1
|
El-Qassas J, Abd El-Atti M, El-Badri N. Harnessing the potency of scorpion venom-derived proteins: applications in cancer therapy. BIORESOUR BIOPROCESS 2024; 11:93. [PMID: 39361208 PMCID: PMC11450130 DOI: 10.1186/s40643-024-00805-0] [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: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Despite breakthroughs in the development of cancer diagnosis and therapy, most current therapeutic approaches lack precise specificity and sensitivity, resulting in damage to healthy cells. Selective delivery of anti-cancer agents is thus an important goal of cancer therapy. Scorpion venom (SV) and/or body parts have been used since early civilizations for medicinal purposes, and in cultures, SV is still applied to the treatment of several diseases including cancer. SV contains numerous active micro and macromolecules with diverse pharmacological effects. These include potent anti-microbial, anti-viral, anti-inflammatory, and anti-cancer properties. This review focuses on the recent advances of SV-derived peptides as promising anti-cancer agents and their diagnostic and therapeutic potential applications in cancers such as glioma, breast cancer, prostate cancer, and colon cancer. Well-characterized SV-derived peptides are thus needed to serve as potent and selective adjuvant therapy for cancer, to significantly enhance the patients' survival and wellbeing.
Collapse
Affiliation(s)
- Jihad El-Qassas
- Department of Zoology, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, 6th of October City, Giza, 12578, Egypt
| | - Mahmoud Abd El-Atti
- Department of Zoology, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, 6th of October City, Giza, 12578, Egypt.
| |
Collapse
|
2
|
Seyfizadeh N, Kalbermatter D, Imhof T, Ries M, Müller C, Jenner L, Blumenschein E, Yendrzheyevskiy A, Grün F, Moog K, Eckert D, Engel R, Diebolder P, Chami M, Krauss J, Schaller T, Arndt M. Development of a highly effective combination monoclonal antibody therapy against Herpes simplex virus. J Biomed Sci 2024; 31:56. [PMID: 38807208 PMCID: PMC11134845 DOI: 10.1186/s12929-024-01045-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Infections with Herpes simplex virus (HSV)-1 or -2 usually present as mild chronic recurrent disease, however in rare cases can result in life-threatening conditions with a large spectrum of pathology. Monoclonal antibody therapy has great potential especially to treat infections with virus resistant to standard therapies. HDIT101, a humanized IgG targeting HSV-1/2 gB was previously investigated in phase 2 clinical trials. The aim of this study was to develop a next-generation therapy by combining different antiviral monoclonal antibodies. METHODS A lymph-node derived phage display library (LYNDAL) was screened against recombinant gB from Herpes simplex virus (HSV) -1 and HDIT102 scFv was selected for its binding characteristics using bio-layer interferometry. HDIT102 was further developed as fully human IgG and tested alone or in combination with HDIT101, a clinically tested humanized anti-HSV IgG, in vitro and in vivo. T-cell stimulating activities by antigen-presenting cells treated with IgG-HSV immune complexes were analyzed using primary human cells. To determine the epitopes, the cryo-EM structures of HDIT101 or HDIT102 Fab bound to HSV-1F as well as HSV-2G gB protein were solved at resolutions < 3.5 Å. RESULTS HDIT102 Fab showed strong binding to HSV-1F gB with Kd of 8.95 × 10-11 M and to HSV-2G gB with Kd of 3.29 × 10-11 M. Neutralization of cell-free virus and inhibition of cell-to-cell spread were comparable between HDIT101 and HDIT102. Both antibodies induced internalization of gB from the cell surface into acidic endosomes by binding distinct epitopes in domain I of gB and compete for binding. CryoEM analyses revealed the ability to form heterogenic immune complexes consisting of two HDIT102 and one HDIT101 Fab bound to one gB trimeric molecule. Both antibodies mediated antibody-dependent phagocytosis by antigen presenting cells which stimulated autologous T-cell activation. In vivo, the combination of HDIT101 and HDIT102 demonstrated synergistic effects on survival and clinical outcome in immunocompetent BALB/cOlaHsd mice. CONCLUSION This biochemical and immunological study showcases the potential of an effective combination therapy with two monoclonal anti-gB IgGs for the treatment of HSV-1/2 induced disease conditions.
Collapse
Affiliation(s)
- Narges Seyfizadeh
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - David Kalbermatter
- Biozentrum, University of Basel, Spitalstrasse 41, Basel, CH - 4056, Switzerland
- Present address: University of Bern, Institute of Anatomy, Balzerstrasse 2, Bern, 3012, Switzerland
| | - Thomas Imhof
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Moritz Ries
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Christian Müller
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Leonie Jenner
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | | | | | - Frank Grün
- Vanudis GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Kevin Moog
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Daniel Eckert
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Ronja Engel
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Philipp Diebolder
- National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, Heidelberg, 69120, Germany
- Present address: Bio-Rad AbD Serotec GmbH, Anna-Sigmund-Str. 5, Neuried, 82061, Germany
| | - Mohamed Chami
- Biozentrum, University of Basel, Spitalstrasse 41, Basel, CH - 4056, Switzerland
| | - Jürgen Krauss
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Torsten Schaller
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany
| | - Michaela Arndt
- Heidelberg ImmunoTherapeutics GmbH, Max-Jarecki Str. 21, Heidelberg, 69115, Germany.
| |
Collapse
|
3
|
Islam MR, Islam Sovon MS, Amena U, Rahman M, Hosen ME, Kumer A, Bourhia M, Bin Jardan YA, Ibenmoussa S, Wondmie GF. Ligand-based drug design against Herpes Simplex Virus-1 capsid protein by modification of limonene through in silico approaches. Sci Rep 2024; 14:9828. [PMID: 38684729 PMCID: PMC11058824 DOI: 10.1038/s41598-024-59577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
The pharmacological effects of limonene, especially their derivatives, are currently at the forefront of research for drug development and discovery as well and structure-based drug design using huge chemical libraries are already widespread in the early stages of therapeutic and drug development. Here, various limonene derivatives are studied computationally for their potential utilization against the capsid protein of Herpes Simplex Virus-1. Firstly, limonene derivatives were designed by structural modification followed by conducting a molecular docking experiment against the capsid protein of Herpes Simplex Virus-1. In this research, the obtained molecular docking score exhibited better efficiency against the capsid protein of Herpes Simplex Virus-1 and hence we conducted further in silico investigation including molecular dynamic simulation, quantum calculation, and ADMET analysis. Molecular docking experiment has documented that Ligands 02 and 03 had much better binding affinities (- 7.4 kcal/mol and - 7.1 kcal/mol) to capsid protein of Herpes Simplex Virus-1 than Standard Acyclovir (- 6.5 kcal/mol). Upon further investigation, the binding affinities of primary limonene were observed to be slightly poor. But including the various functional groups also increases the affinities and capacity to prevent viral infection of the capsid protein of Herpes Simplex Virus-1. Then, the molecular dynamic simulation confirmed that the mentioned ligands might be stable during the formation of drug-protein complexes. Finally, the analysis of ADMET was essential in establishing them as safe and human-useable prospective chemicals. According to the present findings, limonene derivatives might be a promising candidate against the capsid protein of Herpes Simplex Virus-1 which ultimately inhibits Herpes Simplex Virus-induced encephalitis that causes interventions in brain inflammation. Our findings suggested further experimental screening to determine their practical value and utility.
Collapse
Affiliation(s)
- Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh, 1207
| | | | - Ummy Amena
- Department of Pharmacy, Faculty of Life & Earth Sciences, Jagannath University, Dhaka, Bangladesh
| | - Miadur Rahman
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1219, Bangladesh
| | - Md Eram Hosen
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Ajoy Kumer
- Department of Chemistry, College of Arts and Sciences, International University of Business Agriculture and Technology (IUBAT), Dhaka, 1216, Bangladesh
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences in Saveetha Medical College and Hospital, Chennai, India
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco.
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 11451, Riyadh, Saudi Arabia
| | - Samir Ibenmoussa
- Laboratory of Therapeutic and Organic Chemistry, Faculty of Pharmacy, University of Montpellier, 34000, Montpellier, France
| | | |
Collapse
|
4
|
Zhou N, Zheng D, You Q, Chen T, Jiang J, Shen W, Zhang D, Liu J, Chen D, Hu K. Therapeutic Potential of Biochanin A in Herpes Simplex Keratitis. Pharmaceuticals (Basel) 2023; 16:1240. [PMID: 37765049 PMCID: PMC10536220 DOI: 10.3390/ph16091240] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Herpes simplex keratitis (HSK) is a blinding eye disease that is initiated by the herpes simplex virus type 1 (HSV-1). Resistance to acyclovir (ACV) and the side effects of corticosteroid drugs have become concerning issues, so it is crucial to develop new antivirals for treating HSK. In this study, we report that biochanin A (BCA), a naturally occurring flavonoid compound, provides multifaceted protective effects with anti-viral, anti-inflammatory, anti-oxidative stress and anti-apoptotic activities to alleviate HSK. The results show that BCA significantly inhibited HSV-1 replication in vitro and further proved that BCA principally influenced the early stage of virus infection. We reveal that BCA downregulated the expression of pro-inflammatory factors triggered by HSV-1, including TNF-α, RANTES, IL-1β and IL-6. Furthermore, BCA treatment alleviated oxidative stress and apoptotic arising from HSV-1 infection. Lastly, we induced HSK in male C57BL/6 mice and treated them with either BCA or phosphate buffer solution (PBS) eye drops. We observed the ocular surface lesions; determined the virus load in the tear fluid, corneas as well as trigeminal ganglions (TGs); and detected the levels of inflammation and apoptosis in the corneas simultaneously. These results show that BCA inhibits HSV-1 and alleviates the corneal lesion degree. Our study illustrates that BCA is a promising therapeutic approach for application in treating HSK.
Collapse
Affiliation(s)
- Nan Zhou
- Department of Ophthalmology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (N.Z.)
| | - Deyuan Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Qiao You
- Center for Public Health Research, Medical School of Nanjing University, Nanjing 210093, China
| | - Taige Chen
- Department of Ophthalmology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (N.Z.)
| | - Jiaxuan Jiang
- Department of Ophthalmology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (N.Z.)
| | - Wenhao Shen
- Department of Ophthalmology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (N.Z.)
| | - Di Zhang
- Department of Ophthalmology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (N.Z.)
| | - Junpeng Liu
- Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Deyan Chen
- Center for Public Health Research, Medical School of Nanjing University, Nanjing 210093, China
| | - Kai Hu
- Department of Ophthalmology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; (N.Z.)
| |
Collapse
|
5
|
Roa CL, Cipolla-Neto J, Reiter RJ, Linhares IM, Lepique AP, de Aguiar LM, Seganfredo IB, Ferreira-Filho ES, de Medeiros SF, Baracat EC, Soares-Júnior JM. Effects of Melatonin Alone or Associated with Acyclovir on the Suppressive Treatment of Recurrent Genital Herpes: A Prospective, Randomized, and Double-Blind Study. Biomedicines 2023; 11:biomedicines11041088. [PMID: 37189706 DOI: 10.3390/biomedicines11041088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Suppressive therapy of recurrent genital herpes is a challenge, and melatonin may be an alternative. Objective: To evaluate the action of melatonin, acyclovir, or the association of melatonin with acyclovir as a suppressive treatment in women with recurrent genital herpes. Design: The study was prospective, double-blind, and randomized, including 56 patients as follows: (a) The melatonin group received 180 placebo capsules in the ‘day’ container and 180 melatonin 3 mg capsules in the ‘night’ container (n = 19); (b) The acyclovir group received 360 capsules of 400 mg acyclovir twice a day (one capsule during the day and another during the night) (n = 15); (c) the melatonin group received 180 placebo capsules in the ‘day’ container and 180 melatonin 3 mg capsules in the ‘night’ container (n = 22). The length of treatment was six months. The follow-up after treatment was six months. Patients were evaluated before, during, and after treatment through clinical visits, laboratory tests, and the application of four questionnaires (QSF-36, Beck, Epworth, VAS, and LANNS). Results: No statistically significant difference was observed for the depression and sleepiness questionnaires. However, in the Lanns scale for pain, all groups decreased the mean and median values in time (p = 0.001), without differentiation among the groups (p = 0.188). The recurrence rates of genital herpes within 60 days after treatment were 15.8%, 33.3%, and 36.4% in the melatonin, acyclovir, and association of melatonin with acyclovir groups, respectively. Conclusion: Our data suggest that melatonin may be an option for the suppressive treatment of recurrent genital herpes.
Collapse
Affiliation(s)
- Cristiane Lima Roa
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 05508-000, SP, Brazil
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX 78229, USA
| | - Iara Moreno Linhares
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Ana Paula Lepique
- Biomedical Building IV—Department of Immunology, Instituto de Ciências Biomédicas—USP, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Lana Maria de Aguiar
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Isadora Braga Seganfredo
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - Edson Santos Ferreira-Filho
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | | | - Edmund Chada Baracat
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| | - José Maria Soares-Júnior
- Discipline of Gynecology, Obstetrics and Gynecology Department, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-000, SP, Brazil
| |
Collapse
|
6
|
Alt M, Wolf S, van de Sand L, Dittrich R, Tertel T, Brochhagen L, Dirks M, Aufderhorst UW, Thümmler L, Otte M, Rainer K, Dittmer U, Giebel B, Trilling M, Silke Heilingloh C, Lotfi R, Roggendorf M, Witzke O, Krawczyk A. Cell-to-cell spread inhibiting antibodies constitute a correlate of protection against herpes simplex virus type 1 reactivations: A retrospective study. Front Immunol 2023; 14:1143870. [PMID: 37006290 PMCID: PMC10061111 DOI: 10.3389/fimmu.2023.1143870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
BackgroundHerpes simplex viruses (HSV) cause ubiquitous human infections. For vaccine development, knowledge concerning correlates of protection is essential. Therefore, we investigated (I) if humans are in principle capable producing cell-to-cell spread inhibiting antibodies against HSV and (II) whether this capacity is associated with a reduced HSV-1 reactivation risk.MethodsWe established a high-throughput HSV-1-ΔgE-GFP reporter virus-based assay and evaluated 2,496 human plasma samples for HSV-1 glycoprotein E (gE) independent cell-to-cell spread inhibiting antibodies. Subsequently, we conducted a retrospective survey among the blood donors to analyze the correlation between the presence of cell-to-cell spread inhibiting antibodies in plasma and the frequency of HSV reactivations.ResultsIn total, 128 of the 2,496 blood donors (5.1%) exhibited high levels of HSV-1 gE independent cell-to-cell spread inhibiting antibodies in the plasma. None of the 147 HSV-1 seronegative plasmas exhibited partial or complete cell-to-cell spread inhibition, demonstrating the specificity of our assay. Individuals with cell-to-cell spread inhibiting antibodies showed a significantly lower frequency of HSV reactivations compared to subjects without sufficient levels of such antibodies.ConclusionThis study contains two important findings: (I) upon natural HSV infection, some humans produce cell-to-cell spread inhibiting antibodies and (II) such antibodies correlate with protection against recurrent HSV-1. Moreover, these elite neutralizers may provide promising material for immunoglobulin therapy and information for the design of a protective vaccine against HSV-1.
Collapse
Affiliation(s)
- Mira Alt
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Susanne Wolf
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lukas van de Sand
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robin Dittrich
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leonie Brochhagen
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Miriam Dirks
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Wilhelm Aufderhorst
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Laura Thümmler
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mona Otte
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kordula Rainer
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Ulm, Germany
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Ulm, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Ramin Lotfi
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen and University Hospital Ulm, Ulm, Germany
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Michael Roggendorf
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Adalbert Krawczyk,
| |
Collapse
|
7
|
Kuraoka M, Aschner CB, Windsor IW, Mahant AM, Garforth SJ, Kong SL, Achkar JM, Almo SC, Kelsoe G, Herold BC. A non-neutralizing glycoprotein B monoclonal antibody protects against herpes simplex virus disease in mice. J Clin Invest 2023; 133:161968. [PMID: 36454639 PMCID: PMC9888390 DOI: 10.1172/jci161968] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
There is an unmet need for monoclonal antibodies (mAbs) for prevention or as adjunctive treatment of herpes simplex virus (HSV) disease. Most vaccine and mAb efforts focus on neutralizing antibodies, but for HSV this strategy has proven ineffective. Preclinical studies with a candidate HSV vaccine strain, ΔgD-2, demonstrated that non-neutralizing antibodies that activate Fcγ receptors (FcγRs) to mediate antibody-dependent cellular cytotoxicity (ADCC) provide active and passive protection against HSV-1 and HSV-2. We hypothesized that this vaccine provides a tool to identify and characterize protective mAbs. We isolated HSV-specific mAbs from germinal center and memory B cells and bone marrow plasmacytes of ΔgD-2-vaccinated mice and evaluated these mAbs for binding, neutralizing, and FcγR-activating activity and for protective efficacy in mice. The most potent protective mAb, BMPC-23, was not neutralizing but activated murine FcγRIV, a biomarker of ADCC. The cryo-electron microscopic structure of the Fab-glycoprotein B (gB) assembly identified domain IV of gB as the epitope. A single dose of BMPC-23 administered 24 hours before or after viral challenge provided significant protection when configured as mouse IgG2c and protected mice expressing human FcγRIII when engineered as a human IgG1. These results highlight the importance of FcR-activating antibodies in protecting against HSV.
Collapse
Affiliation(s)
- Masayuki Kuraoka
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Clare Burn Aschner
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Ian W. Windsor
- Department of Laboratory of Molecular Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Aakash Mahant Mahant
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Susan Luozheng Kong
- Department of Laboratory of Molecular Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jacqueline M. Achkar
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA.,Department of Medicine, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Garnett Kelsoe
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA.,Department of Surgery and,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Betsy C. Herold
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA.,Department of Pediatrics Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
8
|
Engineering antiviral immune-like systems for autonomous virus detection and inhibition in mice. Nat Commun 2022; 13:7629. [PMID: 36494373 PMCID: PMC9734111 DOI: 10.1038/s41467-022-35425-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
The ongoing COVID-19 pandemic has demonstrated that viral diseases represent an enormous public health and economic threat to mankind and that individuals with compromised immune systems are at greater risk of complications and death from viral diseases. The development of broad-spectrum antivirals is an important part of pandemic preparedness. Here, we have engineer a series of designer cells which we term autonomous, intelligent, virus-inducible immune-like (ALICE) cells as sense-and-destroy antiviral system. After developing a destabilized STING-based sensor to detect viruses from seven different genera, we have used a synthetic signal transduction system to link viral detection to the expression of multiple antiviral effector molecules, including antiviral cytokines, a CRISPR-Cas9 module for viral degradation and the secretion of a neutralizing antibody. We perform a proof-of-concept study using multiple iterations of our ALICE system in vitro, followed by in vivo functionality testing in mice. We show that dual output ALICESaCas9+Ab system delivered by an AAV-vector inhibited viral infection in herpetic simplex keratitis (HSK) mouse model. Our work demonstrates that viral detection and antiviral countermeasures can be paired for intelligent sense-and-destroy applications as a flexible and innovative method against virus infection.
Collapse
|
9
|
Backes IM, Byrd BK, Slein MD, Patel CD, Taylor SA, Garland CR, MacDonald SW, Balazs AB, Davis SC, Ackerman ME, Leib DA. Maternally transferred mAbs protect neonatal mice from HSV-induced mortality and morbidity. J Exp Med 2022; 219:e20220110. [PMID: 36156707 PMCID: PMC9516843 DOI: 10.1084/jem.20220110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/29/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2023] Open
Abstract
Neonatal herpes simplex virus (nHSV) infections often result in significant mortality and neurological morbidity despite antiviral drug therapy. Maternally transferred herpes simplex virus (HSV)-specific antibodies reduce the risk of clinically overt nHSV, but this observation has not been translationally applied. Using a neonatal mouse model, we tested the hypothesis that passive transfer of HSV-specific human mAbs can prevent mortality and morbidity associated with nHSV. The mAbs were expressed in vivo via vectored immunoprophylaxis or recombinantly. Through these maternally derived routes or through direct administration to pups, diverse mAbs to HSV glycoprotein D protected against neonatal HSV-1 and HSV-2 infection. Using in vivo bioluminescent imaging, both pre- and post-exposure mAb treatment significantly reduced viral load in mouse pups. Together these studies support the notion that HSV-specific mAb-based therapies could prevent or improve HSV infection outcomes in neonates.
Collapse
Affiliation(s)
- Iara M. Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Brook K. Byrd
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Matthew D. Slein
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Chaya D. Patel
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Sean A. Taylor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Callaghan R. Garland
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | | | | | - Scott C. Davis
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
- Thayer School of Engineering, Dartmouth College, Hanover, NH
| | - David A. Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| |
Collapse
|
10
|
Blank A, Hohmann N, Dettmer M, Manka‐Stuhlik A, Mikus G, Stoll F, Stützle‐Schnetz M, Thomas D, Exner E, Schmitt‐Bormann B, Schaller T, Laage R, Schönborn‐Kellenberger O, Arndt M, Haefeli WE, Krauss J. First-in-human, randomized, double-blind, placebo-controlled, dose escalation trial of the anti-herpes simplex virus monoclonal antibody HDIT101 in healthy volunteers. Clin Transl Sci 2022; 15:2366-2377. [PMID: 35869929 PMCID: PMC9579396 DOI: 10.1111/cts.13365] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/21/2022] [Accepted: 06/26/2022] [Indexed: 01/25/2023] Open
Abstract
HDIT101 is a first-in-class humanized monoclonal antibody recognizing a conserved epitope in glycoprotein B, a target present on the surface of herpes simplex virus 1 (HSV-1) and HSV-2 particles as well as on virus-infected cells. This was a first-in-human, single-center, double-blind, placebo-controlled trial in 24 healthy volunteers, randomized 3:1 (placebo:active) in each of the six dose levels with escalating doses up to 12,150 mg HDIT101. HDIT101 was administered intravenously, to study safety, pharmacokinetics (PKs), and immunogenicity. HDIT101 was well-tolerated in all recipients and no serious or severe adverse events, no infusion-related reactions, and no events suggestive of dose limiting off-target toxicity occurred. The mean serum exposure (area under the curve from zero to infinity [AUC0-∞ ]) of HDIT101 showed a linear increase from 4340 h*μg/ml at a dose of 50 mg to 1,122,247 h*μg/ml at a dose of 12,150 mg. No immunogenic effects following HDIT101 exposure were observed at any of the applied doses. HDIT101 demonstrated the expected PK properties of a monoclonal antibody was well-tolerated, and could be safely administered even at excessively high doses that may be required for treatment of patients with septical HSV spread.
Collapse
Affiliation(s)
- Antje Blank
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | - Nicolas Hohmann
- NCT, National Center for Tumor Diseases, Department of Medical OncologyHeidelberg University HospitalHeidelbergGermany
| | - Marlen Dettmer
- NCT, National Center for Tumor Diseases, Department of Medical OncologyHeidelberg University HospitalHeidelbergGermany
| | - Anette Manka‐Stuhlik
- NCT, National Center for Tumor Diseases, Department of Medical OncologyHeidelberg University HospitalHeidelbergGermany
| | - Gerd Mikus
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | - Felicitas Stoll
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | - Marlies Stützle‐Schnetz
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | | | - Evelyn Exner
- Heidelberg ImmunoTherapeutics GmbHHeidelbergGermany
| | | | | | - Rico Laage
- Heidelberg ImmunoTherapeutics GmbHHeidelbergGermany
| | | | | | - Walter E. Haefeli
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany
| | - Jürgen Krauss
- NCT, National Center for Tumor Diseases, Department of Medical OncologyHeidelberg University HospitalHeidelbergGermany
| |
Collapse
|
11
|
Backes IM, Leib DA, Ackerman ME. Monoclonal antibody therapy of herpes simplex virus: An opportunity to decrease congenital and perinatal infections. Front Immunol 2022; 13:959603. [PMID: 36016956 PMCID: PMC9398215 DOI: 10.3389/fimmu.2022.959603] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022] Open
Abstract
The fetal/neonatal period represents both a unique window of opportunity for interventions as well as vulnerability to a number of viral infections. While Herpesviruses such as herpes simplex virus (HSV) are highly prevalent and typically of little consequence among healthy adults, they are among the most consequential infections of early life. Despite treatment with antiviral drugs, neonatal HSV (nHSV) infections can still result in significant mortality and lifelong neurological morbidity. Fortunately, newborns in our pathogen-rich world inherit some of the protection provided by the maternal immune system in the form of transferred antibodies. Maternal seropositivity, resulting in placental transfer of antibodies capable of neutralizing virus and eliciting the diverse effector functions of the innate immune system are associated with dramatically decreased risk of nHSV. Given this clear epidemiological evidence of reduced risk of infection and its sequelae, we present what is known about the ability of monoclonal antibody therapies to treat or prevent HSV infection and explore how effective antibody-based interventions in conjunction with antiviral therapy might reduce early life mortality and long-term morbidity.
Collapse
Affiliation(s)
- Iara M Backes
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - David A Leib
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | | |
Collapse
|
12
|
Tian R, Ju F, Yu M, Liang Z, Xu Z, Zhao M, Qin Y, Lin Y, Huang X, Chang Y, Li S, Ren W, Lin C, Xia N, Huang C. A potent neutralizing and protective antibody against a conserved continuous epitope on HSV glycoprotein D. Antiviral Res 2022; 201:105298. [PMID: 35341808 DOI: 10.1016/j.antiviral.2022.105298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 11/25/2022]
Abstract
Infections caused by herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) remain a serious global health issue, and the medical countermeasures available thus far are limited. Virus-neutralizing monoclonal antibodies (NAbs) are crucial tools for studying host-virus interactions and designing effective vaccines, and the discovery and development of these NAbs could be one approach to treat or prevent HSV infection. Here, we report the isolation of five HSV NAbs from mice immunized with both HSV-1 and HSV-2. Among these were two antibodies that potently cross-neutralized both HSV-1 and HSV-2 with the 50% virus-inhibitory concentrations (IC50) below 200 ng/ml, one of which (4A3) exhibited high potency against HSV-2, with an IC50 of 59.88 ng/ml. 4A3 neutralized HSV at the prebinding stage and prevented HSV infection and cell-to-cell spread. Significantly, administration of 4A3 completely prevented weight loss and improved survival of mice challenged with a lethal dose of HSV-2. Using structure-guided molecular modeling combined with alanine-scanning mutagenesis, we observed that 4A3 bound to a highly conserved continuous epitope (residues 216 to 220) within the receptor-binding domain of glycoprotein D (gD) that is essential for viral infection and the triggering of membrane fusion. Our results provide guidance for developing NAb drugs and vaccines against HSV.
Collapse
Affiliation(s)
- Rui Tian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Fei Ju
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Mengqin Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhiqi Liang
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zilong Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Min Zhao
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yaning Qin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yanhua Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaoxuan Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yating Chang
- School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shaopeng Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenfeng Ren
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chaolong Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chenghao Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
13
|
Greenan E, Gallagher S, Khalil R, Murphy CC, Ní Gabhann-Dromgoole J. Advancing Our Understanding of Corneal Herpes Simplex Virus-1 Immune Evasion Mechanisms and Future Therapeutics. Viruses 2021; 13:v13091856. [PMID: 34578437 PMCID: PMC8473450 DOI: 10.3390/v13091856] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/24/2022] Open
Abstract
Herpes stromal keratitis (HSK) is a disease that commonly affects the cornea and external eye and is caused by Herpes Simplex Virus type 1 (HSV-1). This virus infects approximately 66% of people worldwide; however, only a small portion of these people will develop symptoms in their lifetime. There is no cure or vaccine available for HSV-1; however, there are treatments available that aim to control the inflammation caused by the virus and prevent its recurrence. While these treatments are beneficial to those suffering with HSK, there is a need for more effective treatments to minimise the need for topical steroids, which can have harmful effects, and to prevent bouts of disease reactivation, which can lead to progressive corneal scarring and visual impairment. This review details the current understanding of HSV-1 infection and discusses potential novel treatment options including microRNAs, TLRs, mAbs, and aptamers.
Collapse
Affiliation(s)
- Emily Greenan
- Department of Ophthalmology, Royal College of Surgeons in Ireland, D02 XK51 Dublin, Ireland; (E.G.); (C.C.M.)
- School of Pharmacy and Biomolecular Sciences (PBS), RSCI Research Institute, Royal College of Surgeons in Ireland, D02 XK51 Dublin, Ireland;
| | - Sophie Gallagher
- School of Biological and Health Sciences, Technological University (TU) Dublin, Kevin Street, D02 XK51 Dublin, Ireland;
| | - Rana Khalil
- School of Pharmacy and Biomolecular Sciences (PBS), RSCI Research Institute, Royal College of Surgeons in Ireland, D02 XK51 Dublin, Ireland;
| | - Conor C. Murphy
- Department of Ophthalmology, Royal College of Surgeons in Ireland, D02 XK51 Dublin, Ireland; (E.G.); (C.C.M.)
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, D02 XK51 Dublin, Ireland
| | - Joan Ní Gabhann-Dromgoole
- Department of Ophthalmology, Royal College of Surgeons in Ireland, D02 XK51 Dublin, Ireland; (E.G.); (C.C.M.)
- School of Pharmacy and Biomolecular Sciences (PBS), RSCI Research Institute, Royal College of Surgeons in Ireland, D02 XK51 Dublin, Ireland;
- Correspondence:
| |
Collapse
|
14
|
Peptide Derivatives of Platelet-Derived Growth Factor Receptor Alpha Inhibit Cell-Associated Spread of Human Cytomegalovirus. Viruses 2021; 13:v13091780. [PMID: 34578361 PMCID: PMC8473290 DOI: 10.3390/v13091780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 12/27/2022] Open
Abstract
Cell-free human cytomegalovirus (HCMV) can be inhibited by a soluble form of the cellular HCMV-receptor PDGFRα, resembling neutralization by antibodies. The cell-associated growth of recent HCMV isolates, however, is resistant against antibodies. We investigated whether PDGFRα-derivatives can inhibit this transmission mode. A protein containing the extracellular PDGFRα-domain and 40-mer peptides derived therefrom were tested regarding the inhibition of the cell-associated HCMV strain Merlin-pAL1502, hits were validated with recent isolates, and the most effective peptide was modified to increase its potency. The modified peptide was further analyzed regarding its mode of action on the virion level. While full-length PDGFRα failed to inhibit HCMV isolates, three peptides significantly reduced virus growth. A 30-mer version of the lead peptide (GD30) proved even more effective against the cell-free virus, and this effect was HCMV-specific and depended on the viral glycoprotein O. In cell-associated spread, GD30 reduced both the number of transferred particles and their penetration. This effect was reversible after peptide removal, which allowed the synchronized analysis of particle transfer, showing that two virions per hour were transferred to neighboring cells and one virion was sufficient for infection. In conclusion, PDGFRα-derived peptides are novel inhibitors of the cell-associated spread of HCMV and facilitate the investigation of this transmission mode.
Collapse
|
15
|
Brenner S, Braun B, Read C, Weil T, Walther P, Schrader T, Münch J, von Einem J. The Molecular Tweezer CLR01 Inhibits Antibody-Resistant Cell-to-Cell Spread of Human Cytomegalovirus. Viruses 2021; 13:v13091685. [PMID: 34578265 PMCID: PMC8472163 DOI: 10.3390/v13091685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) uses two major ways for virus dissemination: infection by cell-free virus and direct cell-to-cell spread. Neutralizing antibodies can efficiently inhibit infection by cell-free virus but mostly fail to prevent cell-to-cell transmission. Here, we show that the ‘molecular tweezer’ CLR01, a broad-spectrum antiviral agent, is not only highly active against infection with cell-free virus but most remarkably inhibits antibody-resistant direct cell-to-cell spread of HCMV. The inhibition of cell-to-cell spread by CLR01 was not limited to HCMV but was also shown for the alphaherpesviruses herpes simplex viruses 1 and 2 (HSV-1, -2). CLR01 is a rapid acting small molecule that inhibits HCMV entry at the attachment and penetration steps. Electron microscopy of extracellular virus particles indicated damage of the viral envelope by CLR01, which likely impairs the infectivity of virus particles. The rapid inactivation of viral particles by CLR01, the viral envelope as the main target, and the inhibition of virus entry at different stages are presumably the key to inhibition of cell-free virus infection and cell-to-cell spread by CLR01. Importance: While cell-free spread enables the human cytomegalovirus (HCMV) and other herpesviruses to transmit between hosts, direct cell-to-cell spread is thought to be more relevant for in vivo dissemination within infected tissues. Cell-to-cell spread is resistant to neutralizing antibodies, thus contributing to the maintenance of virus infection and virus dissemination in the presence of an intact immune system. Therefore, it would be therapeutically interesting to target this mode of spread in order to treat severe HCMV infections and to prevent dissemination of virus within the infected host. The molecular tweezer CLR01 exhibits broad-spectrum antiviral activity against a number of enveloped viruses and efficiently blocks antibody-resistant cell-to-cell spread of HCMV, thus representing a novel class of small molecules with promising antiviral activity.
Collapse
Affiliation(s)
- Sina Brenner
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
| | - Berenike Braun
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
| | - Clarissa Read
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany;
| | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (T.W.); (J.M.)
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany;
| | - Thomas Schrader
- Faculty of Chemistry, University of Duisburg-Essen, 45117 Essen, Germany;
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (T.W.); (J.M.)
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jens von Einem
- Institute of Virology, Ulm University Medical Center, 89081 Ulm, Germany; (S.B.); (B.B.); (C.R.)
- Correspondence: ; Tel.: +49-(0)731-500-65104; Fax: +49-(0)731-500-65102
| |
Collapse
|
16
|
Obisesan O, Katata-Seru L, Mufamadi S, Mufhandu H. Applications of Nanoparticles for Herpes Simplex Virus (HSV) and Human Immunodeficiency Virus (HIV) Treatment. J Biomed Nanotechnol 2021; 17:793-808. [PMID: 34082867 DOI: 10.1166/jbn.2021.3074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent years, the growing studies focused on the immunotherapy of hepatocellular carcinoma and proved the preclinical and clinical promises of host antitumor immune response. However, there were still various obstacles in meeting satisfactory clinic need, such as low response rate, primary resistance and secondary resistance to immunotherapy. Tackling these barriers required a deeper understanding of immune underpinnings and a broader understanding of advanced technology. This review described immune microenvironment of liver and HCC which naturally decided the complexity of immunotherapy, and summarized recent immunotherapy focusing on different points. The ever-growing clues indicated that the instant killing of tumor cell and the subsequent relive of immunosuppressive microenvironment were both indis- pensables. The nanotechnology applied in immunotherapy and the combination with intervention technology was also discussed.
Collapse
Affiliation(s)
- Oluwafemi Obisesan
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Lebogang Katata-Seru
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Steven Mufamadi
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| | - Hazel Mufhandu
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110000, China
| |
Collapse
|
17
|
Koganti R, Yadavalli T, Naqvi RA, Shukla D, Naqvi AR. Pathobiology and treatment of viral keratitis. Exp Eye Res 2021; 205:108483. [PMID: 33556334 DOI: 10.1016/j.exer.2021.108483] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/12/2021] [Accepted: 01/29/2021] [Indexed: 12/17/2022]
Abstract
Keratitis is one of the most prevalent ocular diseases manifested by partial or total loss of vision. Amongst infectious (viz., microbes including bacteria, fungi, amebae, and viruses) and non-infectious (viz., eye trauma, chemical exposure, and ultraviolet exposure, contact lens) risk factors, viral keratitis has been demonstrated as one of the leading causes of corneal opacity. While many viruses have been shown to cause keratitis (such as rhabdoviruses, coxsackieviruses, etc.), herpesviruses are the predominant etiologic agent of viral keratitis. This chapter will summarize current knowledge on the prevalence, diagnosis, and pathobiology of viral keratitis. Virus-mediated immunomodulation of host innate and adaptive immune components is critical for viral persistence, and dysfunctional immune responses may cause destruction of ocular tissues leading to keratitis. Immunosuppressed or immunocompromised individuals may display recurring disease with pronounced severity. Early diagnosis of viral keratitis is beneficial for disease management and response to treatment. Finally, we have discussed current and emerging therapies to treat viral keratitis.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA
| | - Raza Ali Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA; Department of Microbiology and Immunology, University of Illinois at Chicago, IL, 60612, USA.
| | - Afsar R Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
18
|
Padmanabhanunni A, Pretorius T. The unbearable loneliness of COVID-19: COVID-19-related correlates of loneliness in South Africa in young adults. Psychiatry Res 2021; 296:113658. [PMID: 33360428 PMCID: PMC9754815 DOI: 10.1016/j.psychres.2020.113658] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/14/2020] [Indexed: 01/10/2023]
Abstract
This is the first study to examine the association between COVID-19 related variables and loneliness among young adults in South Africa during COVID-19. Participants (N=337) were university students who completed the UCLA Loneliness Scale and five selected subscales of the WHO COVID-19 Behavioural Insights Tool. The mean loneliness scores were significantly higher than previous studies in other contexts as well as studies conducted in the time of COVID-19. Correlational analysis found that greater perceived risk of infection, limited perceived knowledge of COVID-19 and lower appraisals of resilience were associated with increased loneliness. In a regression analysis, when all COVID-19 variables were considered simultaneously, only resilience, self-rated knowledge, and risk perception emerged as significant correlates of loneliness. These findings suggest that loneliness is a significant public health concern in South Africa in the time of COVID-19. It also suggests that self-efficacy and resilience can potentially be reinforced by public health campaigns that focus on enhancing COVID-19-related knowledge and preparedness.
Collapse
Affiliation(s)
- A. Padmanabhanunni
- Department of Psychology, University of the Western Cape, Western Cape Province, South Africa,Corresponding author at:Private Bag X17, Department of Psychology, University of the Western Cape, Bellville, Cape Town, Western Cape, South Africa
| | - T.B. Pretorius
- Department of Psychology, University of the Western Cape, Western Cape Province, South Africa
| |
Collapse
|
19
|
Ruzsics Z, Hoffmann K, Riedl A, Krawczyk A, Widera M, Sertznig H, Schipper L, Kapper-Falcone V, Debreczeny M, Ernst W, Grabherr R, Hengel H, Harant H. A Novel, Broad-Acting Peptide Inhibitor of Double-Stranded DNA Virus Gene Expression and Replication. Front Microbiol 2020; 11:601555. [PMID: 33281801 PMCID: PMC7705112 DOI: 10.3389/fmicb.2020.601555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/27/2020] [Indexed: 12/30/2022] Open
Abstract
Viral infections are a global disease burden with only a limited number of antiviral agents available. Due to newly emerging viral pathogens and increasing occurrence of drug resistance, there is a continuous need for additional therapeutic options, preferably with extended target range. In the present study, we describe a novel antiviral peptide with broad activity against several double-stranded DNA viruses. The 22-mer peptide TAT-I24 potently neutralized viruses such as herpes simplex viruses, adenovirus type 5, cytomegalovirus, vaccinia virus, and simian virus 40 in cell culture models, while being less active against RNA viruses. The peptide TAT-I24 therefore represents a novel and promising drug candidate for use against double-stranded DNA viruses.
Collapse
Affiliation(s)
- Zsolt Ruzsics
- Institute of Virology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Consulting Laboratory for HSV and VZV, Medical Center-University of Freiburg, Freiburg, Germany
| | - Katja Hoffmann
- Institute of Virology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - André Riedl
- Institute of Virology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adalbert Krawczyk
- Department of Infectious Diseases, West German Centre of Infectious Diseases, Universitätsmedizin Essen, University Duisburg-Essen, Essen, Germany.,Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Marek Widera
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Institute of Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Helene Sertznig
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leonie Schipper
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Valeria Kapper-Falcone
- Institute of Virology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Consulting Laboratory for HSV and VZV, Medical Center-University of Freiburg, Freiburg, Germany
| | - Monika Debreczeny
- VIBT Imaging Center, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Wolfgang Ernst
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Reingard Grabherr
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Hartmut Hengel
- Institute of Virology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Consulting Laboratory for HSV and VZV, Medical Center-University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
20
|
Cell Fusion Induced by a Fusion-Active Form of Human Cytomegalovirus Glycoprotein B (gB) Is Inhibited by Antibodies Directed at Antigenic Domain 5 in the Ectodomain of gB. J Virol 2020; 94:JVI.01276-20. [PMID: 32641474 DOI: 10.1128/jvi.01276-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen that can cause severe clinical disease in allograft recipients and infants infected in utero Virus-neutralizing antibodies defined in vitro have been proposed to confer protection against HCMV infection, and the virion envelope glycoprotein B (gB) serves as a major target of neutralizing antibodies. The viral fusion protein gB is nonfusogenic on its own and requires glycoproteins H (gH) and L (gL) for membrane fusion, which is in contrast to requirements of related class III fusion proteins, including vesicular stomatitis virus glycoprotein G (VSV-G) or baculovirus gp64. To explore requirements for gB's fusion activity, we generated a set of chimeras composed of gB and VSV-G or gp64, respectively. These gB chimeras were intrinsically fusion active and led to the formation of multinucleated cell syncytia when expressed in the absence of other viral proteins. Utilizing a panel of virus-neutralizing gB-specific monoclonal antibodies (MAbs), we could demonstrate that syncytium formation of the fusogenic gB/VSV-G chimera can be significantly inhibited by only a subset of neutralizing MAbs which target antigenic domain 5 (AD-5) of gB. This observation argues for differential modes of action of neutralizing anti-gB MAbs and suggests that blocking the membrane fusion function of gB could be one mechanism of antibody-mediated virus neutralization. In addition, our data have important implications for the further understanding of the conformation of gB that promotes membrane fusion as well as the identification of structures in AD-5 that could be targeted by antibodies to block this early step in HCMV infection.IMPORTANCE HCMV is a major global health concern, and antiviral chemotherapy remains problematic due to toxicity of available compounds and the emergence of drug-resistant viruses. Thus, an HCMV vaccine represents a priority for both governmental and pharmaceutical research programs. A major obstacle for the development of a vaccine is a lack of knowledge of the nature and specificities of protective immune responses that should be induced by such a vaccine. Glycoprotein B of HCMV is an important target for neutralizing antibodies and, hence, is often included as a component of intervention strategies. By generation of fusion-active gB chimeras, we were able to identify target structures of neutralizing antibodies that potently block gB-induced membrane fusion. This experimental system provides an approach to screen for antibodies that interfere with gB's fusogenic activity. In summary, our data will likely contribute to both rational vaccine design and the development of antibody-based therapies against HCMV.
Collapse
|
21
|
Herpes Simplex Virus Type 2 Is More Difficult to Neutralize by Antibodies Than Herpes Simplex Virus Type 1. Vaccines (Basel) 2020; 8:vaccines8030478. [PMID: 32867086 PMCID: PMC7563860 DOI: 10.3390/vaccines8030478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/28/2022] Open
Abstract
Infections with herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) are a global health burden. Besides painful oral or genital lesions in otherwise healthy subjects, both viruses can cause devastating morbidity and mortality in immune-compromised and immune-immature individuals. The latter are particularly susceptible to a disseminated, life-threatening disease. Neutralizing antibodies (NAb) constitute a correlate of protection from disease, and are promising candidates for the prophylactic or therapeutic treatment of severe HSV infections. However, a clinical vaccine trial suggested that HSV-2 might be more resistant to NAbs than HSV-1. In the present study, we investigated the antiviral efficacy of the well-characterized humanized monoclonal antibody (mAb) hu2c against HSV-2, in a NOD/SCID immunodeficiency mouse model. Despite the fact that hu2c recognizes a fully conserved epitope and binds HSV-1 and HSV-2 glycoprotein B with equal affinity, it was much less effective against HSV-2 in vitro and in NOD/SCID mice. Although intravenous antibody treatment prolonged the survival of HSV-2-infected mice, complete protection from death was not achieved. Our data demonstrate that HSV-2 is more resistant to NAbs than HSV-1, even if the same antibody and antigen are concerned, making the development of a vaccine or therapeutic antibodies more challenging.
Collapse
|
22
|
Birzer A, Krawczyk A, Draßner C, Kuhnt C, Mühl-Zürbes P, Heilingloh CS, Steinkasserer A, Popella L. HSV-1 Modulates IL-6 Receptor Expression on Human Dendritic Cells. Front Immunol 2020; 11:1970. [PMID: 32983130 PMCID: PMC7479228 DOI: 10.3389/fimmu.2020.01970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are the guardians of the immune system since they are located in the majority of peripheral tissues. In addition, they are crucial for the induction of an effective immune response based on their unique capacity to stimulate naive T cells. During co-evolution, the human pathogen herpes simplex virus type 1 (HSV-1) has evolved several immune evasion mechanisms in order to subvert the host's immune system especially by targeting DC biology and function. Here we demonstrate that HSV-1 infection influences the IL-6 receptor (IL6R) expression both on protein and mRNA levels in/on human monocyte-derived mature DCs (mDCs). Surprisingly, reduced IL6R expression levels were also observed on uninfected bystander mDCs. Mechanistically, we clearly show that HSV-1-derived non-infectious light (L-) particles are sufficient to trigger IL6R regulation on uninfected bystander mDCs. These L-particles lack the viral DNA-loaded capsid and are predominantly produced during infection of mDCs. Our results show that the deletion of the HSV-1 tegument protein vhs partially rescued the reduced IL6R surface expression levels on/in bystander mDCs. Using a neutralizing antibody, which perturbs the transfer of L-particles to bystander mDCs, was sufficient to rescue the modulation of IL6R surface expression on uninfected bystander mDCs. This study provides evidence that L-particles transfer specific viral proteins to uninfected bystander mDCs, thereby negatively interfering with their IL6R expression levels, however, to a lesser extend compared to H-particles. Due to their immune-modulatory capacity, L-particles represent an elaborated approach of HSV-1-mediated immune evasion.
Collapse
Affiliation(s)
- Alexandra Birzer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christina Draßner
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christiane Silke Heilingloh
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Linda Popella
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
23
|
Lang J, Bohn P, Bhat H, Jastrow H, Walkenfort B, Cansiz F, Fink J, Bauer M, Olszewski D, Ramos-Nascimento A, Duhan V, Friedrich SK, Becker KA, Krawczyk A, Edwards MJ, Burchert A, Huber M, Friebus-Kardash J, Göthert JR, Hardt C, Probst HC, Schumacher F, Köhrer K, Kleuser B, Babiychuk EB, Sodeik B, Seibel J, Greber UF, Lang PA, Gulbins E, Lang KS. Acid ceramidase of macrophages traps herpes simplex virus in multivesicular bodies and protects from severe disease. Nat Commun 2020; 11:1338. [PMID: 32165633 PMCID: PMC7067866 DOI: 10.1038/s41467-020-15072-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/17/2020] [Indexed: 12/20/2022] Open
Abstract
Macrophages have important protective functions during infection with herpes simplex virus type 1 (HSV-1). However, molecular mechanisms that restrict viral propagation and protect from severe disease are unclear. Here we show that macrophages take up HSV-1 via endocytosis and transport the virions into multivesicular bodies (MVBs). In MVBs, acid ceramidase (aCDase) converts ceramide into sphingosine and increases the formation of sphingosine-rich intraluminal vesicles (ILVs). Once HSV-1 particles reach MVBs, sphingosine-rich ILVs bind to HSV-1 particles, which restricts fusion with the limiting endosomal membrane and prevents cellular infection. Lack of aCDase in macrophage cultures or in Asah1-/- mice results in replication of HSV-1 and Asah1-/- mice die soon after systemic or intravaginal inoculation. The treatment of macrophages with sphingosine enhancing compounds blocks HSV-1 propagation, suggesting a therapeutic potential of this pathway. In conclusion, aCDase loads ILVs with sphingosine, which prevents HSV-1 capsids from penetrating into the cytosol.
Collapse
Affiliation(s)
- Judith Lang
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Patrick Bohn
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Hilal Bhat
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Holger Jastrow
- Institute of Anatomy, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Institut for Experimental Immunology and Imaging, Imaging Center Essen, Electron Microscopy Unit, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Bernd Walkenfort
- Institut for Experimental Immunology and Imaging, Imaging Center Essen, Electron Microscopy Unit, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Feyza Cansiz
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Julian Fink
- Institute of Organic Chemistry, Julius-Maximilians University of Würzburg, Am Hubland, Würzburg, D-97074, Germany
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstr. 190, CH-8057, Zurich, Switzerland
| | - Dominik Olszewski
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstr. 190, CH-8057, Zurich, Switzerland
| | - Ana Ramos-Nascimento
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, D-30625, Germany
| | - Vikas Duhan
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Sarah-Kim Friedrich
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Katrin Anne Becker
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Department of Infectious Diseases, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Michael J Edwards
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, University Hospital Giessen and Marburg, Campus Marburg, Baldingerstr., Marburg, D-35043, Germany
| | - Magdalena Huber
- Institute of Medical Microbiology and Hospital Hygiene, Philipps-University Marburg, Hans-Meerwein Str. 2, Marburg, D-35043, Germany
| | - Justa Friebus-Kardash
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Joachim R Göthert
- Department of Hematology, West German Cancer Center, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Cornelia Hardt
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany
| | - Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Langenbeckstr. 1, Mainz, D-55131, Germany
| | - Fabian Schumacher
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert Allee 114-116, Nuthetal, D-14558, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Heinrich-Heine-University, Universitätsstr. 1, Düsseldorf, D-40225, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert Allee 114-116, Nuthetal, D-14558, Germany
| | - Eduard B Babiychuk
- Institute of Anatomy, University of Bern, Baltzerstr. 4, CH-3012, Bern, Switzerland
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, D-30625, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, D-30625, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, Julius-Maximilians University of Würzburg, Am Hubland, Würzburg, D-97074, Germany
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstr. 190, CH-8057, Zurich, Switzerland
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, Düsseldorf, D-40225, Germany
| | - Erich Gulbins
- Institute of Molecular Biology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.,Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Karl S Lang
- Institute of Immunology, University of Duisburg-Essen, Hufelandstr. 55, Essen, D-45147, Germany.
| |
Collapse
|
24
|
Naidu SK, Nabi R, Cheemarla NR, Stanfield BA, Rider PJ, Jambunathan N, Chouljenko VN, Carter R, Del Piero F, Langohr I, Kousoulas KG. Intramuscular vaccination of mice with the human herpes simplex virus type-1(HSV-1) VC2 vaccine, but not its parental strain HSV-1(F) confers full protection against lethal ocular HSV-1 (McKrae) pathogenesis. PLoS One 2020; 15:e0228252. [PMID: 32027675 PMCID: PMC7004361 DOI: 10.1371/journal.pone.0228252] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/11/2020] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) can cause severe ocular infection and blindness. We have previously shown that the HSV-1 VC2 vaccine strain is protective in mice and guinea pigs against genital herpes infection following vaginal challenge with HSV-1 or HSV-2. In this study, we evaluated the efficacy of VC2 intramuscular vaccination in mice against herpetic keratitis following ocular challenge with lethal human clinical strain HSV-1(McKrae). VC2 vaccination in mice produced superior protection and morbidity control in comparison to its parental strain HSV-1(F). Specifically, after HSV-1(McKrae) ocular challenge, all VC2 vaccinated- mice survived, while 30% of the HSV-1(F)- vaccinated and 100% of the mock-vaccinated mice died post challenge. VC2-vaccinated mice did not exhibit any symptoms of ocular infection and completely recovered from initial conjunctivitis. In contrast, HSV-1(F)-vaccinated mice developed time-dependent progressive keratitis characterized by corneal opacification, while mock-vaccinated animals exhibited more severe stromal keratitis characterized by immune cell infiltration and neovascularization in corneal stroma with corneal opacification. Cornea in VC2-immunized mice exhibited significantly increased infiltration of CD3+ T lymphocytes and decreased infiltration of Iba1+ macrophages in comparison to mock- or HSV-1(F)-vaccinated groups. VC2 immunization produced higher virus neutralization titers than HSV-1(F) post challenge. Furthermore, VC-vaccination significantly increased the CD4 T central memory (TCM) subsets and CD8 T effector memory (TEM) subsets in the draining lymph nodes following ocular HSV-1 (McKrae) challenge, then mock- or HSV-1(F)-vaccination. These results indicate that VC2 vaccination produces a protective immune response at the site of challenge to protect against HSV-1-induced ocular pathogenesis.
Collapse
Affiliation(s)
- Shan K. Naidu
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Rafiq Nabi
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Nagarjuna R. Cheemarla
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Brent A. Stanfield
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Paul J. Rider
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Nithya Jambunathan
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Vladimir N. Chouljenko
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Renee Carter
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Fabio Del Piero
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Ingeborg Langohr
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Konstantin G. Kousoulas
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
25
|
Koganti R, Yadavalli T, Shukla D. Current and Emerging Therapies for Ocular Herpes Simplex Virus Type-1 Infections. Microorganisms 2019; 7:microorganisms7100429. [PMID: 31658632 PMCID: PMC6843252 DOI: 10.3390/microorganisms7100429] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) is a neurotropic, double-stranded DNA virus that can cause a wide variety of diseases, including many ocular pathologies. It is one of the leading causes of infectious blindness in the United States. Because of its ubiquitous nature and its potential to cause serious ocular maladies, there is a significant need for more effective antiviral therapies against ocular HSV-1. In this review, we discuss the lifecycle of HSV-1 as it pertains to corneal infections and the clinically approved as well as emerging treatments to combat HSV-1 infections. We also highlight some newly identified host targets for the antiviral drug development.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL 60612, USA.
| | - Tejabhiram Yadavalli
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL 60612, USA.
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL 60612, USA.
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
26
|
Cell-to-Cell Spread Blocking Activity Is Extremely Limited in the Sera of Herpes Simplex Virus 1 (HSV-1)- and HSV-2-Infected Subjects. J Virol 2019; 93:JVI.00070-19. [PMID: 30867302 DOI: 10.1128/jvi.00070-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) and HSV-2 can evade serum antibody-mediated neutralization through cell-to-cell transmission mechanisms, which represent one of the central steps in disease reactivation. To address the role of humoral immunity in controlling HSV-1 and HSV-2 replication, we analyzed serum samples from 44 HSV-1 and HSV-2 seropositive subjects by evaluating (i) their efficiency in binding both the purified viral particles and recombinant gD and gB viral glycoproteins, (ii) their neutralizing activity, and (iii) their capacity to inhibit the cell-to-cell virus passage in vitro All of the sera were capable of binding gD, gB, and whole virions, and all sera significantly neutralized cell-free virus. However, neither whole sera nor purified serum IgG fraction was able to inhibit significantly cell-to-cell virus spreading in in vitro post-virus-entry infectious assays. Conversely, when spiked with an already described anti-gD human monoclonal neutralizing antibody capable of inhibiting HSV-1 and -2 cell-to-cell transmission, each serum boosted both its neutralizing and post-virus-entry inhibitory activity, with no interference exerted by serum antibody subpopulations.IMPORTANCE Despite its importance in the physiopathology of HSV-1 and -2 infections, the cell-to-cell spreading mechanism is still poorly understood. The data shown here suggest that infection-elicited neutralizing antibodies capable of inhibiting cell-to-cell virus spread can be underrepresented in most infected subjects. These observations can be of great help in better understanding the role of humoral immunity in controlling virus reactivation and in the perspective of developing novel therapeutic strategies, studying novel correlates of protection, and designing effective vaccines.
Collapse
|
27
|
Lee DH, Zuckerman RA. Herpes simplex virus infections in solid organ transplantation: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019; 33:e13526. [PMID: 30859647 DOI: 10.1111/ctr.13526] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/27/2019] [Indexed: 12/19/2022]
Abstract
These updated guidelines from the Infectious Diseases Community of Practice of the American Society of Transplantation review the diagnosis, prevention, and management of HSV in the pre- and post-transplant period. A majority of transplant recipients are seropositive for HSV-1 or 2. Compared with immunocompetent persons, SOT recipients shed HSV more frequently, have more severe clinical manifestations, and are slower to respond to therapy. Most HSV infection is diagnosed on clinical grounds, but patients may present with atypical lesions and/or other clinical manifestations. Acquisition from the donor is rare. Polymerase chain reaction is the preferred diagnostic test unless culture is needed for resistance testing. For limited mucocutaneous lesions, oral therapy can be used; however, in severe, disseminated, visceral or CNS involvement, acyclovir doses of up to 10 mg/kg every 8 hours intravenously should be initiated. Acyclovir-resistant HSV is less common in SOT patients than in HSCT and can be treated with foscarnet, though other novel therapies are currently under investigation. HSV-specific prophylaxis should be considered for all HSV-1 and HSV-2-seropositive organ recipients who are not receiving antiviral medication for CMV prevention that has activity against HSV.
Collapse
Affiliation(s)
- Dong H Lee
- Division of Infectious Diseases and HIV Medicine, College of Medicine, Drexel University, Philadelphia, Pennsylvania
| | - Richard A Zuckerman
- Infectious Disease Service for Transplant and Immunocompromised Hosts, Section of Infectious Disease and International Health, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | |
Collapse
|
28
|
Mohseni AH, Taghinezhad-S S, Keyvani H, Ghobadi N. Comparison of Acyclovir and Multistrain Lactobacillus brevis in Women with Recurrent Genital Herpes Infections: a Double-Blind, Randomized, Controlled Study. Probiotics Antimicrob Proteins 2019; 10:740-747. [PMID: 28852985 DOI: 10.1007/s12602-017-9320-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We performed a randomized double-blind controlled trial to compare the efficacy and safety of multistrain probiotic and acyclovir in women patients with recurrent genital herpes simplex virus type 2 (HSV-2) infections. Eighty-one patients enrolled in the study were being treated with multistrain Lactobacillus brevis one vaginal capsule every 12 h and oral acyclovir 400 mg twice daily for 6 months. Of 53 patients who completed both treatment courses, no important differences were identified between acyclovir and probiotic for the primary and secondary efficacy endpoint, resolution of episode (hazard ratio, 0.60; 95% CI, 0.3429 to 1.0663; P = 0.08), lesion healing time (hazard ratio, 0.57; 95% CI, 0.3034 to 1.0717, P = 0.08), viral shedding (hazard ratio, 0.54; 95% CI, 0.3027 to 0.9750, P = 0.04), and percentage of pain (hazard ratio, 0.48; 95% CI, 0.2708 to 0.8545, P = 0.01). The median time to first and second recurrence after treatment were 43 and 121 days in patients receiving acyclovir and 33 and 118 days in patients receiving probiotic (HR 2.61; 95% CI, 1.4427 to 4.7546, P = 0.001, and HR 0.62; 95% CI, 0.3500 to 1.1133, P = 0.1, respectively). No clinically important effects happened during the probiotic treatment but some of adverse events reported in patients taking acyclovir. Easy availability, low cost, and no side effect of L. brevis are valuable properties of probiotic therapy compared with acyclovir. Therefore, we concluded that multistrain L. brevis could play an important role in suppression of recurrent genital herpes simplex virus infection.
Collapse
Affiliation(s)
- Amir Hossein Mohseni
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Research and Development (R&D) Department, Keyvan Virology Specialty Laboratory (KVSL), Tehran, Iran
| | - Sedigheh Taghinezhad-S
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran. .,Research and Development (R&D) Department, Keyvan Virology Specialty Laboratory (KVSL), Tehran, Iran.
| | - Hossein Keyvani
- Research and Development (R&D) Department, Keyvan Virology Specialty Laboratory (KVSL), Tehran, Iran. .,Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Narges Ghobadi
- Research and Development (R&D) Department, Keyvan Virology Specialty Laboratory (KVSL), Tehran, Iran
| |
Collapse
|
29
|
Koujah L, Suryawanshi RK, Shukla D. Pathological processes activated by herpes simplex virus-1 (HSV-1) infection in the cornea. Cell Mol Life Sci 2019; 76:405-419. [PMID: 30327839 PMCID: PMC6349487 DOI: 10.1007/s00018-018-2938-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/03/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Abstract
Herpes simplex virus type-1 (HSV-1) is a ubiquitous pathogen that infects a large majority of the human population worldwide. It is also a leading cause of infection-related blindness in the developed world. HSV-1 infection of the cornea begins with viral entry into resident cells via a multistep process that involves interaction of viral glycoproteins and host cell surface receptors. Once inside, HSV-1 infection induces a chronic immune-inflammatory response resulting in corneal scarring, thinning and neovascularization. This leads to development of various ocular diseases such as herpes stromal keratitis, resulting in visual impairment and eventual blindness. HSV-1 can also invade the central nervous system and lead to encephalitis, a relatively common cause of sporadic fetal encephalitis worldwide. In this review, we discuss the pathological processes activated by corneal HSV-1 infection and existing antiviral therapies as well as novel therapeutic options currently under development.
Collapse
Affiliation(s)
- Lulia Koujah
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rahul K Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA.
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
30
|
Induction of herpes simplex virus type 1 cell-to-cell spread inhibiting antibodies by a calcium phosphate nanoparticle-based vaccine. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 16:138-148. [PMID: 30594660 DOI: 10.1016/j.nano.2018.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/23/2018] [Accepted: 12/10/2018] [Indexed: 11/20/2022]
Abstract
Herpes simplex viruses 1 and 2 are among the most ubiquitous human infections and persist lifelong in their host. Upon primary infection or reactivation from ganglia, the viruses spread by direct cell-cell contacts (cell-to-cell spread) and thus escape from the host immune response. We have developed a monoclonal antibody (mAb 2c), which inhibits the HSV cell-to-cell spread, thereby protecting from lethal genital infection and blindness in animal models. In the present study we have designed a nanoparticle-based vaccine to induce protective antibody responses exceeding the cell-to-cell spread inhibiting properties of mAb 2c. We used biodegradable calcium phosphate (CaP) nanoparticles coated with a synthetic peptide that represents the conformational epitope on HSV-1 gB recognized by mAb 2c. The CaP nanoparticles additionally contained a TLR-ligand CpGm and were formulated with adjuvants to facilitate the humoral immune response. This vaccine effectively protected mice from lethal HSV-1 infection by inducing cell-to-cell spread inhibiting antibodies.
Collapse
|
31
|
Tajpara P, Mildner M, Schmidt R, Vierhapper M, Matiasek J, Popow-Kraupp T, Schuster C, Elbe-Bürger A. A Preclinical Model for Studying Herpes Simplex Virus Infection. J Invest Dermatol 2018; 139:673-682. [PMID: 30414908 PMCID: PMC7100788 DOI: 10.1016/j.jid.2018.08.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/03/2018] [Accepted: 08/12/2018] [Indexed: 01/29/2023]
Abstract
Herpes simplex virus (HSV) infections can cause considerable morbidity. Currently, nucleoside analogues such as acyclovir are widely used for treatment. However, HSV infections resistant to these drugs are a clinical problem among immunocompromised patients. To provide more efficient therapy and to counteract resistance, a different class of antiviral compounds has been developed. Pritelivir, a helicase primase inhibitor, represents a promising candidate for improved therapy. Here, we established an HSV-1 infection model on microneedle-pretreated human skin ex vivo. We identified HSV-1–specific histological changes (e.g., cytopathic effects, multinucleated giant cells), down-regulation of nectin-1, nuclear translocation of NF-κB (p65), interferon regulatory factor 3 (IRF3), and signaling of the IFN-inducible protein MxA. Accordingly, this model was used to test the potency of pritelivir compared with the standard drug acyclovir. We discovered that both drugs had a comparable efficacy for inhibiting HSV-1 replication, suggesting that pritelivir could be an alternative therapeutic agent for patients infected with acyclovir-resistant strains. To our knowledge, we present a previously unreported ex vivo HSV-1 infection model with abdominal human skin to test antiviral drugs, thus bridging the gap between in vitro and in vivo drug screening and providing a valuable preclinical platform for HSV research.
Collapse
Affiliation(s)
- Poojabahen Tajpara
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Ralf Schmidt
- Department of Laboratory Medicine, Division of Clinical Virology, Medical University of Vienna, Vienna, Austria
| | - Martin Vierhapper
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Johannes Matiasek
- Department of Plastic, Aesthetic and Reconstructive Surgery, St. Josef Hospital, Vienna, Austria
| | - Theresia Popow-Kraupp
- Department of Laboratory Medicine, Division of Clinical Virology, Medical University of Vienna, Vienna, Austria
| | - Christopher Schuster
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - Adelheid Elbe-Bürger
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
32
|
Alt M, Falk J, Eis-Hübinger AM, Kropff B, Sinzger C, Krawczyk A. Detection of antibody-secreting cells specific for the cytomegalovirus and herpes simplex virus surface antigens. J Immunol Methods 2018; 462:13-22. [PMID: 30056033 PMCID: PMC7094464 DOI: 10.1016/j.jim.2018.07.010] [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: 02/08/2018] [Revised: 06/29/2018] [Accepted: 07/25/2018] [Indexed: 11/24/2022]
Abstract
Infections with the herpes simplex virus (HSV) and the human cytomegalovirus (HCMV) can lead to life-threatening diseases, particularly in immunosuppressed patients. Furthermore, HSV infections at birth (herpes neonatorum) can result in a disseminated disease associated with a fatal multiorgan failure. Congenital HCMV infections can result in miscarriage, serious birth defects or developmental disabilities. Antibody-based interventions with hyperimmunoglobulins showed encouraging results in clinical studies, but clearly need to be improved. The isolation of highly neutralizing monoclonal antibodies is a promising strategy to establish potent therapy options against HSV and HCMV infections. Monoclonal antibodies are commonly isolated from hybridomas or EBV-immortalized B-cell clones. The screening procedure to identify virus-specific cells from a cell mixture is a challenging step, since most of the highly neutralizing antibodies target complex conformational epitopes on the virus surface. Conventional assays such as ELISA are based on purified viral proteins and inappropriate to display complex epitopes. To overcome this obstacle, we have established two full-virus based methods that allow screening for cells and antibodies targeting complex conformational epitopes on viral surface antigens. The methods are suitable to detect surface antigen-specific cells from a cell mixture and may facilitate the isolation of highly neutralizing antibodies against HSV and HCMV.
Collapse
Affiliation(s)
- Mira Alt
- Institute for Virology, University Hospital of Essen, 45147 Essen, Germany
| | - Jessica Falk
- Institute for Virology, University Hospital of Ulm, 89081 Ulm, Germany
| | | | - Barbara Kropff
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Christian Sinzger
- Institute for Virology, University Hospital of Ulm, 89081 Ulm, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital of Essen, 45147 Essen, Germany.
| |
Collapse
|
33
|
Falk JJ, Winkelmann M, Stöhr D, Alt M, Schrezenmeier H, Krawczyk A, Lotfi R, Sinzger C. Identification of Elite Neutralizers With Broad and Potent Neutralizing Activity Against Human Cytomegalovirus (HCMV) in a Population of HCMV-Seropositive Blood Donors. J Infect Dis 2018; 218:876-885. [DOI: 10.1093/infdis/jiy229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/16/2018] [Indexed: 11/14/2022] Open
Affiliation(s)
| | - Martina Winkelmann
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood-Transfusion Service Baden-Württemberg–Hessen and University Hospital, Ulm, Germany
| | - Dagmar Stöhr
- Institute for Virology, Ulm University Medical Center, Ulm, Germany
| | - Mira Alt
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood-Transfusion Service Baden-Württemberg–Hessen and University Hospital, Ulm, Germany
- Institute for Transfusion Medicine, Ulm University, Ulm, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ramin Lotfi
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood-Transfusion Service Baden-Württemberg–Hessen and University Hospital, Ulm, Germany
- Institute for Transfusion Medicine, Ulm University, Ulm, Germany
| | | |
Collapse
|
34
|
Ariza-Heredia EJ, Chemaly RF, Shahani LR, Jang Y, Champlin RE, Mulanovich VE. Delay of alternative antiviral therapy and poor outcomes of acyclovir-resistant herpes simplex virus infections in recipients of allogeneic stem cell transplant - a retrospective study. Transpl Int 2018; 31:639-648. [PMID: 29464765 DOI: 10.1111/tri.13142] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/23/2017] [Accepted: 02/14/2018] [Indexed: 12/16/2022]
Abstract
Acyclovir is commonly used to prevent and treat herpes simplex virus (HSV) reactivation after hematopoietic cell transplant (HCT), and only few reports have been published on acyclovir-resistant HSV in HCT recipients. We reviewed the medical records of patients with a microbiologic diagnosis of acyclovir-resistant HSV by plaque reduction test who received an HCT from 2002 through 2014. A total of 4 028 HCTs were performed during the study period, and 18 of the recipients met the diagnostic criteria for acyclovir-resistant HSV. All cases had undergone allogeneic HCTs. Most patients were in the pre-engraftment period or on systemic corticosteroid therapy for graft-versus-host disease (GVHD). The median time between diagnosis and susceptibility testing was 15 days, and antiviral therapy was changed at a median of 27 days. Patients required prolonged therapy (~80 days), and many had serious complications including renal failure and hospitalization. In conclusion, acyclovir-resistant HSV infection is more likely during the period of profound deficit in T-cell-mediated immunity and is associated with significant morbidities. Higher doses of acyclovir prophylaxis might be needed for patients with history of HSV during pre-engraftment or GVHD treatment. In patients who do not respond or progress after 1 week of acyclovir therapy, testing for drug-resistant HSV, and early switch to an alternative antiviral should be considered.
Collapse
Affiliation(s)
- Ella J Ariza-Heredia
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roy F Chemaly
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lokesh R Shahani
- McGovern Medical School, The University of Texas Science Center at Houston, Houston, TX, USA
| | - Ying Jang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victor E Mulanovich
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
35
|
Antiviral resistance in herpes simplex virus and varicella-zoster virus infections: diagnosis and management. Curr Opin Infect Dis 2018; 29:654-662. [PMID: 27306564 DOI: 10.1097/qco.0000000000000288] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Aciclovir (ACV) is the first-line drug for the management of herpes simplex virus (HSV) and varicella-zoster virus (VZV) infections. Long-term administration of ACV for the treatment of severe infections in immunocompromised patients can lead to the development of drug resistance. Furthermore, the emergence of isolates resistant to ACV is increasingly recognized in immunocompetent individuals with herpetic keratitis. This review describes the mechanisms involved in drug resistance for HSV and VZV, the laboratory diagnosis and management of patients with infections refractory to ACV therapy. RECENT FINDINGS Genotypic testing is more frequently performed for the diagnosis of infections caused by drug-resistant HSV or VZV isolates. Molecular biology-based systems for the generation of recombinant viruses have been developed to link unknown mutations with their drug phenotypes. Fast and sensitive methods based on next-generation sequencing will improve the detection of heterogeneous viral populations of drug-resistant viruses and their temporal changes during antiviral therapy, which could allow better patient management. Novel promising compounds acting on targets that differ from the viral DNA polymerase are under clinical development. SUMMARY Antiviral drug resistance monitoring for HSV and VZV is required for a rational use of antiviral therapy in high-risk populations.
Collapse
|
36
|
Dehghanpir SD, Birkenheuer CH, Yang K, Murelli RP, Morrison LA, Le Grice SFJ, Baines JD. Broad anti-herpesviral activity of α-hydroxytropolones. Vet Microbiol 2017; 214:125-131. [PMID: 29408023 DOI: 10.1016/j.vetmic.2017.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 01/02/2023]
Abstract
Herpesviruses are ubiquitous in animals and cause economic losses concomitant with many diseases. Most of the domestic animal herpesviruses are within the subfamily Alphaherpesvirinae, which includes human herpes simplex virus 1 (HSV-1). Suppression of HSV-1 replication has been reported with α-hydroxytropolones (αHTs), aromatic ring compounds that have broad bioactivity due to potent chelating activity. It is postulated that αHTs inhibit enzymes within the nucleotidyltransferase superfamily (NTS). These enzymes require divalent cations for nucleic acid cleavage activity. Potential targets include the nuclease component of the herpesvirus terminase (pUL15C), a highly conserved NTS-like enzyme that cleaves viral DNA into genomic lengths prior to packaging into capsids. Inhibition of pUL15C activity in biochemical assays by various αHTs previously revealed a spectrum of potencies. Interestingly, the most potent anti-pUL15C αHT inhibited HSV-1 replication to a limited extent in cell culture. The aim of this study was to evaluate three different αHT molecules with varying biochemical anti-pUL15C activity for a capacity to inhibit replication of veterinary herpesviruses (BoHV-1, EHV-1, and FHV-1) and HSV-1. Given the known discordant potencies between anti-pUL15C and HSV-1 replication inhibition, a second objective was to elucidate the mechanism of action of these compounds. The results show that αHTs broadly inhibit herpesviruses, with similar inhibitory effect against HSV-1, BoHV-1, EHV-1, and FHV-1. Based on immunoblotting, Southern blotting, and real-time qPCR, the compounds were found to specifically inhibit viral DNA replication. Thus, αHTs represent a new class of broadly active anti-herpesviral compounds with potential veterinary applications.
Collapse
Affiliation(s)
- Shannon D Dehghanpir
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Claire H Birkenheuer
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Kui Yang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Ryan P Murelli
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, PhD Program in Chemistry, The Graduate Center, The City University of New York, New York, NY, United States
| | - Lynda A Morrison
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine,St. Louis, MO, United States
| | - Stuart F J Le Grice
- Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Joel D Baines
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States.
| |
Collapse
|
37
|
Heilingloh CS, Krawczyk A. Role of L-Particles during Herpes Simplex Virus Infection. Front Microbiol 2017; 8:2565. [PMID: 29312245 PMCID: PMC5742154 DOI: 10.3389/fmicb.2017.02565] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 12/11/2017] [Indexed: 11/28/2022] Open
Abstract
Infection of eukaryotic cells with α-herpesviruses results in the formation and secretion of infectious heavy particles (virions; H-particles) and non-infectious light particles (L-particles). Herpes simplex virus type 1 (HSV-1) H-particles consist of a genome-containing capsid surrounded by tegument proteins and a glycoprotein-rich lipid bilayer. Non-infectious L-particles are composed mainly of envelope and tegument proteins and are devoid of capsids and viral DNA. L-particles were first described in the early nineties and from then on investigated for their formation and role during virus infection. The development and secretion of L-particles occur simultaneously to the assembly of complete viral particles. HSV-1 L-particles are assembled by budding of condensed tegument into Golgi-delivered vesicles and are capable of delivering their functional content to non-infected cells. Thereby, HSV-1 L-particles contribute to viral pathogenesis within the infected host by enhancing virion infectivity and providing immune evasion functions. In this review we discuss the emergence of HSV-1 L-particles during virus replication and their biological functions described thus far.
Collapse
Affiliation(s)
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
38
|
Bauer D, Keller J, Alt M, Schubert A, Aufderhorst UW, Palapys V, Kasper M, Heilingloh CS, Dittmer U, Laffer B, Eis-Hübinger AM, Verjans GM, Heiligenhaus A, Roggendorf M, Krawczyk A. Antibody-based immunotherapy of aciclovir resistant ocular herpes simplex virus infections. Virology 2017; 512:194-200. [DOI: 10.1016/j.virol.2017.09.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/20/2017] [Accepted: 09/22/2017] [Indexed: 11/29/2022]
|
39
|
Zinser E, Krawczyk A, Mühl-Zürbes P, Aufderhorst U, Draßner C, Stich L, Zaja M, Strobl S, Steinkasserer A, Heilingloh CS. A new promising candidate to overcome drug resistant herpes simplex virus infections. Antiviral Res 2017; 149:202-210. [PMID: 29155164 DOI: 10.1016/j.antiviral.2017.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 01/08/2023]
Abstract
Infections with Herpes simplex viruses (HSV) belong to the most common human diseases worldwide, resulting in symptoms ranging from painful, but commonly self-limiting lesions of the orofacial or genital tract to severe infections of the eye or life-threatening generalized infections. Frequent HSV-reactivations at the eye may lead to the development of herpetic stromal keratitis, which is one of the major causes of infectious blindness in developed countries. The vast majority of life-threatening generalized infections occur in immunocompromised individuals, such as transplant recipients or patients suffering from advanced human immunodeficiency virus (HIV) infection with concurrent HSV-reactivation. Over the past decades, Acyclovir (ACV) became the golden standard for the treatment of HSV infections. However, long-term antiviral treatment, as it is required mainly in immunocompromised patients, led to the emergence of resistances towards ACV and other antivirals. Therefore, there is a clear need for the development of new potent antivirals which combine good oral bioavailability and tolerability with low side effects. In the current study we present SC93305 as a novel potent antiviral substance that proved to be highly effective not only against different HSV-1 and HSV-2 strains but also towards ACV- and multi-resistant HSV-1 and HSV-2 isolates. SC93305 shows comparable antiviral activity as reported for ACV and very importantly it does not interfere with the activation of specific immune cells. Here we report that SC93305 does not affect the biological function of dendritic cells (DC), the most potent antigen presenting cells of the immune system to induce antiviral immune responses, nor T cell stimulation or the release of inflammatory cytokines. Thus, SC93305 is a new and promising candidate for the treatment of HSV-1 and HSV-2 infections and in particular also for the inhibition of drug-resistant HSV-1/2 strains.
Collapse
Affiliation(s)
- Elisabeth Zinser
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Ulrich Aufderhorst
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christina Draßner
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Mirko Zaja
- 4SC Discovery GmbH, Martinsried, Germany
| | | | | | | |
Collapse
|
40
|
Bauer D, Alt M, Dirks M, Buch A, Heilingloh CS, Dittmer U, Giebel B, Görgens A, Palapys V, Kasper M, Eis-Hübinger AM, Sodeik B, Heiligenhaus A, Roggendorf M, Krawczyk A. A Therapeutic Antiviral Antibody Inhibits the Anterograde Directed Neuron-to-Cell Spread of Herpes Simplex Virus and Protects against Ocular Disease. Front Microbiol 2017; 8:2115. [PMID: 29163407 PMCID: PMC5671610 DOI: 10.3389/fmicb.2017.02115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus (HSV) is a leading cause of blindness and viral encephalitis in the developed world. Upon reactivation from sensory neurons, HSV returns via axonal transport to peripheral tissues where it causes, e.g., severe, potentially blinding ocular diseases. In the present study we investigated whether the HSV-1/2 glycoprotein B-specific antibody mAb 2c or its humanized counterpart mAb hu2c can protect from ocular disease in a mouse model of HSV-1-induced acute retinal necrosis (ARN). In this model the viral spread from the initially infected to the contralateral eye resembles the routes taken in humans upon HSV reactivation. Systemic antibody treatment prior or early after infection effectively protected the mice from the development of ARN. These observations suggest that the antibody potently neutralized the infection and inhibited the viral transmission, since there was almost no virus detectable in the contralateral eyes and trigeminal ganglia of antibody treated mice. Besides of neutralizing free virus or limiting the infection via activating the complement or cellular effector functions, blocking of the anterograde directed neuron-to-cell spread of HSV represents a viable mode of action how mAb 2c protected the mice from ARN. We proved this hypothesis using a microfluidic chamber system. Neurons and epithelial cells were cultured in two separate compartments where the neurons sent axons via connecting microgrooves to the epithelial cells. Neurons were infected with a reporter HSV-1 strain expressing mCherry, and the co-culture was treated with neutralizing antibodies. In contrast to commercial polyclonal human HSV-neutralizing immunoglobulins, mAb 2c effectively blocked the anterograde directed neuron-to-cell transmission of the virus. Our data suggest that the humanized HSV-1/2-gB antibody protects mice from ocular disease by blocking the neuronal spread of HSV. Therefore, mAb hu2c may become a potent novel therapeutic option for severe ocular HSV infections.
Collapse
Affiliation(s)
- Dirk Bauer
- Department of Ophthalmology, Ophtha Lab, St. Franziskus-Hospital, Münster, Germany
| | - Mira Alt
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Miriam Dirks
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Ulf Dittmer
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Vivien Palapys
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Kasper
- Department of Ophthalmology, Ophtha Lab, St. Franziskus-Hospital, Münster, Germany
| | | | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Arnd Heiligenhaus
- Department of Ophthalmology, Ophtha Lab, St. Franziskus-Hospital, Münster, Germany.,Department of Ophthalmology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Roggendorf
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Adalbert Krawczyk
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
41
|
Vilas Boas LCP, de Lima LMP, Migliolo L, Mendes GDS, de Jesus MG, Franco OL, Silva PA. Linear antimicrobial peptides with activity against herpes simplex virus 1 and Aichi virus. Biopolymers 2017; 108. [PMID: 27161201 DOI: 10.1002/bip.22871] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/30/2016] [Accepted: 05/04/2016] [Indexed: 01/01/2023]
Abstract
Viruses are the major cause of disease and mortality worldwide. Nowadays there are treatments based on antivirals or prophylaxis with vaccines. However, the rising number of reports of viral resistance to current antivirals and the emergence of new types of virus has concerned the scientific community. In this scenario, the search for alternative treatments has led scientists to the discovery of antimicrobial peptides (AMPs) derived from many different sources. Since some of them have shown antiviral activities, here we challenged 10 synthetic peptides from different animal and plant sources against, herpes simplex virus 1 (HSV-1), and Aichi virus. Among them, the highlight was Pa-MAP from the polar fish Pleuronectes americanus, which caused around 90% of inhibition of the HSV with a selectivity index of 5 and a virucidal mechanism of action. Moreover, LL-37 from human neutrophils showed 96% of inhibition against the Aichi virus, showing a selectivity index of 3.4. The other evaluated peptides did not show significant antiviral activity. In conclusion, the present study demonstrated that Pa-MAP seems to be a reliable candidate for a possible alternative drug to treat HSV-1 infections. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 108: 1-6, 2017.
Collapse
Affiliation(s)
- Liana Costa Pereira Vilas Boas
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Lídia Maria Pinto de Lima
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Ludovico Migliolo
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil.,S-Inova Biotech, Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Gabriele Dos Santos Mendes
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Maianne Gonçalves de Jesus
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| | - Octávio Luiz Franco
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil.,S-Inova Biotech, Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil
| | - Paula Andréia Silva
- Centro De Análises Proteômicas E Bioquímicas: Programa De Pós-Graduação Em Ciências Genômicas E Biotecnologia, Universidade Católica De Brasília, Distrito Federal, Brazil
| |
Collapse
|
42
|
Clementi N, Criscuolo E, Cappelletti F, Quaranta P, Pistello M, Diotti RA, Sautto GA, Tarr AW, Mailland F, Concas D, Burioni R, Clementi M, Mancini N. Entry inhibition of HSV-1 and -2 protects mice from viral lethal challenge. Antiviral Res 2017; 143:48-61. [PMID: 28396205 DOI: 10.1016/j.antiviral.2017.03.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 11/26/2022]
Abstract
The present study focused on inhibition of HSV-1 and -2 replication and pathogenesis in vitro and in vivo, through the selective targeting of the envelope glycoprotein D. Firstly, a human monoclonal antibody (Hu-mAb#33) was identified that could neutralise both HSV-1 and -2 at nM concentrations, including clinical isolates from patients affected by different clinical manifestations and featuring different susceptibility to acyclovir in vitro. Secondly, the potency of inhibition of both infection by cell-free viruses and cell-to-cell virus transmission was also assessed. Finally, mice receiving a single systemic injection of Hu-mAb#33 were protected from death and severe clinical manifestations following both ocular and vaginal HSV-1 and -2 lethal challenge. These results pave the way for further studies reassessing the importance of HSV entry as a novel target for therapeutic intervention and inhibition of cell-to-cell virus transmission.
Collapse
Affiliation(s)
- Nicola Clementi
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy.
| | - Elena Criscuolo
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy
| | | | - Paola Quaranta
- Department of Translational Research, University of Pisa, Pisa, Italy
| | - Mauro Pistello
- Department of Translational Research, University of Pisa, Pisa, Italy
| | - Roberta A Diotti
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy
| | - Giuseppe A Sautto
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy
| | - Alexander W Tarr
- School of Life Sciences & NIHR Biomedical Research Unit in Gastrointestinal & Liver Diseases, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Daniela Concas
- Wezen Bio AG, Fondation pour Recherches Medicales, Geneva, Switzerland
| | - Roberto Burioni
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy; Laboratory of Microbiology and Virology, San Raffaele Hospital, Milan, Italy
| | - Massimo Clementi
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy; Laboratory of Microbiology and Virology, San Raffaele Hospital, Milan, Italy
| | - Nicasio Mancini
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, Milan, Italy; Laboratory of Microbiology and Virology, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
43
|
Wahid B, Ali A, Idrees M, Rafique S. Immunotherapeutic strategies for sexually transmitted viral infections: HIV, HSV and HPV. Cell Immunol 2016; 310:1-13. [PMID: 27514252 PMCID: PMC7124316 DOI: 10.1016/j.cellimm.2016.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/22/2016] [Accepted: 08/02/2016] [Indexed: 12/24/2022]
Abstract
More than 1 million sexually transmitted infections (STIs) are acquired each day globally. Etiotropic drugs cannot effectively control infectious diseases therefore, there is a dire need to explore alternative strategies especially those based on the regulation of immune system. The review discusses all rational approaches to develop better understanding towards immunotherapeutic strategies based on modulation of immune system in an attempt to curb the elevating risk of infectious diseases such as HIV, HPV and HSV because of their high prevalence. Development of monoclonal antibodies, vaccines and several other immune based treatments are promising alternative strategies that are offering new opportunities to eradicate pathogens.
Collapse
Affiliation(s)
- Braira Wahid
- Centre for Applied Molecular Biology, 87-West Canal Bank Road, Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan.
| | - Amjad Ali
- Centre for Applied Molecular Biology, 87-West Canal Bank Road, Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan.
| | - Muhammad Idrees
- Centre for Applied Molecular Biology, 87-West Canal Bank Road, Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan; Vice Chancellor Hazara University Mansehra, Pakistan.
| | - Shazia Rafique
- Centre for Applied Molecular Biology, 87-West Canal Bank Road, Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
44
|
Edwards JL, Jennings MP, Apicella MA, Seib KL. Is gonococcal disease preventable? The importance of understanding immunity and pathogenesis in vaccine development. Crit Rev Microbiol 2016; 42:928-41. [PMID: 26805040 PMCID: PMC4958600 DOI: 10.3109/1040841x.2015.1105782] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/01/2015] [Accepted: 10/06/2015] [Indexed: 12/31/2022]
Abstract
Gonorrhea is a major, global public health problem for which there is no vaccine. The continuing emergence of antibiotic-resistant strains raises concerns that untreatable Neisseria gonorrhoeae may become widespread in the near future. Consequently, there is an urgent need for increased efforts towards the development of new anti-gonococcal therapeutics and vaccines, as well as suitable models for potential pre-clinical vaccine trials. Several current issues regarding gonorrhea are discussed herein, including the global burden of disease, the emergence of antibiotic-resistance, the status of vaccine development and, in particular, a focus on the model systems available to evaluate drug and vaccine candidates. Finally, alternative approaches to evaluate vaccine candidates are presented. Such approaches may provide valuable insights into the protective mechanisms, and correlates of protection, required to prevent gonococcal transmission, local infection and disease sequelae.
Collapse
Affiliation(s)
- Jennifer L. Edwards
- Department of Pediatrics, The Research Institute at Nationwide Children's Hospital and The Ohio State UniversityColumbus,
OH,
USA
| | | | | | - Kate L. Seib
- Institute for Glycomics, Griffith University,
Gold Coast,
Australia
| |
Collapse
|
45
|
Heikkilä O, Nygårdas M, Paavilainen H, Ryödi E, Hukkanen V. Interleukin-27 Inhibits Herpes Simplex Virus Type 1 Infection by Activating STAT1 and 3, Interleukin-6, and Chemokines IP-10 and MIG. J Interferon Cytokine Res 2016; 36:617-629. [DOI: 10.1089/jir.2016.0015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Outi Heikkilä
- Department of Virology, University of Turku, Turku, Finland
| | | | - Henrik Paavilainen
- Department of Virology, University of Turku, Turku, Finland
- Drug Research Doctoral Programme, University of Turku, Turku, Finland
| | - Elina Ryödi
- Department of Virology, University of Turku, Turku, Finland
| | - Veijo Hukkanen
- Department of Virology, University of Turku, Turku, Finland
| |
Collapse
|
46
|
Converting monoclonal antibody-based immunotherapies from passive to active: bringing immune complexes into play. Emerg Microbes Infect 2016; 5:e92. [PMID: 27530750 PMCID: PMC5034104 DOI: 10.1038/emi.2016.97] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/13/2022]
Abstract
Monoclonal antibodies (mAbs), which currently constitute the main class of biotherapeutics, are now recognized as major medical tools that are increasingly being considered to fight severe viral infections. Indeed, the number of antiviral mAbs developed in recent years has grown exponentially. Although their direct effects on viral blunting have been studied in detail, their potential immunomodulatory actions have been overlooked until recently. The ability of antiviral mAbs to modulate antiviral immune responses in infected organisms has recently been revealed. More specifically, upon recognition of their cognate antigens, mAbs form immune complexes (ICs) that can be recognized by the Fc receptors expressed on different immune cells of infected individuals. This binding may be followed by the modulation of the host immune responses. Harnessing this immunomodulatory property may facilitate improvements in the therapeutic potential of antiviral mAbs. This review focuses on the role of ICs formed with different viral determinants and mAbs in the induction of antiviral immune responses in the context of both passive immunotherapies and vaccination strategies. Potential deleterious effects of ICs on the host immune response are also discussed.
Collapse
|
47
|
Pelegrin M, Naranjo-Gomez M, Piechaczyk M. Antiviral Monoclonal Antibodies: Can They Be More Than Simple Neutralizing Agents? Trends Microbiol 2016; 23:653-665. [PMID: 26433697 PMCID: PMC7127033 DOI: 10.1016/j.tim.2015.07.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/06/2015] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) are increasingly being considered as agents to fight severe viral diseases. So far, they have essentially been selected and used on the basis of their virus-neutralizing activity and/or cell-killing activity to blunt viral propagation via direct mechanisms. There is, however, accumulating evidence that they can also induce long-lasting protective antiviral immunity by recruiting the endogenous immune system of infected individuals during the period of immunotherapy. Exploiting this property may revolutionize antiviral mAb-based immunotherapies, with benefits for both patients and healthcare systems. Antiviral monoclonal antibodies (mAbs) are promising, high-added-value biotherapeutics. During recent years, the number of antiviral mAbs developed against both acute and chronic viruses has grown exponentially, some of them being currently tested in clinical trials. Antiviral mAbs can be used to blunt viral propagation through direct effects. They can also engage the host's immune system, leading to the induction of long-lasting protective vaccine-like effects. The assessment of mechanisms at play in the induction of vaccine-like effects by antiviral mAbs will help in improving antiviral treatments. Exploiting this effect will translate into therapeutic benefit for patients. The benefit will also help healthcare systems through the reduction of treatment costs.
Collapse
Affiliation(s)
- Mireia Pelegrin
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France.
| | - Mar Naranjo-Gomez
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Marc Piechaczyk
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| |
Collapse
|
48
|
Clementi N, Criscuolo E, Cappelletti F, Burioni R, Clementi M, Mancini N. Novel therapeutic investigational strategies to treat severe and disseminated HSV infections suggested by a deeper understanding of in vitro virus entry processes. Drug Discov Today 2016; 21:682-91. [PMID: 26976690 DOI: 10.1016/j.drudis.2016.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/11/2016] [Accepted: 03/04/2016] [Indexed: 01/28/2023]
Abstract
The global burden of herpes simplex virus (HSV) legitimates the critical need to develop new prevention strategies, such as drugs and vaccines that are able to fight either primary HSV infections or reactivations. Moreover, the ever-growing number of patients receiving transplants increases the number of severe HSV infections that are unresponsive to current therapies. Finally, the high global incidence of genital HSV-2 infection increases the risk of perinatal transmission to newborns, in which disseminated infection or central nervous system (CNS) involvement is frequent, with associated high morbidity and mortality rates. There are several key features shared by novel anti-HSV drugs, from currently available optimized drugs to small molecules able to interfere with various virus replication steps. However, several virological aspects of the disease and associated clinical needs highlight why an ideal anti-HSV drug has yet to be developed.
Collapse
Affiliation(s)
- Nicola Clementi
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, 20132 Milan, Italy.
| | - Elena Criscuolo
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, 20132 Milan, Italy
| | - Francesca Cappelletti
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, 20132 Milan, Italy
| | - Roberto Burioni
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, 20132 Milan, Italy
| | - Massimo Clementi
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, 20132 Milan, Italy
| | - Nicasio Mancini
- Microbiology and Virology Unit, 'Vita-Salute San Raffaele' University, 20132 Milan, Italy
| |
Collapse
|
49
|
Le-Trilling VTK, Megger DA, Katschinski B, Landsberg CD, Rückborn MU, Tao S, Krawczyk A, Bayer W, Drexler I, Tenbusch M, Sitek B, Trilling M. Broad and potent antiviral activity of the NAE inhibitor MLN4924. Sci Rep 2016; 6:19977. [PMID: 26829401 PMCID: PMC4734293 DOI: 10.1038/srep19977] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/22/2015] [Indexed: 02/01/2023] Open
Abstract
In terms of infected human individuals, herpesviruses range among the most successful virus families. Subclinical herpesviral infections in healthy individuals contrast with life-threatening syndromes under immunocompromising and immunoimmature conditions. Based on our finding that cytomegaloviruses interact with Cullin Roc ubiquitin ligases (CRLs) in the context of interferon antagonism, we systematically assessed viral dependency on CRLs by utilizing the drug MLN4924. CRL activity is regulated through the conjugation of Cullins with the ubiquitin-like molecule Nedd8. By inhibiting the Nedd8-activating Enzyme (NAE), MLN4924 interferes with Nedd8 conjugation and CRL activity. MLN4924 exhibited pronounced antiviral activity against mouse and human cytomegalovirus, herpes simplex virus (HSV)- 1 (including multi-drug resistant clinical isolates), HSV-2, adeno and influenza viruses. Human cytomegalovirus genome amplification was blocked at nanomolar MLN4924 concentrations. Global proteome analyses revealed that MLN4924 blocks cytomegaloviral replication despite increased IE1 amounts. Expression of dominant negative Cullins assigned this IE regulation to defined Cullin molecules and phenocopied the antiviral effect of MLN4924.
Collapse
Affiliation(s)
| | - Dominik A Megger
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Bochum, Germany
| | - Benjamin Katschinski
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christine D Landsberg
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Meike U Rückborn
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sha Tao
- Institute for Virology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Adalbert Krawczyk
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ingo Drexler
- Institute for Virology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr Universität Bochum, Bochum, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
50
|
Masaoka T, Zhao H, Hirsch DR, D'Erasmo MP, Meck C, Varnado B, Gupta A, Meyers MJ, Baines J, Beutler JA, Murelli RP, Tang L, Le Grice SFJ. Characterization of the C-Terminal Nuclease Domain of Herpes Simplex Virus pUL15 as a Target of Nucleotidyltransferase Inhibitors. Biochemistry 2016; 55:809-19. [PMID: 26829613 DOI: 10.1021/acs.biochem.5b01254] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The natural product α-hydroxytropolones manicol and β-thujaplicinol inhibit replication of herpes simplex viruses 1 and 2 (HSV-1 and HSV-2, respectively) at nontoxic concentrations. Because these were originally developed as divalent metal-sequestering inhibitors of the ribonuclease H activity of HIV-1 reverse transcriptase, α-hydroxytropolones likely target related HSV proteins of the nucleotidyltransferase (NTase) superfamily, which share an "RNase H-like" fold. One potential candidate is pUL15, a component of the viral terminase molecular motor complex, whose C-terminal nuclease domain, pUL15C, has recently been crystallized. Crystallography also provided a working model for DNA occupancy of the nuclease active site, suggesting potential protein-nucleic acid contacts over a region of ∼ 14 bp. In this work, we extend crystallographic analysis by examining pUL15C-mediated hydrolysis of short, closely related DNA duplexes. In addition to defining a minimal substrate length, this strategy facilitated construction of a dual-probe fluorescence assay for rapid kinetic analysis of wild-type and mutant nucleases. On the basis of its proposed role in binding the phosphate backbone, studies with pUL15C variant Lys700Ala showed that this mutation affected neither binding of duplex DNA nor binding of small molecule to the active site but caused a 17-fold reduction in the turnover rate (kcat), possibly by slowing conversion of the enzyme-substrate complex to the enzyme-product complex and/or inhibiting dissociation from the hydrolysis product. Finally, with a view of pUL15-associated nuclease activity as an antiviral target, the dual-probe fluorescence assay, in combination with differential scanning fluorimetry, was used to demonstrate inhibition by several classes of small molecules that target divalent metal at the active site.
Collapse
Affiliation(s)
- Takashi Masaoka
- Basic Research Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | - Haiyan Zhao
- Department of Molecular Biosciences, University of Kansas , Lawrence, Kansas 66045, United States
| | - Danielle R Hirsch
- Department of Chemistry, Brooklyn College, City University of New York , Brooklyn, New York 11210, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| | - Michael P D'Erasmo
- Department of Chemistry, Brooklyn College, City University of New York , Brooklyn, New York 11210, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| | - Christine Meck
- Department of Chemistry, Brooklyn College, City University of New York , Brooklyn, New York 11210, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| | - Brittany Varnado
- Department of Molecular Biosciences, University of Kansas , Lawrence, Kansas 66045, United States
| | - Ankit Gupta
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine , St. Louis, Missouri 63104, United States
| | - Marvin J Meyers
- Department of Chemistry, St. Louis University , St. Louis, Missouri 63103, United States
| | - Joel Baines
- School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana 70803, United States
| | - John A Beutler
- Molecular Targets Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | - Ryan P Murelli
- Department of Chemistry, Brooklyn College, City University of New York , Brooklyn, New York 11210, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , New York, New York 10016, United States
| | - Liang Tang
- Department of Molecular Biosciences, University of Kansas , Lawrence, Kansas 66045, United States
| | - Stuart F J Le Grice
- Basic Research Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| |
Collapse
|