 ROD. Additionally, it demonstrated selectivity indices exceeding 4.0 and 6.8 against HIV-1 NL 4.3 and HIV-2 ROD, respectively.
Collapse
Affiliation(s)
- Lanfei Ji
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Sihao Deng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Jin-Bu Xu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Xiaohuan Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| | - Steven De Jonghe
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, KU Leuven, Herestraat 49, Box 1043, 3000 Leuven, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, KU Leuven, Herestraat 49, Box 1043, 3000 Leuven, Belgium
| | - Feng Gao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, Sichuan, People's Republic of China
| |
Collapse
|
2
|
Andrysik Z, Espinosa JM. Harnessing p53 for targeted cancer therapy: new advances and future directions. Transcription 2025; 16:3-46. [PMID: 40031988 PMCID: PMC11970777 DOI: 10.1080/21541264.2025.2452711] [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: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 03/05/2025] Open
Abstract
The transcription factor p53 is the most frequently impaired tumor suppressor in human cancers. In response to various stress stimuli, p53 activates transcription of genes that mediate its tumor-suppressive functions. Distinctive characteristics of p53 outlined here enable a well-defined program of genes involved in cell cycle arrest, apoptosis, senescence, differentiation, metabolism, autophagy, DNA repair, anti-viral response, and anti-metastatic functions, as well as facilitating autoregulation within the p53 network. This versatile, anti-cancer network governed chiefly by a single protein represents an immense opportunity for targeted cancer treatment, since about half of human tumors retain unmutated p53. During the last two decades, numerous compounds have been developed to block the interaction of p53 with the main negative regulator MDM2. However, small molecule inhibitors of MDM2 only induce a therapeutically desirable apoptotic response in a limited number of cancer types. Moreover, clinical trials of the MDM2 inhibitors as monotherapies have not met expectations and have revealed hematological toxicity as a characteristic adverse effect across this drug class. Currently, combination treatments are the leading strategy for enhancing efficacy and reducing adverse effects of MDM2 inhibitors. This review summarizes efforts to identify and test therapeutics that work synergistically with MDM2 inhibitors. Two main types of drugs have emerged among compounds used in the following combination treatments: first, modulators of the p53-regulated transcriptome (including chromatin modifiers), translatome, and proteome, and second, drugs targeting the downstream pathways such as apoptosis, cell cycle arrest, DNA repair, metabolic stress response, immune response, ferroptosis, and growth factor signaling. Here, we review the current literature in this field, while also highlighting overarching principles that could guide target selection in future combination treatments.
Collapse
Affiliation(s)
- Zdenek Andrysik
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joaquin M. Espinosa
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
3
|
Ni Q, Liu X, Song Z, Ma Y. Nickel-Catalyzed Cross-Coupling of Aziridines with Thioesters toward Atom-Economic Synthesis of β-Sulfanyl Amides. Org Lett 2024; 26:8457-8462. [PMID: 39331476 DOI: 10.1021/acs.orglett.4c02823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Thioesters have been recognized as a class of powerful bifunctional reagents, namely, great donors of acyl and sulfide moieties. However, such application in value-added synthesis is still very limited to date. Herein, a nickel-catalyzed cross-coupling reaction system of aziridines with thioesters was developed under redox-neutral and mild conditions. This catalytic method provides an atom-economic route for the synthesis of diverse β-sulfanyl amide derivatives with wide substrate scope (43 examples), good functional group tolerance, and regioselectivity.
Collapse
Affiliation(s)
- Qian Ni
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), Key Laboratory of Phytochemical R&D of Hunan Province, and Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Institute of Interdisciplinary Studies, College of Chemistry and Chemical Engineering, Hunan Normal University, 410081 Changsha, P. R. China
| | - Xianmao Liu
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), Key Laboratory of Phytochemical R&D of Hunan Province, and Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Institute of Interdisciplinary Studies, College of Chemistry and Chemical Engineering, Hunan Normal University, 410081 Changsha, P. R. China
| | - Zhiyong Song
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), Key Laboratory of Phytochemical R&D of Hunan Province, and Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Institute of Interdisciplinary Studies, College of Chemistry and Chemical Engineering, Hunan Normal University, 410081 Changsha, P. R. China
| | - Yuanhong Ma
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Ministry of Education), Key Laboratory of Phytochemical R&D of Hunan Province, and Key Laboratory of the Assembly and Application of Organic Functional Molecules of Hunan Province, Institute of Interdisciplinary Studies, College of Chemistry and Chemical Engineering, Hunan Normal University, 410081 Changsha, P. R. China
| |
Collapse
|
4
|
Liu Y, Ouyang L, Jiang S, Liang L, Chen Y, Mao C, Jiang Y, Cong L. PPP2R1A silencing suppresses LUAD progression by sensitizing cells to nelfinavir-induced apoptosis and pyroptosis. Cancer Cell Int 2024; 24:145. [PMID: 38654331 DOI: 10.1186/s12935-024-03321-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024] Open
Abstract
Lung adenocarcinoma is a major public health problem with the low 5-year survival rate (15%) among cancers. Aberrant alterations of meiotic genes, which have gained increased attention recently, might contribute to elevated tumor risks. However, systematic and comprehensive studies based on the relationship between meiotic genes and LUAD recurrence and treatment response are still lacking. In this manuscript, we first confirmed that the meiosis related prognostic model (MRPM) was strongly related to LUAD progression via LASSO-Cox regression analyses. Furthermore, we identified the role of PPP2R1A in LUAD, which showed more contributions to LUAD process compared with other meiotic genes in our prognostic model. Additionally, repression of PPP2R1A enhances cellular susceptibility to nelfinavir-induced apoptosis and pyroptosis. Collectively, our findings indicated that meiosis-related genes might be therapeutic targets in LUAD and provided crucial guidelines for LUAD clinical intervention.
Collapse
Affiliation(s)
- Yating Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Lianlian Ouyang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-Related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, 410011, China
| | - Shiyao Jiang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China
| | - Lu Liang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China
| | - Yuanbing Chen
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Chao Mao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yiqun Jiang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China.
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China.
| | - Li Cong
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China.
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
5
|
Carraro M, Are C, Azzena U, De Luca L, Gaspa S, Satta G, Holzer W, Pace V, Pisano L. Electronic Effects in a Green Protocol for (Hetero)Aryl-S Coupling. Molecules 2024; 29:1714. [PMID: 38675533 PMCID: PMC11051792 DOI: 10.3390/molecules29081714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Aryl and heteroaryl iodides have been efficiently converted into the corresponding thioacetates in cyclopentyl methyl ether (CPME), a green solvent, under Cu catalysis. The chemoselectivity of the reaction is mainly controlled by electronic factors, enabling the conversion of both electron-rich and electron-deficient substrates into the corresponding thioacetates in good to excellent yields. The products can be easily deprotected to the corresponding thiolates to carry out additional synthetic transformations in situ. Surprisingly, despite CPME's relatively low dielectric constant, the reaction rate significantly increased when conducted under microwave irradiation conditions. This synthetic methodology exhibits a remarkable tolerance to functional groups, mild reaction conditions, and a wide substrate scope, utilizing a safe and inexpensive CuI pre-catalyst in the green solvent CPME. A non-aqueous workup allowing for the complete recovery of both catalyst and solvent makes this approach an environmentally sustainable protocol for C(sp2) sulfur functionalization. Additionally, the reaction shows selective cross-coupling with iodides in competition with chlorides and bromides, allowing its use in multistep syntheses. To demonstrate the potential of this methodology, it was applied to the high-yield synthesis of a photochromic dithienylethene, where a selective synthesis had not been reported before.
Collapse
Affiliation(s)
- Massimo Carraro
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| | - Camillo Are
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| | - Ugo Azzena
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| | - Lidia De Luca
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| | - Silvia Gaspa
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| | - Giuseppe Satta
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| | - Wolfgang Holzer
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, A-1090 Vienna, Austria;
| | - Vittorio Pace
- Dipartimento di Chimica, Università di Torino, Via Giuria 7, 10125 Torino, Italy;
| | - Luisa Pisano
- Dipartimento di Scienze Chimiche, Fisiche, Matematiche e Naturali, Università degli Studi di Sassari, Via Vienna 2, 07100 Sassari, Italy; (M.C.); (L.D.L.); (S.G.)
| |
Collapse
|
6
|
Hossain MI, Asha AT, Hossain MA, Mahmud S, Chowdhury K, Mohiuddin RB, Nahar N, Sarker S, Napis S, Hossain MS, Mohiuddin A. Investigating the role of hypothetical protein (AAB33144.1) in HIV-1 virus pathogenicity: A comparative study with FDA-Approved inhibitor compounds through In silico analysis and molecular docking. Heliyon 2024; 10:e23183. [PMID: 38163140 PMCID: PMC10755284 DOI: 10.1016/j.heliyon.2023.e23183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Aim and objective Due to the a lot of unexplored proteins in HIV-1, this research aimed to explore the functional roles of a hypothetical protein (AAB33144.1) that might play a key role in HIV-1 pathogenicity. Methods The homologous protein was identified along with building and validating the 3D structure by searching several bioinformatics tools. Results Retroviral aspartyl protease and retropepsin like functional domains and motifs, folding pattern (cupredoxins), and subcellular localization in cytoplasmic membrane were determined as biological activity. Besides, the functional annotation revealed that the chosen hypothetical protein possessed protease-like activity. To validate our generated protein 3D structure, molecular docking was performed with five compounds where nelfinavir showed (-8.2 kcal/mol) best binding affinity against HXB2 viral protease (PDB ID: 7SJX) and main protease (PDB ID: 4EYR) protein. Conclusions This study suggests that the annotated hypothetical protein related to protease action, which may be useful in viral genetics and drug discovery.
Collapse
Affiliation(s)
- Md. Imran Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Anika Tabassum Asha
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Md. Arju Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Shahin Mahmud
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Kamal Chowdhury
- Biology Department, Claflin University, 400 Magnolia St, Orangeburg, SC 29115, USA
| | - Ramisa Binti Mohiuddin
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Nazneen Nahar
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Saborni Sarker
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | - Suhaimi Napis
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor D.E., Malaysia
| | - Md Sanower Hossain
- Centre for Sustainability of Mineral and Resource Recovery Technology (Pusat SMaRRT), Universiti Malaysia Pahang Al-Sultan Abdullah, Kuantan 26300, Malaysia
| | - A.K.M. Mohiuddin
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| |
Collapse
|
7
|
Peng Y, Zong Y, Wang D, Chen J, Chen ZS, Peng F, Liu Z. Current drugs for HIV-1: from challenges to potential in HIV/AIDS. Front Pharmacol 2023; 14:1294966. [PMID: 37954841 PMCID: PMC10637376 DOI: 10.3389/fphar.2023.1294966] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
The human immunodeficiency virus (HIV) persists in latently infected CD4+T cells and integrates with the host genome until cell death. Acquired immunodeficiency syndrome (AIDS) is associated with HIV-1. Possibly, treating HIV/AIDS is an essential but challenging clinical goal. This review provides a detailed account of the types and mechanisms of monotherapy and combination therapy against HIV-1 and describes nanoparticle and hydrogel delivery systems. In particular, the recently developed capsid inhibitor (Lenacapavir) and the Ainuovirine/tenofovir disoproxil fumarate/lamivudine combination (ACC008) are described. It is interestingly to note that the lack of the multipass transmembrane proteins serine incorporator 3 (SERINC3) and the multipass transmembrane proteins serine incorporator 5 (SERINC5) may be one of the reasons for the enhanced infectivity of HIV-1. This discovery of SERINC3 and SERINC5 provides new ideas for HIV-1 medication development. Therefore, we believe that in treating AIDS, antiviral medications should be rationally selected for pre-exposure and post-exposure prophylaxis to avoid the emergence of drug resistance. Attention should be paid to the research and development of new drugs to predict HIV mutations as accurately as possible and to develop immune antibodies to provide multiple guarantees for the cure of AIDS.
Collapse
Affiliation(s)
- Yuan Peng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yanjun Zong
- Department of Medical Microbiology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Dongfeng Wang
- School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Junbing Chen
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Fujun Peng
- School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Zhijun Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| |
Collapse
|
8
|
Miyazaki T, Hosogaya N, Fukushige Y, Takemori S, Morimoto S, Yamamoto H, Hori M, Ozawa Y, Shiko Y, Inaba Y, Kurokawa T, Hanaoka H, Iwanami S, Kim K, Iwami S, Watashi K, Miyazawa K, Umeyama T, Yamagoe S, Miyazaki Y, Wakita T, Sumiyoshi M, Hirayama T, Izumikawa K, Yanagihara K, Mukae H, Kawasuji H, Yamamoto Y, Tarumoto N, Ishii H, Ohno H, Yatera K, Kakeya H, Kichikawa Y, Kato Y, Matsumoto T, Saito M, Yotsuyanagi H, Kohno S. A Multicenter Randomized Controlled Trial To Evaluate the Efficacy and Safety of Nelfinavir in Patients with Mild COVID-19. Microbiol Spectr 2023; 11:e0431122. [PMID: 37140398 PMCID: PMC10269734 DOI: 10.1128/spectrum.04311-22] [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: 10/27/2022] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Nelfinavir, an orally administered inhibitor of human immunodeficiency virus protease, inhibits the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro. We conducted a randomized controlled trial to evaluate the clinical efficacy and safety of nelfinavir in patients with SARS-CoV-2 infection. We included unvaccinated asymptomatic or mildly symptomatic adult patients who tested positive for SARS-CoV-2 infection within 3 days before enrollment. The patients were randomly assigned (1:1) to receive oral nelfinavir (750 mg; thrice daily for 14 days) combined with standard-of-care or standard-of-care alone. The primary endpoint was the time to viral clearance, confirmed using quantitative reverse-transcription PCR by assessors blinded to the assigned treatment. A total of 123 patients (63 in the nelfinavir group and 60 in the control group) were included. The median time to viral clearance was 8.0 (95% confidence interval [CI], 7.0 to 12.0) days in the nelfinavir group and 8.0 (95% CI, 7.0 to 10.0) days in the control group, with no significant difference between the treatment groups (hazard ratio, 0.815; 95% CI, 0.563 to 1.182; P = 0.1870). Adverse events were reported in 47 (74.6%) and 20 (33.3%) patients in the nelfinavir and control groups, respectively. The most common adverse event in the nelfinavir group was diarrhea (49.2%). Nelfinavir did not reduce the time to viral clearance in this setting. Our findings indicate that nelfinavir should not be recommended in asymptomatic or mildly symptomatic patients infected with SARS-CoV-2. The study is registered with the Japan Registry of Clinical Trials (jRCT2071200023). IMPORTANCE The anti-HIV drug nelfinavir suppresses the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro. However, its efficacy in patients with COVID-19 has not been studied. We conducted a multicenter, randomized controlled trial to evaluate the efficacy and safety of orally administered nelfinavir in patients with asymptomatic or mildly symptomatic COVID-19. Compared to standard-of-care alone, nelfinavir (750 mg, thrice daily) did not reduce the time to viral clearance, viral load, or the time to resolution of symptoms. More patients had adverse events in the nelfinavir group than in the control group (74.6% [47/63 patients] versus 33.3% [20/60 patients]). Our clinical study provides evidence that nelfinavir, despite its antiviral effects on SARS-CoV-2 in vitro, should not be recommended for the treatment of patients with COVID-19 having no or mild symptoms.
Collapse
Affiliation(s)
- Taiga Miyazaki
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Nagasaki University, Nagasaki, Japan
| | - Naoki Hosogaya
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Yuri Fukushige
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Sachiko Takemori
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Shinpei Morimoto
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroshi Yamamoto
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Makoto Hori
- Division of Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Yoshihito Ozawa
- Division of Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Yuki Shiko
- Division of Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Yosuke Inaba
- Division of Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Tomoya Kurokawa
- Division of Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Hideki Hanaoka
- Division of Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Shoya Iwanami
- interdisciplinary Biology Laboratory (iBLab), Division of Biological Sciences, Graduate School of Science, Nagoya University, Aichi, Japan
| | - Kwangsu Kim
- interdisciplinary Biology Laboratory (iBLab), Division of Biological Sciences, Graduate School of Science, Nagoya University, Aichi, Japan
| | - Shingo Iwami
- interdisciplinary Biology Laboratory (iBLab), Division of Biological Sciences, Graduate School of Science, Nagoya University, Aichi, Japan
| | - Koichi Watashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ken Miyazawa
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takashi Umeyama
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Satoshi Yamagoe
- Section of Infectious Resource Coordination, Department of Research Resource, Center of Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yoshitsugu Miyazaki
- Department of Fungal Infection, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takaji Wakita
- National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Sumiyoshi
- Division of Respirology, Rheumatology, Infectious Diseases, and Neurology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tatsuro Hirayama
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
- Department of Pharmacotherapeutics, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Koichi Izumikawa
- Infection Control and Education Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Katsunori Yanagihara
- Department of Laboratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Hitoshi Kawasuji
- Department of Infectious Disease, Toyama University Hospital, Toyama, Japan
| | - Yoshihiro Yamamoto
- Department of Infectious Disease, Toyama University Hospital, Toyama, Japan
| | - Norihito Tarumoto
- Department of Infectious Disease and Infection Control, Saitama Medical University Hospital, Saitama, Japan
| | - Hiroshi Ishii
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Hideaki Ohno
- Department of Infectious Disease and Infection Control, Saitama Medical Center, Saitama, Japan
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, Hospital of the University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Hiroshi Kakeya
- Department of Infection Control Science, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | | | - Yasuyuki Kato
- Department of Infectious Diseases, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Tetsuya Matsumoto
- Department of Infectious Diseases, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Makoto Saito
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
9
|
Wang N, Wang H, Wan LX, Li XH, Zhou XL, Li JH, De Jonghe S, Schols D, Xu JB, Gao F. Visible-Light-Promoted Tandem Thiol-Ene Click Reaction/Transannular Cyclization and Regioselective Cyclopropane Ring-Opening to Construct Sulfur-Containing Euphorbia Diterpenes. Org Lett 2023; 25:597-602. [PMID: 36662155 DOI: 10.1021/acs.orglett.2c04116] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The biorelevant sulfur-containing Euphorbia diterpenes with scarce 5/7/6/3 premyrsinane- and 5/7/6 myrsinane-type backbones were easily constructed from naturally abundant lathyrane-type Euphorbia factor L3 by visible-light-triggered tandem thiol-ene click reaction/transannular cyclization and regioselective cyclopropane ring-opening. The selenide diterpene was also successfully obtained to verify the system universality. This concise synthesis route gives an efficient strategy for obtaining structurally diverse Euphorbia diterpenes under very mild conditions and provides a promising anti-HIV bioactive premyrsinane diterpene 3h.
Collapse
Affiliation(s)
- Neng Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Hang Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Lin-Xi Wan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Xiao-Huan Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Xian-Li Zhou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Jia-Hong Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Steven De Jonghe
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Herestraat 49, Box 1043, 3000 Leuven, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Herestraat 49, Box 1043, 3000 Leuven, Belgium
| | - Jin-Bu Xu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| | - Feng Gao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, People's Republic of China
| |
Collapse
|
10
|
Lee J, Kim J, Kang J, Lee HJ. COVID-19 drugs: potential interaction with ATP-binding cassette transporters P-glycoprotein and breast cancer resistance protein. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2023; 53:1-22. [PMID: 36320434 PMCID: PMC9607806 DOI: 10.1007/s40005-022-00596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/30/2022] [Indexed: 01/08/2023]
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2, has resulted in acute respiratory distress, fatal systemic manifestations (extrapulmonary as well as pulmonary), and premature mortality among many patients. Therapy for COVID-19 has focused on the treatment of symptoms and of acute inflammation (cytokine storm) and the prevention of viral infection. Although the mechanism of COVID-19 is not fully understood, potential clinical targets have been identified for pharmacological, immunological, and vaccinal approaches. Area covered Pharmacological approaches including drug repositioning have been a priority for initial COVID-19 therapy due to the time-consuming nature of the vaccine development process. COVID-19 drugs have been shown to manage the antiviral infection cycle (cell entry and replication of proteins and genomic RNA) and anti-inflammation. In this review, we evaluated the interaction of current COVID-19 drugs with two ATP-binding cassette transporters [P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP)] and potential drug-drug interactions (DDIs) among COVID-19 drugs, especially those associated with P-gp and BCRP efflux transporters. Expert opinion Overall, understanding the pharmacodynamic/pharmacokinetic DDIs of COVID-19 drugs can be useful for pharmacological therapy in COVID-19 patients.
Collapse
Affiliation(s)
- Jaeok Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Jihye Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Jiyeon Kang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Hwa Jeong Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| |
Collapse
|
11
|
Peng J, Ladumor MK, Unadkat JD. Estimation of Fetal-to-Maternal Unbound Steady-State Plasma Concentration Ratio of P-Glycoprotein and/or Breast Cancer Resistance Protein Substrate Drugs Using a Maternal-Fetal Physiologically Based Pharmacokinetic Model. Drug Metab Dispos 2022; 50:613-623. [PMID: 35149540 PMCID: PMC9073947 DOI: 10.1124/dmd.121.000733] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/18/2022] [Indexed: 11/22/2022] Open
Abstract
Pregnant women are frequently prescribed drugs to treat chronic diseases such as human immunodeficiency virus infection, but little is known about the benefits and risks of these drugs to the fetus that are driven by fetal drug exposure. The latter can be estimated by fetal-to-maternal unbound plasma concentration at steady state (Kp,uu,fetal). For drugs that are substrates of placental efflux transporters [i.e., P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP)], Kp,uu,fetal is expected to be <1. Here, we estimated the in vivo Kp,uu,fetal of selective P-gp and BCRP substrate drugs by maternal-fetal physiologically based pharmacokinetic (m-f-PBPK) modeling of umbilical vein (UV) plasma and maternal plasma (MP) concentrations obtained simultaneously at term from multiple maternal-fetal dyads. To do so, three drugs were selected: nelfinavir (P-gp substrate), efavirenz (BCRP substrate), and imatinib (P-gp/BCRP substrate). An m-f-PBPK model for each drug was developed and validated for the nonpregnant population and pregnant women using the Simcyp simulator (v20). Then, after incorporating placental passive diffusion clearance, the in vivo Kp,uu,fetal of the drug was estimated by adjusting the placental efflux clearance until the predicted UV/MP values best matched the observed data (Kp,uu,fetal) of nelfinavir = 0.41, efavirenz = 0.39, and imatinib = 0.35. Furthermore, Kp,uu,fetal of nelfinavir and efavirenz at gestational weeks (GWs) 25 and 15 were predicted to be 0.34 and 0.23 (GW25) and 0.33 and 0.27 (GW15). These Kp,uu,fetal values can be used to adjust dosing regimens of these drugs to optimize maternal-fetal drug therapy throughout pregnancy, to assess fetal benefits and risks of these dosing regimens, and to determine if these estimated in vivo Kp,uu,fetal values can be predicted from in vitro studies. SIGNIFICANCE STATEMENT: The in vivo fetal-to-maternal unbound steady-state plasma concentration ratio (Kp,uu,fetal) of nelfinavir [P-glycoprotein (P-gp) substrate], efavirenz [breast cancer resistance protein (BCRP) substrate], and imatinib (P-gp and BCRP substrate) was successfully estimated using maternal-fetal physiologically based pharmacokinetic (m-f-PBPK) modeling. These Kp,uu,fetal values can be used to adjust dosing regimens of these drugs to optimize maternal-fetal drug therapy throughout pregnancy, to assess fetal benefits and risks of these dosing regimens, and to determine if these estimated in vivo Kp,uu,fetal values can be predicted from in vitro studies.
Collapse
Affiliation(s)
- Jinfu Peng
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (J.P., M.K.L., J.D.U.) and Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China (J.P.)
| | - Mayur K Ladumor
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (J.P., M.K.L., J.D.U.) and Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China (J.P.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (J.P., M.K.L., J.D.U.) and Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China (J.P.)
| |
Collapse
|
12
|
Foo CS, Abdelnabi R, Kaptein SJF, Zhang X, Ter Horst S, Mols R, Delang L, Rocha-Pereira J, Coelmont L, Leyssen P, Dallmeier K, Vergote V, Heylen E, Vangeel L, Chatterjee AK, Annaert PP, Augustijns PF, De Jonghe S, Jochmans D, Gouwy M, Cambier S, Vandooren J, Proost P, van Laer C, Weynand B, Neyts J. HIV protease inhibitors Nelfinavir and Lopinavir/Ritonavir markedly improve lung pathology in SARS-CoV-2-infected Syrian hamsters despite lack of an antiviral effect. Antiviral Res 2022; 202:105311. [PMID: 35390430 PMCID: PMC8978445 DOI: 10.1016/j.antiviral.2022.105311] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 12/24/2022]
Abstract
Nelfinavir is an HIV protease inhibitor that has been widely prescribed as a component of highly active antiretroviral therapy, and has been reported to exert in vitro antiviral activity against SARS-CoV-2. We here assessed the effect of Nelfinavir in a SARS-CoV-2 infection model in hamsters. Despite the fact that Nelfinavir, [50 mg/kg twice daily (BID) for four consecutive days], did not reduce viral RNA load and infectious virus titres in the lung of infected animals, treatment resulted in a substantial improvement of SARS-CoV-2-induced lung pathology. This was accompanied by a dense infiltration of neutrophils in the lung interstitium which was similarly observed in non-infected hamsters. Nelfinavir resulted also in a marked increase in activated neutrophils in the blood, as observed in non-infected animals. Although Nelfinavir treatment did not alter the expression of chemoattractant receptors or adhesion molecules on human neutrophils, in vitro migration of human neutrophils to the major human neutrophil attractant CXCL8 was augmented by this protease inhibitor. Nelfinavir appears to induce an immunomodulatory effect associated with increasing neutrophil number and functionality, which may be linked to the marked improvement in SARS-CoV-2 lung pathology independent of its lack of antiviral activity. Since Nelfinavir is no longer used for the treatment of HIV, we studied the effect of two other HIV protease inhibitors, namely the combination Lopinavir/Ritonavir (Kaletra™) in this model. This combination resulted in a similar protective effect as Nelfinavir against SARS-CoV2 induced lung pathology in hamsters. Nelfinavir and lopinavir/ritonavir are FDA-approved HIV-protease inhibitors that inhibit SARS-CoV-2 replication in vitro. In hamsters, both compounds did not reduce viral loads but resulted in marked improvement of virus-induced lung pathology. Histopathology revealed a dense infiltration of neutrophils in the lungs of animals treated with these protease inhibitors. Nelfinavir treatment resulted also in a marked increase in activated neutrophils in the blood of treated hamsters. These data suggest that these compounds induce immunomodulatory effects, resulting in improvement of the lung pathology.
Collapse
Affiliation(s)
- Caroline S Foo
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Suzanne J F Kaptein
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Xin Zhang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Sebastiaan Ter Horst
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Raf Mols
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery & Disposition, Box 921, 3000, Leuven, Belgium
| | - Leen Delang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Joana Rocha-Pereira
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Pieter Leyssen
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Valentijn Vergote
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Elisabeth Heylen
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Laura Vangeel
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | | | - Pieter P Annaert
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery & Disposition, Box 921, 3000, Leuven, Belgium
| | - Patrick F Augustijns
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery & Disposition, Box 921, 3000, Leuven, Belgium
| | - Steven De Jonghe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Dirk Jochmans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Mieke Gouwy
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Immunology, B-3000, Leuven, Belgium
| | - Seppe Cambier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Immunology, B-3000, Leuven, Belgium
| | - Jennifer Vandooren
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Immunobiology, B-3000, Leuven, Belgium
| | - Paul Proost
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Immunology, B-3000, Leuven, Belgium
| | - Christine van Laer
- Clinical Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Division of Translational Cell and Tissue Research, B-3000, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium; GVN, Global Virus Network, Baltimore, MD, USA.
| |
Collapse
|
13
|
Krieg SC, Grimmer J, Kramer P, Bolte M, Kelm H, Manolikakes G. Oxyenamides as Versatile Building Blocks for a Highly Stereoselective One-Pot Synthesis of the 1,3-Diamino-2-ol-Scaffold Containing Three Continuous Stereocenters. Angew Chem Int Ed Engl 2021; 60:23667-23671. [PMID: 34463410 PMCID: PMC8597051 DOI: 10.1002/anie.202109752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/22/2021] [Indexed: 12/15/2022]
Abstract
A highly diastereoselective one-pot synthesis of the 1,3-diamino-2-alcohol unit bearing three continuous stereocenters is described. This method utilizes 2-oxyenamides as a novel type of building block for the rapid assembly of the 1,3-diamine scaffold containing an additional stereogenic oxygen functionality at the C2 position. A stereoselective preparation of the required (Z)-oxyenamides is reported as well.
Collapse
Affiliation(s)
- Sara-Cathrin Krieg
- Department of Chemistry, Technical University Kaiserslautern, Erwin-Schrödinger-Strasse Geb. 54, 67663, Kaiserslautern, Germany
| | - Jennifer Grimmer
- Department of Chemistry, Technical University Kaiserslautern, Erwin-Schrödinger-Strasse Geb. 54, 67663, Kaiserslautern, Germany
| | - Philipp Kramer
- Department of Chemistry, Technical University Kaiserslautern, Erwin-Schrödinger-Strasse Geb. 54, 67663, Kaiserslautern, Germany
| | - Michael Bolte
- Institute of Inorganic and Analytical Chemistry, Goethe University Frankfurt am Main, Max-von-Laue-Strasse 7, 60438, Frankfurt am Main, Germany
| | - Harald Kelm
- Department of Chemistry, Technical University Kaiserslautern, Erwin-Schrödinger-Strasse Geb. 54, 67663, Kaiserslautern, Germany
| | - Georg Manolikakes
- Department of Chemistry, Technical University Kaiserslautern, Erwin-Schrödinger-Strasse Geb. 54, 67663, Kaiserslautern, Germany
| |
Collapse
|
14
|
Krieg S, Grimmer J, Kramer P, Bolte M, Kelm H, Manolikakes G. Oxyenamide als vielseitige Bausteine für eine hochgradig stereoselektive Eintopf‐Synthese der 1,3‐Diamino‐2‐ol‐Einheit mit drei fortlaufenden Stereozentren. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sara‐Cathrin Krieg
- Fachbereich Chemie Technische Universität Kaiserslautern Erwin-Schrödinger-Straße, Geb. 54 67663 Kaiserslautern Deutschland
| | - Jennifer Grimmer
- Fachbereich Chemie Technische Universität Kaiserslautern Erwin-Schrödinger-Straße, Geb. 54 67663 Kaiserslautern Deutschland
| | - Philipp Kramer
- Fachbereich Chemie Technische Universität Kaiserslautern Erwin-Schrödinger-Straße, Geb. 54 67663 Kaiserslautern Deutschland
| | - Michael Bolte
- Institut für Anorganische und Analytische Chemie Goethe Universität Frankfurt am Main Max-von-Laue-Straße 7 60438 Frankfurt am Main Deutschland
| | - Harald Kelm
- Fachbereich Chemie Technische Universität Kaiserslautern Erwin-Schrödinger-Straße, Geb. 54 67663 Kaiserslautern Deutschland
| | - Georg Manolikakes
- Fachbereich Chemie Technische Universität Kaiserslautern Erwin-Schrödinger-Straße, Geb. 54 67663 Kaiserslautern Deutschland
| |
Collapse
|
15
|
Naidu ECS, Olojede SO, Lawal SK, Rennie CO, Azu OO. Nanoparticle delivery system, highly active antiretroviral therapy, and testicular morphology: The role of stereology. Pharmacol Res Perspect 2021; 9:e00776. [PMID: 34107163 PMCID: PMC8189564 DOI: 10.1002/prp2.776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
The conjugation of nanoparticles (NPs) with antiretroviral drugs is a drug delivery approach with great potential for managing HIV infections. Despite their promise, recent studies have highlighted the toxic effects of nanoparticles on testicular tissue and their impact on sperm morphology. This review explores the role of stereological techniques in assessing the testicular morphology in highly active antiretroviral therapy (HAART) when a nanoparticle drug delivery system is used. Also, NPs penetration and pharmacokinetics concerning the testicular tissue and blood-testis barrier form the vital part of this review. More so, various classes of NPs employed in biomedical and clinical research to deliver antiretroviral drugs were thoroughly discussed. In addition, considerations for minimizing nanoparticle-drugs toxicity, ensuring enhanced permeability of nanoparticles, maximizing drug efficacy, ensuring adequate bioavailability, and formulation of HAART-NPs fabrication are well discussed.
Collapse
Affiliation(s)
- Edwin Coleridge S. Naidu
- Discipline of Clinical AnatomySchool of Laboratory Medicine & Medical SciencesNelson R Mandela School of MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Samuel Oluwaseun Olojede
- Discipline of Clinical AnatomySchool of Laboratory Medicine & Medical SciencesNelson R Mandela School of MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Sodiq Kolawole Lawal
- Discipline of Clinical AnatomySchool of Laboratory Medicine & Medical SciencesNelson R Mandela School of MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Carmen Olivia Rennie
- Discipline of Clinical AnatomySchool of Laboratory Medicine & Medical SciencesNelson R Mandela School of MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Onyemaechi Okpara Azu
- Discipline of Clinical AnatomySchool of Laboratory Medicine & Medical SciencesNelson R Mandela School of MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
- Department of AnatomySchool of MedicineUniversity of NamibiaWindhoekNamibia
| |
Collapse
|
16
|
Addressing Antiretroviral Drug Resistance with Host-Targeting Drugs-First Steps towards Developing a Host-Targeting HIV-1 Assembly Inhibitor. Viruses 2021; 13:v13030451. [PMID: 33802145 PMCID: PMC8001593 DOI: 10.3390/v13030451] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
The concerning increase in HIV-1 resistance argues for prioritizing the development of host-targeting antiviral drugs because such drugs can offer high genetic barriers to the selection of drug-resistant viral variants. Targeting host proteins could also yield drugs that act on viral life cycle events that have proven elusive to inhibition, such as intracellular events of HIV-1 immature capsid assembly. Here, we review small molecule inhibitors identified primarily through HIV-1 self-assembly screens and describe how all act either narrowly post-entry or broadly on early and late events of the HIV-1 life cycle. We propose that a different screening approach could identify compounds that specifically inhibit HIV-1 Gag assembly, as was observed when a potent rabies virus inhibitor was identified using a host-catalyzed rabies assembly screen. As an example of this possibility, we discuss an antiretroviral small molecule recently identified using a screen that recapitulates the host-catalyzed HIV-1 capsid assembly pathway. This chemotype potently blocks HIV-1 replication in T cells by specifically inhibiting immature HIV-1 capsid assembly but fails to select for resistant viral variants over 37 passages, suggesting a host protein target. Development of such small molecules could yield novel host-targeting antiretroviral drugs and provide insight into chronic diseases resulting from dysregulation of host machinery targeted by these drugs.
Collapse
|
17
|
Dash P, Mohapatra S, Ghosh S, Nayak B. A Scoping Insight on Potential Prophylactics, Vaccines and Therapeutic Weaponry for the Ongoing Novel Coronavirus (COVID-19) Pandemic- A Comprehensive Review. Front Pharmacol 2021; 11:590154. [PMID: 33815095 PMCID: PMC8015872 DOI: 10.3389/fphar.2020.590154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/29/2020] [Indexed: 12/18/2022] Open
Abstract
The emergence of highly virulent CoVs (SARS-CoV-2), the etiologic agent of novel ongoing "COVID-19" pandemics has been marked as an alarming case of pneumonia posing a large global healthcare crisis of unprecedented magnitude. Currently, the COVID-19 outbreak has fueled an international demand in the biomedical field for the mitigation of the fast-spreading illness, all through the urgent deployment of safe, effective, and rational therapeutic strategies along with epidemiological control. Confronted with such contagious respiratory distress, the global population has taken significant steps towards a more robust strategy of containment and quarantine to halt the total number of positive cases but such a strategy can only delay the spread. A substantial number of potential vaccine candidates are undergoing multiple clinical trials to combat COVID-19 disease, includes live-attenuated, inactivated, viral-vectored based, sub-unit vaccines, DNA, mRNA, peptide, adjuvant, plant, and nanoparticle-based vaccines. However, there are no licensed anti-COVID-19 drugs/therapies or vaccines that have proven to work as more effective therapeutic candidates in open-label clinical trial studies. To counteract the infection (SARS-CoV-2), many people are under prolonged treatment of many chemical drugs that inhibit the PLpro activity (Ribavirin), viral proteases (Lopinavir/Ritonavir), RdRp activity (Favipiravir, Remdesivir), viral membrane fusion (Umifenovir, Chloroquine phosphate (CQ), Hydroxychloroquine phosphate (HCQ), IL-6 overexpression (Tocilizumab, Siltuximab, Sarilumab). Mesenchymal Stem Cell therapy and Convalescent Plasma Therapy have emerged as a promising therapeutic strategy against SARS-CoV-2 virion. On the other hand, repurposing previously designed antiviral agents with tolerable safety profile and efficacy could be the only promising approach and fast response to the novel virion. In addition, research institutions and corporations have commenced the redesign of the available therapeutic strategy to manage the global crisis. Herein, we present succinct information on selected anti-COVID-19 therapeutic medications repurposed to combat SARS-CoV-2 infection. Finally, this review will provide exhaustive detail on recent prophylactic strategies and ongoing clinical trials to curb this deadly pandemic, outlining the major therapeutic areas for researchers to step in.
Collapse
Affiliation(s)
| | | | | | - Bismita Nayak
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| |
Collapse
|
18
|
Identification of an Antiretroviral Small Molecule That Appears To Be a Host-Targeting Inhibitor of HIV-1 Assembly. J Virol 2021; 95:JVI.00883-20. [PMID: 33148797 PMCID: PMC7925099 DOI: 10.1128/jvi.00883-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/25/2020] [Indexed: 12/16/2022] Open
Abstract
Given the projected increase in multidrug-resistant HIV-1, there is an urgent need for development of antiretrovirals that act on virus life cycle stages not targeted by drugs currently in use. Host-targeting compounds are of particular interest because they can offer a high barrier to resistance. Here, we report identification of two related small molecules that inhibit HIV-1 late events, a part of the HIV-1 life cycle for which potent and specific inhibitors are lacking. This chemotype was discovered using cell-free protein synthesis and assembly systems that recapitulate intracellular host-catalyzed viral capsid assembly pathways. These compounds inhibit replication of HIV-1 in human T cell lines and peripheral blood mononuclear cells, and are effective against a primary isolate. They reduce virus production, likely by inhibiting a posttranslational step in HIV-1 Gag assembly. Notably, the compound colocalizes with HIV-1 Gag in situ; however, unexpectedly, selection experiments failed to identify compound-specific resistance mutations in gag or pol, even though known resistance mutations developed upon parallel nelfinavir selection. Thus, we hypothesized that instead of binding to Gag directly, these compounds localize to assembly intermediates, the intracellular multiprotein complexes containing Gag and host factors that form during immature HIV-1 capsid assembly. Indeed, imaging of infected cells shows compound colocalized with two host enzymes found in assembly intermediates, ABCE1 and DDX6, but not two host proteins found in other complexes. While the exact target and mechanism of action of this chemotype remain to be determined, our findings suggest that these compounds represent first-in-class, host-targeting inhibitors of intracellular events in HIV-1 assembly.IMPORTANCE The success of antiretroviral treatment for HIV-1 is at risk of being undermined by the growing problem of drug resistance. Thus, there is a need to identify antiretrovirals that act on viral life cycle stages not targeted by drugs in use, such as the events of HIV-1 Gag assembly. To address this gap, we developed a compound screen that recapitulates the intracellular events of HIV-1 assembly, including virus-host interactions that promote assembly. This effort led to the identification of a new chemotype that inhibits HIV-1 replication at nanomolar concentrations, likely by acting on assembly. This compound colocalized with Gag and two host enzymes that facilitate capsid assembly. However, resistance selection did not result in compound-specific mutations in gag, suggesting that the chemotype does not directly target Gag. We hypothesize that this chemotype represents a first-in-class inhibitor of virus production that acts by targeting a virus-host complex important for HIV-1 Gag assembly.
Collapse
|
19
|
de Almeida SMV, Santos Soares JC, Dos Santos KL, Alves JEF, Ribeiro AG, Jacob ÍTT, da Silva Ferreira CJ, Dos Santos JC, de Oliveira JF, de Carvalho Junior LB, de Lima MDCA. COVID-19 therapy: What weapons do we bring into battle? Bioorg Med Chem 2020; 28:115757. [PMID: 32992245 PMCID: PMC7481143 DOI: 10.1016/j.bmc.2020.115757] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/29/2020] [Accepted: 09/03/2020] [Indexed: 01/18/2023]
Abstract
Urgent treatments, in any modality, to fight SARS-CoV-2 infections are desired by society in general, by health professionals, by Estate-leaders and, mainly, by the scientific community, because one thing is certain amidst the numerous uncertainties regarding COVID-19: knowledge is the means to discover or to produce an effective treatment against this global disease. Scientists from several areas in the world are still committed to this mission, as shown by the accelerated scientific production in the first half of 2020 with over 25,000 published articles related to the new coronavirus. Three great lines of publications related to COVID-19 were identified for building this article: The first refers to knowledge production concerning the virus and pathophysiology of COVID-19; the second regards efforts to produce vaccines against SARS-CoV-2 at a speed without precedent in the history of science; the third comprehends the attempts to find a marketed drug that can be used to treat COVID-19 by drug repurposing. In this review, the drugs that have been repurposed so far are grouped according to their chemical class. Their structures will be presented to provide better understanding of their structural similarities and possible correlations with mechanisms of actions. This can help identifying anti-SARS-CoV-2 promising therapeutic agents.
Collapse
Affiliation(s)
- Sinara Mônica Vitalino de Almeida
- Laboratório de Biologia Molecular, Universidade de Pernambuco, Garanhuns, PE, Brazil; Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil; Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil.
| | - José Cleberson Santos Soares
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Keriolaine Lima Dos Santos
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Amélia Galdino Ribeiro
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Íris Trindade Tenório Jacob
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | | | - Jamerson Ferreira de Oliveira
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Maria do Carmo Alves de Lima
- Laboratório de Química e Inovação Terapêutica (LQIT) - Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
20
|
Verrier ER, Weiss A, Bach C, Heydmann L, Turon-Lagot V, Kopp A, El Saghire H, Crouchet E, Pessaux P, Garcia T, Pale P, Zeisel MB, Sureau C, Schuster C, Brino L, Baumert TF. Combined small molecule and loss-of-function screen uncovers estrogen receptor alpha and CAD as host factors for HDV infection and antiviral targets. Gut 2020; 69:158-167. [PMID: 30833451 PMCID: PMC6943243 DOI: 10.1136/gutjnl-2018-317065] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 01/24/2019] [Accepted: 02/10/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hepatitis D virus (HDV) is a circular RNA virus coinfecting hepatocytes with hepatitis B virus. Chronic hepatitis D results in severe liver disease and an increased risk of liver cancer. Efficient therapeutic approaches against HDV are absent. DESIGN Here, we combined an RNAi loss-of-function and small molecule screen to uncover host-dependency factors for HDV infection. RESULTS Functional screening unravelled the hypoxia-inducible factor (HIF)-signalling and insulin-resistance pathways, RNA polymerase II, glycosaminoglycan biosynthesis and the pyrimidine metabolism as virus-hepatocyte dependency networks. Validation studies in primary human hepatocytes identified the carbamoyl-phosphatesynthetase 2, aspartate transcarbamylase and dihydroorotase (CAD) enzyme and estrogen receptor alpha (encoded by ESR1) as key host factors for HDV life cycle. Mechanistic studies revealed that the two host factors are required for viral replication. Inhibition studies using N-(phosphonoacetyl)-L-aspartic acid and fulvestrant, specific CAD and ESR1 inhibitors, respectively, uncovered their impact as antiviral targets. CONCLUSION The discovery of HDV host-dependency factors elucidates the pathogenesis of viral disease biology and opens therapeutic strategies for HDV cure.
Collapse
Affiliation(s)
- Eloi R Verrier
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Amélie Weiss
- IGBMC, Plateforme de Criblage Haut-débit, UMR7104 CNRS U1258 Inserm, Illkirch, France
| | - Charlotte Bach
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Laura Heydmann
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Vincent Turon-Lagot
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Arnaud Kopp
- IGBMC, Plateforme de Criblage Haut-débit, UMR7104 CNRS U1258 Inserm, Illkirch, France
| | - Houssein El Saghire
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Emilie Crouchet
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Patrick Pessaux
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France,Institut Hospitalo-universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Thomas Garcia
- Laboratoire de Synthèse, Réactivité Organiques et Catalyse, Institut de Chimie, UMR 7177 CNRS, Université de Strasbourg, Strasbourg, France
| | - Patrick Pale
- Laboratoire de Synthèse, Réactivité Organiques et Catalyse, Institut de Chimie, UMR 7177 CNRS, Université de Strasbourg, Strasbourg, France
| | - Mirjam B Zeisel
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Camille Sureau
- INTS, Laboratoire de Virologie Moléculaire, Paris, France
| | - Catherine Schuster
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France
| | - Laurent Brino
- IGBMC, Plateforme de Criblage Haut-débit, UMR7104 CNRS U1258 Inserm, Illkirch, France
| | - Thomas F Baumert
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques UMR_S1110, F-67000 Strasbourg, France,Institut Hospitalo-universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France,Institut Universitaire de France, Paris, France
| |
Collapse
|
21
|
Han D, Tan J, Men J, Li C, Zhang X. Quantitative Structure Activity/Pharmacokinetics Relationship Studies of HIV-1 Protease Inhibitors Using Three Modelling Methods. Med Chem 2019; 17:396-406. [PMID: 31448716 DOI: 10.2174/1573406415666190826154505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/21/2019] [Accepted: 08/05/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND HIV-1 protease inhibitor (PIs) is a good choice for AIDS patients. Nevertheless, for PIs, there are several bugs in clinical application, like drug resistance, the large dose, the high costs and so on, among which, the poor pharmacokinetics property is one of the important reasons that leads to the failure of its clinical application. OBJECTIVE We aimed to build computational models for studying the relationship between PIs structure and its pharmacological activities. METHODS We collected experimental values of koff/Ki and structures of 50 PIs through a careful literature and database search. Quantitative structure activity/pharmacokinetics relationship (QSAR/QSPR) models were constructed by support vector machine (SVM), partial-least squares regression (PLSR) and back-propagation neural network (BPNN). RESULTS For QSAR models, SVM, PLSR and BPNN all generated reliable prediction models with the r2 of 0.688, 0.768 and 0.787, respectively, and r2pred of 0.748, 0.696 and 0.640, respectively. For QSPR models, the optimum models of SVM, PLSR and BPNN obtained the r2 of 0.952, 0.869 and 0.960, respectively, and the r2pred of 0.852, 0.628 and 0.814, respectively. CONCLUSION Among these three modelling methods, SVM showed superior ability than PLSR and BPNN both in QSAR/QSPR modelling of PIs, thus, we suspected that SVM was more suitable for predicting activities of PIs. In addition, 3D-MoRSE descriptors may have a tight relationship with the Ki values of PIs, and the GETAWAY descriptors have significant influence on both koff and Ki in PLSR equations.
Collapse
Affiliation(s)
- Dan Han
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Jianjun Tan
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Jingrui Men
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Chunhua Li
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Xiaoyi Zhang
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
22
|
Nagao Y, Hisanaga T, Utsumi T, Egami H, Kawato Y, Hamashima Y. Enantioselective Synthesis of Nelfinavir via Asymmetric Bromocyclization of Bisallylic Amide. J Org Chem 2018; 83:7290-7295. [DOI: 10.1021/acs.joc.8b00039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yoshihiro Nagao
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Tatsunari Hisanaga
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takahiro Utsumi
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiromichi Egami
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yuji Kawato
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoshitaka Hamashima
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
23
|
VanPatten S, Sun S, He M, Cheng KF, Altiti A, Papatheodorou A, Kowal C, Jeganathan V, Crawford JM, Bloom O, Volpe BT, Grant C, Meurice N, Coleman TR, Diamond B, Al-Abed Y. Amending HIV Drugs: A Novel Small-Molecule Approach To Target Lupus Anti-DNA Antibodies. J Med Chem 2016; 59:8859-8867. [PMID: 27603688 DOI: 10.1021/acs.jmedchem.6b00694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Systemic lupus erythematosus is an autoimmune disease that can affect numerous tissues and is characterized by the production of nuclear antigen-directed autoantibodies (e.g., anti-dsDNA). Using a combination of virtual and ELISA-based screens, we made the intriguing discovery that several HIV-protease inhibitors can function as decoy antigens to specifically inhibit the binding of anti-dsDNA antibodies to target antigens such as dsDNA and pentapeptide DWEYS. Computational modeling revealed that HIV-protease inhibitors comprised structural features present in DWEYS and predicted that analogues containing more flexible backbones would possess preferred binding characteristics. To address this, we reduced the internal amide backbone to improve flexibility, producing new small-molecule decoy antigens, which neutralize anti-dsDNA antibodies in vitro, in situ, and in vivo. Pharmacokinetic and SLE model studies demonstrated that peptidomimetic FISLE-412,1 a reduced HIV protease inhibitor analogue, was well-tolerated, altered serum reactivity to DWEYS, reduced glomeruli IgG deposition, preserved kidney histology, and delayed SLE onset in NZB/W F1 mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Christian Grant
- BioMedical Research Models, Inc. , 67 Millbrook Street, Worcester, Massachusetts 01606, United States
| | - Nathalie Meurice
- Department of Research, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | | | | | | |
Collapse
|
24
|
De Bruyn T, Stieger B, Augustijns PF, Annaert PP. Clearance Prediction of HIV Protease Inhibitors in Man: Role of Hepatic Uptake. J Pharm Sci 2016. [DOI: 10.1002/jps.24564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Cheng WH, Chang CH, Lu PL, Lin HC. Bilateral uveitis associated with concurrent administration of rifabutin and nelfinavir. Taiwan J Ophthalmol 2015; 5:187-188. [PMID: 29018696 PMCID: PMC5602138 DOI: 10.1016/j.tjo.2014.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/27/2014] [Accepted: 08/18/2014] [Indexed: 11/17/2022] Open
Abstract
Rifabutin-associated uveitis has been recognized as a dosage-dependent side effect. Previous studies have reported that clarithromycin or fluconazole may elevate concentrations of rifabutin through inhibition of metabolism through the cytochrome P-450 pathway. Nelfinavir is a protease inhibitor widely used in the treatment of human immunodeficiency virus (HIV) infection. The interactions between protease inhibitors and rifabutin have not been reported in clinical practice. Therefore, we present a case of bilateral uveitis associated with coadministration of rifabutin and nelfinavir. Uveitis did not subside until discontinuation of rifabutin. To our knowledge, this is the first report of uveitis with concurrent administration of rifabutin and nelfinavir. Our finding reminds us that rifabutin dosage should be reduced when it is administered with protease inhibitors.
Collapse
Affiliation(s)
- Wen-Hsin Cheng
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Hsien Chang
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Liang Lu
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsien-Chung Lin
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, Yuan's General Hospital, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Papanagnou P, Baltopoulos P, Tsironi M. Marketed nonsteroidal anti-inflammatory agents, antihypertensives, and human immunodeficiency virus protease inhibitors: as-yet-unused weapons of the oncologists' arsenal. Ther Clin Risk Manag 2015; 11:807-19. [PMID: 26056460 PMCID: PMC4445694 DOI: 10.2147/tcrm.s82049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Experimental data indicate that several pharmacological agents that have long been used for the management of various diseases unrelated to cancer exhibit profound in vitro and in vivo anticancer activity. This is of major clinical importance, since it would possibly aid in reassessing the therapeutic use of currently used agents for which clinicians already have experience. Further, this would obviate the time-consuming process required for the development and the approval of novel antineoplastic drugs. Herein, both pre-clinical and clinical data concerning the antineoplastic function of distinct commercially available pharmacological agents that are not currently used in the field of oncology, ie, nonsteroidal anti-inflammatory drugs, antihypertensive agents, and anti-human immunodeficiency virus agents inhibiting viral protease, are reviewed. The aim is to provide integrated information regarding not only the molecular basis of the antitumor function of these agents but also the applicability of the reevaluation of their therapeutic range in the clinical setting.
Collapse
Affiliation(s)
- Panagiota Papanagnou
- Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese, Sparta, Greece
| | - Panagiotis Baltopoulos
- Department of Sports Medicine and Biology of Physical Activity, Faculty of Physical Education and Sport Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Tsironi
- Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese, Sparta, Greece
| |
Collapse
|
27
|
Induction of suicidal erythrocyte death by nelfinavir. Toxins (Basel) 2015; 7:1616-28. [PMID: 26008229 PMCID: PMC4448164 DOI: 10.3390/toxins7051616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/28/2015] [Accepted: 05/05/2015] [Indexed: 12/31/2022] Open
Abstract
The HIV protease inhibitor, nelfinavir, primarily used for the treatment of HIV infections, has later been shown to be effective in various infectious diseases including malaria. Nelfinavir may trigger mitochondria-independent cell death. Erythrocytes may undergo eryptosis, a mitochondria-independent suicidal cell death characterized by cell shrinkage and phosphatidylserine translocation to the erythrocyte surface. Triggers of eryptosis include oxidative stress and increase of cytosolic Ca2+-activity ([Ca2+]i). During malaria, accelerated death of infected erythrocytes may decrease parasitemia and thus favorably influence the clinical course of the disease. In the present study, phosphatidylserine abundance at the cell surface was estimated from annexin V binding, cell volume from forward scatter, reactive oxidant species (ROS) from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence, and [Ca2+]i from Fluo3-fluorescence. A 48 h treatment of human erythrocytes with nelfinavir significantly increased the percentage of annexin-V-binding cells (≥5µg/mL), significantly decreased forward scatter (≥2.5µg/mL), significantly increased ROS abundance (10 µg/mL), and significantly increased [Ca2+]i (≥5 µg/mL). The up-regulation of annexin-V-binding following nelfinavir treatment was significantly blunted, but not abolished by either addition of the antioxidant N-acetylcysteine (1 mM) or removal of extracellular Ca2+. In conclusion, exposure of erythrocytes to nelfinavir induces oxidative stress and Ca2+ entry, thus leading to suicidal erythrocyte death characterized by erythrocyte shrinkage and erythrocyte membrane scrambling.
Collapse
|
28
|
Methaneethorn J, Kunyamee P, Jindasri W, Wattanasaovaluk W, Kraiboot A, Lohitnavy M. Pharmacokinetic modeling of simvastatin, nelfinavir and their interaction in humans. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2014:5715-8. [PMID: 25571293 DOI: 10.1109/embc.2014.6944925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Simvastatin, a commonly used HMG-CoA reductase inhibitor, is extensively metabolized by CYP3A4. Therefore, co-administration of simvastatin and CYP3A4 inhibitor can affect simvastatin pharmacokinetics. Nelfinavir, a protease inhibitor, and its major metabolite (M8) are known to be potent CYP3A4 inhibitors. When simvastatin and nelfinavir are co-administered, simvastatin pharmacokinetics is significantly altered and may result in an increased risk of rhabdomyolysis. OBJECTIVE To develop a mathematical model describing a drug-drug interaction between simvastatin and nelfinavir in humans. METHODS Eligible pharmacokinetic studies were selected from Pubmed database and concentration time course data were digitally extracted and used for model development. Compartmental pharmacokinetic models for simvastatin and nelfinavir were developed separately. A drug-drug interaction model of simvastatin and nelfinavir was subsequently developed using the prior information. Finally, the final drug-drug interaction modeled was validated against observed simvastatin concentrations. RESULTS Three compartmental pharmacokinetic models were successfully developed. Simvastatin pharmacokinetics was best described by a one compartment model for simvastatin linked to its active form, simvastatin hydroxy acid. Nelfinavir pharmacokinetics could be adequately described by a one compartment parent-metabolite model. Our final drug-drug interaction model predicted an increase in simvastatin exposure which is in line with clinical observations linking the simvastatin-nelfinavir combination to an increased risk of rhabdomyolysis. CONCLUSION Simvastatin-nelfinavir pharmacokinetic interaction can be explained by our final model. This model framework will be useful in further advanced developing other mechanism based drug-drug interaction model used to predict the risk of rhabdomyolysis occurrence in patients prescribed simvastatin and nelfinavir concurrently.
Collapse
|
29
|
Fang A, Valluri S, O’Sullivan MJ, Maupin R, Jones T, Delke I, Clax P. Safety and Pharmacokinetics of Nelfinavir During the Second and Third Trimesters of Pregnancy and Postpartum. HIV CLINICAL TRIALS 2015. [DOI: 10.1310/hct1301-46] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
30
|
Gerlach SL, Yeshak M, Göransson U, Roy U, Izadpanah R, Mondal D. Cycloviolacin O2 (CyO2) suppresses productive infection and augments the antiviral efficacy of nelfinavir in HIV-1 infected monocytic cells. Biopolymers 2013; 100:471-9. [DOI: 10.1002/bip.22325] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 04/02/2013] [Accepted: 05/28/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Samantha L. Gerlach
- Department of Pharmacology; Tulane University Health Sciences Center; 1430 Tulane Avenue New Orleans LA 70112
| | - Mariamawit Yeshak
- Division of Pharmacognosy; Department of Medicinal Chemistry, Uppsala University, Biomedical Centre; 574 S-751 23 Uppsala Sweden
| | - Ulf Göransson
- Division of Pharmacognosy; Department of Medicinal Chemistry, Uppsala University, Biomedical Centre; 574 S-751 23 Uppsala Sweden
| | - Upal Roy
- Department of Immunology; Florida International University; 11200 SW 8 Street AHC2 Miami FL 33199
| | - Reza Izadpanah
- Heart and Vascular Institute; Tulane University Health Sciences Center; 1430 Tulane Avenue New Orleans LA 70112
| | - Debasis Mondal
- Department of Pharmacology; Tulane University Health Sciences Center; 1430 Tulane Avenue New Orleans LA 70112
| |
Collapse
|
31
|
Buijsen J, Lammering G, Jansen RLH, Beets GL, Wals J, Sosef M, Den Boer MO, Leijtens J, Riedl RG, Theys J, Lambin P. Phase I trial of the combination of the Akt inhibitor nelfinavir and chemoradiation for locally advanced rectal cancer. Radiother Oncol 2013; 107:184-8. [PMID: 23647753 DOI: 10.1016/j.radonc.2013.03.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/08/2013] [Accepted: 03/17/2013] [Indexed: 11/25/2022]
Abstract
PURPOSE To investigate the toxicity of nelfinavir, administered during preoperative chemoradiotherapy (CRT) in patients with locally advanced rectal cancer. MATERIAL AND METHODS Twelve patients were treated with chemoradiotherapy to 50.4 Gy combined with capecitabine 825 mg/m(2) BID. Three dose levels (DL) of nelfinavir were tested: 750 mg BID (DL1), 1250 mg BID (DL2) and an intermediate level of 1000 mg BID (DL3). Surgery was performed between 8 and 10 weeks after completion of CRT. Primary endpoint was dose-limiting toxicity (DLT), defined as any grade 3 or higher non-hematological or grade 4 or higher hematological toxicity. RESULTS Eleven patients could be analyzed: 5 were treated in DL1, 3 in DL2 and 3 in DL3. The first 3 patients in DL1 did not develop a DLT. In DL2 one patient developed gr 3 diarrhea, 1 patient had gr 3 transaminase elevation and 1 patient had a gr 3 cholangitis with unknown cause. An intermediate dose level was tested in DL3. In this group 2 patients developed gr 3 diarrhea and 1 patient gr 3 transaminase elevation and gr 4 post-operative wound complication. Three patients achieved a pathological complete response (pCR). CONCLUSIONS Nelfinavir 750 mg BID was defined as the recommended phase II dose in combination with capecitabine and 50.4 Gy pre-operative radiotherapy in rectal cancer. First tumor response evaluations are promising, but a further phase II study is needed to get more information about efficacy of this treatment regimen.
Collapse
Affiliation(s)
- Jeroen Buijsen
- Department of Radiation Oncology (MAASTRO Clinic), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Naccarato M, Yoong D, la Porte C, Fong I. Amiodarone and concurrent antiretroviral therapy: a case report and review of the literature. Antivir Ther 2013; 19:329-39. [DOI: 10.3851/imp2715] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2013] [Indexed: 10/25/2022]
|
33
|
Schönthal AH. Endoplasmic reticulum stress: its role in disease and novel prospects for therapy. SCIENTIFICA 2012; 2012:857516. [PMID: 24278747 PMCID: PMC3820435 DOI: 10.6064/2012/857516] [Citation(s) in RCA: 218] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/12/2012] [Indexed: 05/19/2023]
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle required for lipid biosynthesis, calcium storage, and protein folding and processing. A number of physiological and pathological conditions, as well as a variety of pharmacological agents, are able to disturb proper ER function and thereby cause ER stress, which severely impairs protein folding and therefore poses the risk of proteotoxicity. Specific triggers for ER stress include, for example, particular intracellular alterations (e.g., calcium or redox imbalances), certain microenvironmental conditions (e.g., hypoglycemia, hypoxia, and acidosis), high-fat and high-sugar diet, a variety of natural compounds (e.g., thapsigargin, tunicamycin, and geldanamycin), and several prescription drugs (e.g., bortezomib/Velcade, celecoxib/Celebrex, and nelfinavir/Viracept). The cell reacts to ER stress by initiating a defensive process, called the unfolded protein response (UPR), which is comprised of cellular mechanisms aimed at adaptation and safeguarding cellular survival or, in cases of excessively severe stress, at initiation of apoptosis and elimination of the faulty cell. In recent years, this dichotomic stress response system has been linked to several human diseases, and efforts are underway to develop approaches to exploit ER stress mechanisms for therapy. For example, obesity and type 2 diabetes have been linked to ER stress-induced failure of insulin-producing pancreatic beta cells, and current research efforts are aimed at developing drugs that ameliorate cellular stress and thereby protect beta cell function. Other studies seek to pharmacologically aggravate chronic ER stress in cancer cells in order to enhance apoptosis and achieve tumor cell death. In the following, these principles will be presented and discussed.
Collapse
Affiliation(s)
- Axel H. Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, 2011 Zonal Avenue, HMR-405, Los Angeles, CA 90033, USA
| |
Collapse
|
34
|
Wallet MA, Reist CM, Williams JC, Appelberg S, Guiulfo GL, Gardner B, Sleasman JW, Goodenow MM. The HIV-1 protease inhibitor nelfinavir activates PP2 and inhibits MAPK signaling in macrophages: a pathway to reduce inflammation. J Leukoc Biol 2012; 92:795-805. [PMID: 22786868 DOI: 10.1189/jlb.0911447] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The HIV-1 PI NFV has off-target effects upon host enzymes, including inhibition of the 20S proteasome, resulting in activation of PP1. HIV-1-associated monocyte/macrophage activation, in part a result of systemically elevated levels of microbial products including LPS, is associated with risk of mortality, independent of viremia or CD4 T cell loss. This study tested the hypothesis that activation of protein phosphatases by NFV would reduce activation of monocytes/macrophages through dephosphorylation of signal transduction proteins. NFV uniquely blocked LPS-induced production by human monocyte-derived macrophages of the inflammatory cytokines TNF and IL-6, as well as sCD14. Although NFV failed to modulate NF-κB, NFV treatment reduced phosphorylation of AKT and MAPKs. Inhibition of PP2 with okadaic acid blocked the anti-inflammatory effect of NFV, whereas the PP1 inhibitor calyculin A failed to counter the anti-inflammatory effects of NFV. For in vivo studies, plasma sCD14 and LPS were monitored in a cohort of 31 pediatric HIV-1 patients for over 2 years of therapy. Therapy, including NFV, reduced sCD14 levels significantly compared with IDV or RTV, independent of ΔLPS levels, VL, CD4 T cell frequency, or age. The hypothesis was supported as NFV induced activation of PP2 in macrophages, resulting in disruption of inflammatory cell signaling pathways. In vivo evidence supports that NFV may offer beneficial effects independent of antiviral activity by reducing severity of chronic innate immune activation in HIV-1 infection.
Collapse
Affiliation(s)
- Mark A Wallet
- University of Florida, Department of Pathology, Immunology and Laboratory Medicine, Gainesville, FL, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Alfonso Y, Monzote L. HIV Protease Inhibitors: Effect on the Opportunistic Protozoan Parasites. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2011; 5:40-50. [PMID: 21629510 PMCID: PMC3103880 DOI: 10.2174/1874104501105010040] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 05/22/2010] [Accepted: 06/28/2010] [Indexed: 11/22/2022]
Abstract
The impact of highly active antiretroviral therapy (HAART) in the natural history of AIDS disease has been allowed to prolong the survival of people with HIV infection, particularly whose with increased HIV viral load. Additionally, the antiretroviral therapy could exert a certain degree of protection against parasitic diseases. A number of studies have been evidenced a decrease in the incidence of opportunistic parasitic infections in the era of HAART. Although these changes have been attributed to the restoration of cell-mediated immunity, induced by either non-nucleoside reverse transcriptase inhibitors or HIV protease inhibitors, in combination with at least two nucleoside reverse transcriptase inhibitors included in HAART, there are evidence that the control of these parasitic infections in HIV-positive persons under HAART, is also induced by the inhibition of the proteases of the parasites. This review focuses on the principal available data related with therapeutic HIV-protease inhibitors and their in vitro and in vivo effects on the opportunistic protozoan parasites.
Collapse
Affiliation(s)
- Yenisey Alfonso
- Parasitology Department, Institute of Tropical Medicine “Pedro Kourí”, Cuba
| | | |
Collapse
|
36
|
Ye ZW, Camus S, Augustijns P, Annaert P. Interaction of eight HIV protease inhibitors with the canalicular efflux transporter ABCC2 (MRP2) in sandwich-cultured rat and human hepatocytes. Biopharm Drug Dispos 2010; 31:178-88. [PMID: 20238377 DOI: 10.1002/bdd.701] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatotoxicity has been reported as a side-effect in some patients on HIV protease inhibitors (PI). Since transporter interaction has been implicated as a mechanism underlying drug-mediated hepatotoxicity and drug-drug interactions, the interaction of PI with the hepatic canalicular efflux transporter ABCC2 (MRP2; multidrug resistance associated protein-2) was studied. Interaction with ABCC2/Abcc2 was evaluated in human and rat sandwich-cultured hepatocytes using 5(6)-carboxy-2',7'-dichlorofluorescein (CDF) as substrate. In rat hepatocytes, interaction with estradiol-17-beta-D-glucuronide (E17G) efflux was also studied. In human hepatocytes, saquinavir, ritonavir and atazanavir were the most efficient inhibitors of ABCC2-mediated biliary excretion of CDF, whereas in rat hepatocytes indinavir, lopinavir and nelfinavir were the most efficient. No species-similarity was found for ABCC2/Abcc2 inhibition. In rat hepatocytes, the effects on Abcc2 were substrate-dependent as inhibition of biliary excretion of E17G was most pronounced for saquinavir (completely blocked), amprenavir (82% inhibition) and indinavir (68% inhibition). In conclusion, several HIV PI showed substantial ABCC2 inhibition, which, combined with the effects of PI on other hepatobiliary disposition mechanisms, will determine the clinical relevance of these in vitro interaction data.
Collapse
Affiliation(s)
- Zhi-Wei Ye
- Laboratory for Pharmacotechnology and Biopharmacy, Department of Pharmaceutical Sciences, Katholieke Universiteit Leuven, O&N2, Herestraat 49-bus-921, 3000 Leuven, Belgium
| | | | | | | |
Collapse
|
37
|
Damle BD, Uderman H, Biswas P, Crownover P, Lin C, Glue P. Influence of CYP2C19 polymorphism on the pharmacokinetics of nelfinavir and its active metabolite. Br J Clin Pharmacol 2010; 68:682-9. [PMID: 19916992 DOI: 10.1111/j.1365-2125.2009.03499.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
AIMS This study reports the pharmacokinetics of nelfinavir, its active metabolite, M8, and active moiety (nelfinavir + M8) in volunteers genotyped for CYP2C19 as extensive metabolizer (*1*1; n = 38), heterozygous poor metabolizer (PM) (*1*2; n = 22) and homozygous PM (*2*2; n = 6). METHODS Subjects received nelfinavir at normal dose (3.5 days of 1250 mg q12h) or high dose (1250 mg q12h for 3 days and single dose of 3125 mg on day 4). Steady-state plasma samples were analysed by high-performance liquid chromatography/ultraviolet assay to determine pharmacokinetics. RESULTS At steady state, the mean C(max) was 42% [95% confidence interval (CI) 19, 69] and 63% (95% CI 20, 122) higher, and mean AUC was 51% (95% CI 24, 83) and 85% (95% CI 32, 159) higher for *1*2 and *2*2 compared with *1*1 subjects, respectively. For M8, the mean C(max) and AUC were 35% (95% CI 6, 55) and 33% (95% CI -3, 56), respectively, lower for *1*2 compared with *1*1 subjects. M8 was not detectable in *2*2 subjects. The mean C(max) and AUC values for the active moiety were higher by 30-35% for the *1*2 and *2*2 compared with *1*1 subjects. CONCLUSIONS Mutation in CYP2C19 increased the systemic exposure of nelfinavir and reduced the exposure of M8. No significant differences were noted among the heterozygous (*1*2) and homozygous (*2*2) PMs. These changes are not considered to be clinically relevant and hence the use of nelfinavir does not require prior assessment of CYP2C19 genotype.
Collapse
Affiliation(s)
- Bharat D Damle
- Pfizer Inc., Global Medical Research & Development, 685 Third Avenue, 685/13/55, New York, NY 10017, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Wong HL, Chattopadhyay N, Wu XY, Bendayan R. Nanotechnology applications for improved delivery of antiretroviral drugs to the brain. Adv Drug Deliv Rev 2010; 62:503-17. [PMID: 19914319 DOI: 10.1016/j.addr.2009.11.020] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 09/14/2009] [Indexed: 01/16/2023]
Abstract
Human immunodeficiency virus (HIV) can gain access to the central nervous system during the early course of primary infection. Once in the brain compartment the virus actively replicates to form an independent viral reservoir, resulting in debilitating neurological complications, latent infection and drug resistance. Current antiretroviral drugs (ARVs) often fail to effectively reduce the HIV viral load in the brain. This, in part, is due to the poor transport of many ARVs, in particular protease inhibitors, across the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSBF). Studies have shown that nanocarriers including polymeric nanoparticles, liposomes, solid lipid nanoparticles (SLN) and micelles can increase the local drug concentration gradients, facilitate drug transport into the brain via endocytotic pathways and inhibit the ATP-binding cassette (ABC) transporters expressed at the barrier sites. By delivering ARVs with nanocarriers, significant increase in the drug bioavailability to the brain is expected to be achieved. Recent studies show that the specificity and efficiency of ARVs delivery can be further enhanced by using nanocarriers with specific brain targeting, cell penetrating ligands or ABC-transporters inhibitors. Future research should focus on achieving brain delivery of ARVs in a safe, efficient, and yet cost-effective manner.
Collapse
|
39
|
Annaert P, Ye Z, Stieger B, Augustijns P. Interaction of HIV protease inhibitors with OATP1B1, 1B3, and 2B1. Xenobiotica 2010; 40:163-76. [DOI: 10.3109/00498250903509375] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
40
|
Kharasch ED, Walker A, Whittington D, Hoffer C, Bedynek PS. Methadone metabolism and clearance are induced by nelfinavir despite inhibition of cytochrome P4503A (CYP3A) activity. Drug Alcohol Depend 2009; 101:158-68. [PMID: 19232844 PMCID: PMC3582041 DOI: 10.1016/j.drugalcdep.2008.12.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 11/15/2008] [Accepted: 12/06/2008] [Indexed: 11/30/2022]
Abstract
BACKGROUND Methadone plasma concentrations are decreased by nelfinavir. Methadone clearance and the drug interactions have been attributed to CYP3A4, but actual mechanisms of methadone clearance and the nelfinavir interaction are unknown. We assessed nelfinavir effects on methadone pharmacokinetics and pharmacodynamics, intestinal and hepatic CYP3A4/5 activity, and intestinal P-glycoprotein transport activity. CYP3A4/5 and transporters were assessed using alfentanil and fexofenadine, respectively. METHODS Twelve healthy HIV-negative volunteers underwent a sequential crossover. On three consecutive days they received oral alfentanil plus fexofenadine, intravenous alfentanil, and intravenous plus oral methadone. This was repeated after nelfinavir. Plasma and urine analytes were measured by mass spectrometry. Opioid effects were measured by pupil diameter change (miosis). RESULTS Nelfinavir decreased intravenous and oral methadone plasma concentrations 40-50%. Systemic clearance, hepatic clearance, and hepatic extraction all increased 1.6- and 2-fold, respectively, for R- and S-methadone; apparent oral clearance increased 1.7- and 1.9-fold. Nelfinavir stereoselectively increased (S>R) methadone metabolism and metabolite formation clearance, and methadone renal clearance. Methadone bioavailability and P-glycoprotein activity were minimally affected. Nelfinavir decreased alfentanil systemic and apparent oral clearances 50 and 76%, respectively. Nelfinavir appeared to shift the methadone plasma concentration-effect (miosis) curve leftward and upward. CONCLUSIONS Nelfinavir induced methadone clearance by increasing renal clearance, and more so by stereoselectively increasing hepatic metabolism, extraction and clearance. Induction occurred despite 50% inhibition of hepatic CYP3A4/5 activity and more than 75% inhibition of first-pass CYP3A4/5 activity, suggesting little or no role for CYP3A in clinical methadone disposition. Nelfinavir may alter methadone pharmacodynamics, increasing clinical effects.
Collapse
Affiliation(s)
- Evan D. Kharasch
- Department of Anesthesiology, and Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, 63110 USA
| | - Alysa Walker
- Department of Anesthesiology, University of Washington. Seattle, WA, 98195 USA
| | - Dale Whittington
- Department of Anesthesiology, University of Washington. Seattle, WA, 98195 USA
| | - Christine Hoffer
- Department of Anesthesiology, University of Washington. Seattle, WA, 98195 USA
| | | |
Collapse
|
41
|
Damle B, Fosser C, Ito K, Tran A, Clax P, Uderman H, Glue P. Effects of standard and supratherapeutic doses of nelfinavir on cardiac repolarization: a thorough QT study. J Clin Pharmacol 2009; 49:291-300. [PMID: 19246729 DOI: 10.1177/0091270008329551] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This was a randomized, 4-way crossover, third-party-blinded study in 68 healthy subjects to assess the effect of nelfinavir on QTc interval. Treatments included (A) nelfinavir 1250 mg every 12 hours on days 1-4, (B) nelfinavir 1250 mg every 12 hours on days 1-3 plus 3125 mg on day 4, (C) placebo, and (D) moxifloxacin 400 mg every 24 hours on days 1-4. Pharmacokinetics and triplicate 12-lead electrocardiograms were performed over 12 hours on days 1 and 4. Time-matched, placebo-subtracted, baseline-adjusted changes in QT intervals with Fridericia's (QTcF) correction were determined following nelfinavir and moxifloxacin administration. Neither dose of nelfinavir had a clinically relevant effect on the QTcF interval on day 4 (primary endpoint) and day 1 because at every time point the upper 90% confidence limit was below 10 milliseconds and, furthermore, the mean difference was below 5 milliseconds. Additionally, there was no clinically relevant effect on QTcB (Bazett's correction), uncorrected QT, or the RR interval on days 1 or 4. Pharmacokinetics confirmed adequate systemic exposure to nelfinavir and moxifloxacin. While nelfinavir exposure was higher in poor compared with extensive metabolizers of CYP2C19 isozyme, there were no corresponding significant differences in QTcF change from placebo. At clinically relevant, doses nelfinavir is unlikely to cause QTc prolongation.
Collapse
Affiliation(s)
- Bharat Damle
- Pfizer Global Research & Development, Pfizer Inc, 685 Third Ave., 685/13/55, New York, NY 10017, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Audelin AM, Lohse N, Obel N, Gerstoft J, Jørgensen LB. The incidence rate of HIV type-1 drug resistance in patients on antiretroviral therapy: a nationwide population-based Danish cohort study 1999–2005. Antivir Ther 2009; 14:995-1000. [DOI: 10.3851/imp1412] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
43
|
Ronaldson PT, Persidsky Y, Bendayan R. Regulation of ABC membrane transporters in glial cells: Relevance to the pharmacotherapy of brain HIV-1 infection. Glia 2008; 56:1711-35. [DOI: 10.1002/glia.20725] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
44
|
Roustit M, Jlaiel M, Leclercq P, Stanke-Labesque F. Pharmacokinetics and therapeutic drug monitoring of antiretrovirals in pregnant women. Br J Clin Pharmacol 2008; 66:179-95. [PMID: 18537960 PMCID: PMC2492933 DOI: 10.1111/j.1365-2125.2008.03220.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 05/06/2008] [Indexed: 01/07/2023] Open
Abstract
Highly active antiretroviral therapy is recommended for HIV-infected pregnant women to prevent mother-to-child transmission. The specific physiological background induced by pregnancy leads to significant changes in maternal pharmacokinetics, suggesting potential variability in plasma concentrations of antiretrovirals during gestation. Therapeutic drug monitoring (TDM) of protease inhibitors (PIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) is recommended in certain situations, including pregnancy, but its systematic use in HIV-infected pregnant women remains controversial. This review provides an update of the pharmacokinetic data available for PIs and NNRTIs in pregnant women and highlights the clinical interest of systematic TDM of certain antiretroviral drugs during pregnancy, including nevirapine, nelfinavir, saquinavir, indinavir and lopinavir.
Collapse
Affiliation(s)
- Matthieu Roustit
- CHU de Grenoble, Laboratoire de PharmacologieBP217, Grenoble, France
- INSERM ERI 17, Laboratoire HP2BP217, Grenoble, France
| | - Malik Jlaiel
- CHU de Grenoble, Laboratoire de PharmacologieBP217, Grenoble, France
| | - Pascale Leclercq
- CHU de Grenoble, Clinique Infectiologie–CISIHBP217, Grenoble, France
| | - Françoise Stanke-Labesque
- CHU de Grenoble, Laboratoire de PharmacologieBP217, Grenoble, France
- INSERM ERI 17, Laboratoire HP2BP217, Grenoble, France
- Université Joseph Fourier, Faculté de Médecine IFR1BP217, Grenoble, France
| |
Collapse
|
45
|
Fang AF, Damle BD, LaBadie RR, Crownover PH, Hewlett D, Glue PW. Significant decrease in nelfinavir systemic exposure after omeprazole coadministration in healthy subjects. Pharmacotherapy 2008; 28:42-50. [PMID: 18154473 DOI: 10.1592/phco.28.1.42] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
STUDY OBJECTIVES To assess the effect of omeprazole on the multiple-dose (steady-state) pharmacokinetics and safety of nelfinavir, and to evaluate the safety and tolerability of nelfinavir when administered alone and with omeprazole. DESIGN Open-label, two-period, single-fixed-sequence study. SETTING Clinical research unit of a large, teaching hospital. PARTICIPANTS Twenty healthy volunteers (mean age 26 +/- 9 yrs, range 18-48 yrs). Intervention. Subjects received nelfinavir 1250 mg every 12 hours for 4 days (period 1). After a 7-day washout period, subjects were coadministered nelfinavir 1250 mg every 12 hours and omeprazole 40 mg every 24 hours for 4 days (period 2). MEASUREMENTS AND MAIN RESULTS The pharmacokinetics of nelfinavir and its active metabolite M8 were determined on day 4 of both periods. Plasma samples were assayed by a high-performance liquid chromatography-ultraviolet method for nelfinavir and M8 concentrations, and noncompartmental pharmacokinetic analysis was performed by using analytical software. In the presence of omeprazole, nelfinavir area under the concentration-time curve over the dosing interval (AUC(tau)), maximum observed plasma concentration (C(max)), and minimum observed plasma concentration (C(min)) were reduced by an average of 36%, 37%, and 39%, respectively, relative to administration of nelfinavir alone. The AUC(tau), C(max), and C(min) of M8 were reduced by an average of 92%, 89%, and 75%, respectively. The slopes of the terminal elimination phase of nelfinavir and M8 plasma concentration-time curves were similar between treatments. Nelfinavir was well tolerated when administered alone and when coadministered with omeprazole. CONCLUSION The observed reduction in the systemic exposure to both nelfinavir and its active metabolite M8 after coadministration with omeprazole could result in loss of virologic control and potential emergence of drug resistance. Hence, omeprazole should not be coadministered to patients taking nelfinavir.
Collapse
|