Recent Updates on Viral Oncogenesis: Available Preventive and Therapeutic EntitiesClick to copy article linkArticle link copied!
- Shivam ChowdharyShivam ChowdharyDepartment of Industrial Microbiology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, Uttar Pradesh IndiaMore by Shivam Chowdhary
- Rahul DekaRahul DekaDepartment of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, IndiaMore by Rahul Deka
- Kingshuk PandaKingshuk PandaDepartment of Applied Microbiology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, IndiaMore by Kingshuk Panda
- Rohit KumarRohit KumarDepartment of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, IndiaMore by Rohit Kumar
- Abhishikt David SolomonAbhishikt David SolomonDepartment of Molecular & Cellular Engineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, Uttar Pradesh, IndiaMore by Abhishikt David Solomon
- Jimli DasJimli DasCentre for Biotechnology and Bioinformatics, Dibrugarh University, Assam 786004, IndiaMore by Jimli Das
- Supriya KanoujiyaSupriya KanoujiyaSchool of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, IndiaMore by Supriya Kanoujiya
- Ashish Kumar GuptaAshish Kumar GuptaDepartment of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, IndiaMore by Ashish Kumar Gupta
- Somya SinhaSomya SinhaDepartment of Biotechnology, Graphic Era Deemed to Be University, Dehradun 248002, Uttarakhand, IndiaMore by Somya Sinha
- Janne Ruokolainen*Janne Ruokolainen*Email: [email protected]Department of Applied Physics, School of Science, Aalto University, 02150 Espoo, FinlandMore by Janne Ruokolainen
- Kavindra Kumar Kesari*Kavindra Kumar Kesari*Email: [email protected]Department of Applied Physics, School of Science, Aalto University, 02150 Espoo, FinlandDivision of Research and Development, Lovely Professional University, Phagwara 144411, Punjab, IndiaMore by Kavindra Kumar Kesari
- Piyush Kumar Gupta*Piyush Kumar Gupta*Email: [email protected]Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, IndiaDepartment of Biotechnology, Graphic Era Deemed to Be University, Dehradun 248002, Uttarakhand, IndiaFaculty of Health and Life Sciences, INTI International University, Nilai 71800, MalaysiaMore by Piyush Kumar Gupta
Abstract
Human viral oncogenesis is a complex phenomenon and a major contributor to the global cancer burden. Several recent findings revealed cellular and molecular pathways that promote the development and initiation of malignancy when viruses cause an infection. Even, antiviral treatment has become an approach to eliminate the viral infections and prevent the activation of oncogenesis. Therefore, for a better understanding, the molecular pathogenesis of various oncogenic viruses like, hepatitis virus, human immunodeficiency viral (HIV), human papillomavirus (HPV), herpes simplex virus (HSV), and Epstein-Barr virus (EBV), could be explored, especially, to expand many potent antivirals that may escalate the apoptosis of infected malignant cells while sparing normal and healthy ones. Moreover, contemporary therapies, such as engineered antibodies antiviral agents targeting signaling pathways and cell biomarkers, could inhibit viral oncogenesis. This review elaborates the recent advancements in both natural and synthetic antivirals to control viral oncogenesis. The study also highlights the challenges and future perspectives of using antivirals in viral oncogenesis.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
1. Introduction
2. Viruses Associated with Oncogenesis
2.1. Hepatitis B Virus (HBV)
2.2. Human Papillomavirus (HPV)
2.3. Kaposi-Sarcoma Associated Herpesvirus (KSHV)/Human Herpesvirus-8 (HHV-8)
2.4. Epstein–Barr Virus (EBV)/Human Herpes Virus 4 (HHV-4)
2.5. Merkel Cell Polyomavirus (MCPyV)
2.6. Hepatitis C Virus (HCV)
S. No. | Viruses | Genome | Types of Cancer | References |
---|---|---|---|---|
1. | Merkel cell polyomavirus (MCPyV) | Double-stranded DNA Polyomaviridae | Merkel cell carcinoma | (25) |
2. | Hepatitis B virus (HBV) | Partially double-stranded DNA Hepadnaviridae | Hepatocellular carcinoma | (81) |
3. | Human Papilloma virus (HPV) | Double-stranded DNA Papillomaviridae | Cervical, neck, head, and anogenital tract carcinoma | (82,83) |
4. | Kaposi sarcoma herpesvirus (KSHV/ HHV-8) | Double-stranded DNA Herpesviridae | Primary effusion lymphoma, Kaposi sarcoma, multicentric Castleman disease | (84) |
5. | Epstein–Barr virus (EBV/HHV-4) | Double-stranded DNA Herpesviridae | Burkitt lymphoma, Hodgkin lymphoma, nasopharyngeal carcinoma | (85) |
6. | Hepatitis C virus (HCV) | Single-stranded RNA Flaviviridae | Hepatocellular carcinoma, Lymphomas | (86) |
7. | Human T-cell leukemia virus- (HTLV-1) | Single-stranded RNA Retroviridae | Adult T-cell leukemia and myelopathy/tropical spastic paraparesis | (87) |
8. | Human immunodeficiency virus (HIV) | Double-stranded RNA Retroviridae | Elevates the immunosuppression-mediated malignancies by other oncogenic viruses | (88) |
2.7. Human T-cell Lymphotropic Virus (HTLV-I)
2.8. HIV (Human Immunodeficiency Virus)
3. Antiviral Strategies
A. Chemical Antivirals | ||||
---|---|---|---|---|
Viruses | Antivirals | Functions | Target | References |
Epstein–Barr virus | Ganciclovir | Prevent the progression of viral DNA, inhibits host cell DNA polymerase | Viral DNA | (3) |
Acyclovir | ||||
Famciclovir | ||||
Rituximab (Monoclonal antibody) | Targets all B cells expressing CD20 (not specifically target cells containing EBV) | B cells | ||
Rapamycin (Immunosuppressive agent) | Decrease tumor growth and metastasis in a mouse model of EBV-associated oncogenesis | P13K Pathway | ||
Valganciclovir + Alemtuzumab | Suppresses replication of EBV | Reactivation of EBV by targeting CD52 on B- and T-lymphocyte) | (248−250) | |
Maribavir | Inhibits the EBV protein kinases | Viral DNA | (251) | |
Cidofovir | Inhibitors of murine herpesvirus replication and inhibits ribonucleotide reductase (RR) | Phosphodiesterase and EBV-transformed epithelial cells/Xenografts | (249,251) | |
Rutamarin | Inhibiting replication of viral DNA | Topoisomerase II | (251) | |
Hepatitis B and C virus | Lamivudine | HBV DNA chain termination | Reverse transcriptase | (252) |
Adefovir | Inhibits HBV DNA synthesis | Reverse transcriptase | (252,253) | |
Entecavir | Inhibits all steps in the HBV viral replication | HBV polymerase | (254,255) | |
Telbivudine | Inhibits HBV DNA chain termination and viral replication | Second strand of DNA and HBV DNA polymerase (reverse transcriptase) | (256−258) | |
Tenofovir | Reverse transcriptase | (259) | ||
Boceprevir | Inhibits viral HCV replication as well as disrupts the processing of viral proteins | HCV nonstructural 3/4A protease inhibitors | (252) | |
Telaprevir | ||||
Sofosbuvir | Executes chain termination and prevents viral replication of HCV | Inhibits HCV NS5B (nonstructural protein 5B) RNA-dependent RNA polymerase | (260) | |
Simeprevir | Blocks the function of adapters protein | Polyprotein of HCV | (261) | |
Faldaprevir | Cleaves the HCV-encoded polyprotein | NS3/4A protease | (262) | |
Ribavirin | Depletion of intracellular GTP and increased hepatitis C virus mutagenesis | ITP pyrophosphatase (ITPase) | (263) | |
Asunaprevir | Inhibits viral replication | HCV NS3 protease | (264) | |
Danoprevir | Prevents the cleavage and processing of HCV viral proteins | HCV NS3/4A protease | (265) | |
Vaniprevir | Inhibits the enzymatic activity | HCV NS3/4A protease | (266) | |
Daclatasvir | Blocks both virion assembly/secretion in vivo and intracellular viral RNA synthesis | HCV NS5A proteins | (267) | |
Ledipasvir | Prevents hyperphosphorylation of proteins for viral production | (268) | ||
Ombitasvir | Blocking signaling interactions and modification of the HCV replication complex | (269,270) | ||
Human Papillomavirus | Cidofovir | Converted to its active form as triphosphorylated cidofovir and is likely to induce chain termination | HPV DNA | (271,272) |
GS-9191 | Inhibits DNA synthesis | DNA polymerase α and ß | (273) | |
ODE-Bn-PMEG/ABI-1968 | Inhibits HPV origin-dependent plasmid amplification | HPV DNA | (274) | |
Acyclovir | Inhibits the integration of viral DNA and the replication of cellular DNA | HPV DNA | (275) | |
Imiquimod | Stimulates the innate and acquired immune responses followed by apoptosis of infected tissues | Toll-like receptors | (276) | |
Cimetidine | Improves cellular immunity and wart remission | Stimulates Th1 cells to catalyze interleukins (IL)-2, IL-12, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ | (276) | |
Bleomycin | Eliminates pyrimidine and purine bases and affects cellular DNA synthesis | Cellular DNA | (277) | |
Merkel Cell Polyomavirus | Pembrolizumab | Counteracts its interaction with its known ligands | Programmed cell death protein 1 | |
Avelumab | 1. Blocks the interaction between PD-L1 and its receptors PD-1 and B7.1 | Programmed cell death 1, Ligand 1 | (278,279) | |
2. Stimulates ADCC in addition to immune checkpoint inhibition | ||||
Nivolumab | Prevents PD-L1 and PD-L2 from inhibiting the action of T cells | Programmed cell death protein 1 | (280) | |
Ipilimumab | Defects in the DNA repair machinery | CTLA-4 protein | (281) | |
Kaposi Sarcoma Herpesvirus | Cidofovir | Inhibitors of murine herpesvirus replication and inhibits ribonucleotide reductase (RR) | Phosphodiesterase and EBV-transformed epithelial cells/Xenografts | (249) |
Ganciclovir | 1. Prevents the progression of viral DNA, inhibits host cell DNA polymerase | Viral DNA | (3,282) | |
2. Reduces plasma viral load of KSHV and can prevent KS in KSHV-seropositive transplant recipients | ||||
Bortezomib | Stabilizes the cellular proteins involved in suppressing cellular proliferation and promotes apoptosis, including p21, p27, p53, and I kappa-B | Human 26 S proteasome | (283) | |
Tocilizumab (Antibody) | Blocks membrane-bound and soluble gp80 signal transduction | gp80 | (284) | |
Siltuximab (Monoclonal antibody) | Inhibits binding to soluble and membrane-bound IL-6 receptors | Interleukin 6 | (285) | |
Pomalidomide | Induces and elevates the B7–2 and ICAM-1 expression in PEL cells | Activation of T cells and NK cell-mediated killing of PEL cells | (286) | |
Bevacizumab (Monoclonal antibody) | Exerts its effects by binding and inactivating serum VEGF and is unable to interact with its cell surface receptors, thereby proangiogenic signaling is terminated | VEGF-A, subcomponents subunits (A, B, C), and Affinity Immunoglobulin Fc gamma receptors | (286,287) | |
Doxorubicin | Complexes are formed with DNA by intercalation between base pairs, and it inhibits topoisomerase II action by stabilizing the complex of DNA–topoisomerase II | DNA topoisomerase 2-alpha and DNA | (288) | |
Human Immunodeficiency Virus (HIV) | Saquinavir + Ritonavir | Hydroxyethylene scaffold mimics the HIV protease for cleaving, thereby carrying out the proteolysis of Gag polyprotein and producing immature and noninfectious particles | HIV protease | (289−292) |
Lopinavir | ||||
Indinavir | ||||
Nelfinavir | ||||
Amprenavir | ||||
Atazanavir | ||||
Tipranavir | ||||
Darunavir + Ritonavir | Prevents viral replication | HIV protease | (293,294) | |
Lamivudine | Prevents the formation of phosphodiester bonds between the Nucleotide Reverse Transcriptase Inhibitors and nucleoside triphosphate | Reverse transcriptase | (295−297) | |
Zidovudine | ||||
Abacavir | ||||
Didanosine | ||||
Emtricitabine | ||||
Stavudine | ||||
Zalcitabine | ||||
Tenofovir disoprovil fumarate | ||||
Entravirine | Non-Nucleoside Reverse Transcriptase Inhibitor changes the spatial conformation of the substrate binding site and reduces the polymerase activity | Reverse transcriptase | (298) | |
Delavirdine | ||||
Efavirenz | ||||
Nevirapine | ||||
Rilpivirine (Phase 3) | ||||
Raltegravir | 1. Binds between the integrase and viral DNA to a specific complex | Integrase (Strand transfer reaction) | (299−301) | |
MK-0518 | 2. Interaction takes place between the inhibitor and Mg metal ion in the active site of integrase as well as the DNA | |||
Elvitegravir | ||||
Human T-lymphotropic Virus (HTLV) | Alemtuzumab | Blocks the TAC antigen | ATLL cells and HTLV1-infected cells | (302) |
Arsenic trioxide + Interferon-α | Induces cell cycle inhibition and apoptosis | HTLV1-infected cells and malignant ATLL cells | (303,304) | |
Lamivudine | Can inhibit viral replication in HTLV1-infected cells in single-use or combination therapy | Nucleoside analogs | (305,306) | |
Abacavir | ||||
Zidovudine | ||||
Didanosine | ||||
Emtricitabine | ||||
Stavudine | ||||
Zalcitabine | ||||
Tenofovir disoprovil fumarate | ||||
Raltegravir | Potent to inhibit strand transfer reaction and number of integration of events directly | HTLV1 Integrase | (307) | |
Romidepsin | 1. Can suppress the expression of NF-κB and AP-1 | ATLL cells and HTLV1-infected cells | (308,309) | |
2. Induces apoptosis of HTLV-1 infected cells and ATLL cells | ||||
Niclosamide | Induces degradation in protein and inhibits viral gene transcription of HTLV1 | Tax protein | (310) | |
Chondroitin Sulfate Type E | Interacts with the recombinant envelope protein of the virus at the C-terminus and blocks the binding of the virus to the human T cell | Human T cell | (311) | |
Darunavir | Prevents viral replication | HTLV1 Protease | (312) | |
Arsenic trioxide + Interferon-α | Induces cell cycle inhibition and apoptosis | HTLV1-infected cells and malignant ATLL cells | (303,304) |
B. Natural Antivirals | |||
---|---|---|---|
Antivirals | Function | Virus/Target | References |
Terminalia bellerica | Inhibits viral entry, replication, and maturation of HBV particles | Hepatitis B virus | (313−315) |
Phyllanthus Amarus | |||
Hybanthusenneaspermus | |||
Enicostemmaaxillare | |||
Bombyx mori L | |||
Boehmeria nivea | |||
Trichiliadregeana | Inhibits viral entry | Hepatitis C virus | |
Detarium microcarpum | |||
Phragmanthera capitata | |||
Bupleurum kaoi | |||
Anthocyanidin | |||
Alloeocomatellapolycladia | Suppression of the helicase activity of HCV NS3 | ||
Fusarium equiseti | Inhibition of HCV NS3/4A protease | ||
Eclipta alba | Inhibition of HCV NS5B replicase activity | ||
Taraxacum officinale | |||
Swietenia macrophylla | Reduction of HCV protein and HCV–RNA levels | ||
Entada africana | Broad antiviral activity | ||
Grape seed | Suppression of HCV-induced Cox-2 | ||
Flavanone | Release/Assembly | ||
Curcuma longaL | Inhibits immediate–early gene expression | Herpes simplex virus 1 | |
Houttuynia cordata | Blocks viral binding and suppresses NF-κB activation | Herpes simplex virus 1 | |
Herpes simplex virus 2 | |||
Pistacia vera L | Inhibits expression of HSV-1 viral proteins and viral DNA synthesis | Herpes simplex virus 1 | |
Prunus dulcis | Blocks viral binding | ||
Aloe Vera | Reduction of cytopathic effect (CPE) | ||
Cephalotaxaceae | Targets the cellular factor eIF4E | ||
Curcuma longa L | Downregulating expression of oncogenes E6 and E7 | HPV-16 | |
HPV-18 | |||
Ficus carica | HPV-16 | ||
C. longa, A. indica, E. officinalis, A. vera | Prevents the entry of HPV 16 in Hela cells | ||
Green tea | Promoting apoptosis and inhibiting cellular transcriptional factors | ||
Pinelliapedatisecta | HPV-16 | ||
HPV-18 | |||
Cudraniatricuspidata | HPV-16 | ||
Curcuma longa L | HPV-18 | ||
HPV-16 | |||
Bryophyllum pinnata | HPV-18 | ||
Phyllanthus emblica | HPV-16 | ||
HPV-18 | |||
Kaempferia parviflora | HPV-16 | ||
Podophyllum | Mitotic arrest in cell cycle | Genital warts | (316) |
Nostoc ellipsosporum | Breaching inhibitors | HIV | (313−315) |
Griffithsia sp. | |||
Stellettaclavosa | |||
Siliquariaspongia mirabilis | |||
Syzygiumclaviflorum | Maturation inhibitors | ||
Synthetic derivative of betulinic acid | |||
Rheum palmatum | Integrase inhibitors and reverse transcriptase | ||
Morus nigra | |||
Justica gendarussa | |||
Calophyllumlanigerum | |||
Euphorbia kansui | Latency-reversing agents | ||
Theobroma cacao | |||
Andrographis paniculata | Inhibits transcription of IE genes in EVB and the production of virions | Epstein–Barr Virus | (249) |
Polygonum cuspidatum | Inhibits the lytic cycle of EBV | ||
Saururus chinensis | Inhibits replication of EBV lytic cycle | ||
Rhus chinensis | Reduces the number of EBV particles | ||
Thelypteris torresiana | Inhibiting the EBV virus lytic cycle | ||
Psoralea corylifolia | Inhibits early steps of the replicative lytic cycle in EBV | ||
Angelica archangelica | |||
Borrelidin | Inhibits tRNA synthesis | Merkel Cell Carcinoma | (317) |
Mechlorethamine | Causes DNA damage | ||
Plumbagin | ROS/redox-active, proteasome | ||
Mitomycin C | Causes DNA damage | ||
Cladribine | Inhibits DNA synthesis | ||
Clofarabine | |||
Pyrrolidine dithiocarbamate | ROS, proteasome, NFκB | ||
Etoposide | Topoisomerase II/causes DNA damage | ||
Gloxazone | Inhibits DNA synthesis | ||
Panobinostat | Histone deacetylase inhibitors/ROS active | ||
Disulfiram | ROS, proteasome, NF-κB | ||
Thapsigargin | Stress on the endoplasmic reticulum | ||
Englerin A | Protein Kinase C | ||
Furoxanobenzofuroxan | Inhibits monoamine oxidase | ||
5-Nitroso-8-quinolinol | Histone deacetylase inhibitors/ROS active | ||
Phytolacca americana | Depurinates nucleotides/feedback inhibition of HTLV-I gene expression | Human T-lymphotropic virus 1 | (318) |
Scutellaria baicalensis georgi | Inhibits reverse transcriptase activity in HTLV-I-infected cells | (319) | |
Curcumin metabolites (Curcumin-sulfate, curcumin-glucuronide, and tetrahydro-curcumin) | Potent HTLV1 protease inhibitors | (320) | |
Oleandrin | Inhibits virological synapse formation | (321) |
4. Recent Progress on Drug Developments for Oncogenic Virus
S. No. | Title | Intervention | Outcome measures | Sponsors/ Collaborators | Phase | Enrollment | NCT Number |
---|---|---|---|---|---|---|---|
(i) MCPyV | |||||||
1. | Pembrolizumab (MK-3475) as First-line Therapy for Advanced Merkel Cell Carcinoma (MK-3475-913) | Drug: Pembrolizumab (MK-3475) | Objective Response Rate (ORR), Duration of Response (DOR), Progression-free Survival (PFS), Overall Survival (OS), Number of Participants with One or More Adverse Events (AEs), Number of Participants Who Discontinued from Study Treatment Due to an AE | Merck Sharp & Dohme LLC | Phase 3 | 55 | NCT03783078 (https://ClinicalTrials.gov/show/NCT03783078, Accessed on 22 March 2023) |
2. | Testing Pembrolizumab Versus Observation in Patients with Merkel Cell Carcinoma After Surgery, STAMP Study | Other: Best Practice, Procedure: Biospecimen Collection, Procedure: Computed Tomography, Biological: Pembrolizumab, Procedure: Positron Emission Tomography, Radiation: Radiation Therapy | Recurrence-free survival (RFS), OS, Impact of radiation therapy on RFS, Impact of radiation therapy on OS, Impact of radiation therapy on distant metastasis-free survival (DMFS), Incidence of adverse events | National Cancer Institute (NCI) | Phase 3 | 280 | NCT03712605 (https://ClinicalTrials.gov/show/NCT03712605, Accessed on 22 March 2023) |
3. | Adjuvant Avelumab in Merkel Cell Cancer | Drug: Avelumab, Other: Peripheral Blood Collection, Other: Placebo | Relapse-free survival, Disease-specific survival, Distant-metastases free survival, Incidence of adverse events, Overall survival | University of Washington, EMD Serono | Phase 3 | 100 | NCT03271372 (https://ClinicalTrials.gov/show/NCT03271372, Accessed on 22 March 2023) |
4. | Evaluating Length of Treatment With PD-1/PD-L1 Inhibitor in Advanced Solid Tumors | Drug: Continue PD-1/PD-L1 Inhibitors treatment; other: Discontinue PD-1/PD-L1–1 inhibitor | Time to next treatment, Progression-free Survival (PFS) (at between 2 and 3.9 months), Progression-free Survival (PFS) (at between 4 and 7.9 months), Progression-free Survival (PFS), Incidence of irAEs (Immune-Related Adverse Events), Overall Survival (OS), Best Objective Response (BOR) | Jason J. Luke, MD, University of Pittsburgh | Phase 3 | 578 | NCT04157985 (https://ClinicalTrials.gov/show/NCT04157985, Accessed on 22 March 2023) |
5. | Study Comparing the Standard Administration of IO Versus the Same IO Administered Each 3 Months in Patients with Metastatic Cancer in Response After 6 Months of Standard IO | Drug: Reduced dose intensity of IO | Progression-free survival (PFS), Cost-effectiveness analysis of the proposed therapeutic strategy, Immune progression-free survival (iPFS), Objective response rate (ORR), Overall survival (OS), Duration of response (DoR), Quality of life questionnaire - Core 30 (QLQ-C30), The Developed 5-level version of EQ-5D (EQ-5D-5L) questionnaire, Hospital anxiety and depression scale (HADS), Fear of relapse questionnaire, Safety profile | UNICANCER | Phase 3 | 646 | NCT05078047 (https://ClinicalTrials.gov/show/NCT05078047, Accessed on 22 March 2023) |
(ii) HTLV1 | |||||||
1. | Use of Pentoxifylline in Human T-lymphotropic Virus Type-1 (HTLV-1) Diseases | Drug: Pentoxifylline, Drug: Placebo | Functional neurological capacity, Reduced in cytokines and chemokines | Hospital Universitario Professor Edgard Santos | Phase 3 | 48 | NCT01472263 (https://ClinicalTrials.gov/show/NCT01472263, Accessed on 22 March 2023) |
2. | Zidovudine Plus Lamivudine in HTLV-I-associated Myelopathy: A Randomized Trial | Drug: Zidovudine/lamivudine, Drug: Placebos | Timed walk, Osame’s Motor Disability Score, Pain score, Urinary frequency, HTLV-1 proviral load, CD25%, HLA-DR% | Imperial College London | Phases 2 and 3 | 16 | NCT00272480 (https://ClinicalTrials.gov/show/NCT00272480, Accessed on 22 March 2023) |
3. | Ciclosporin in HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP) | Drug: Ciclosporin | Number of Patients with Lack of Objective Clinical Improvement, Change in Timed Walk Sl. No. Between Baseline and 12 Weeks | Imperial College London, Medical Research Council, University Hospital Birmingham, Imperial College Healthcare NHS Trust | Phases 2 and 3 | 7 | NCT00773292 (https://ClinicalTrials.gov/show/NCT00773292, Accessed on 22 March 2023) |
4. | Use of Valproic Acid to Treat Tropical Spastic Paraparesis/HTLV-1-Associated Myelopathy (TSP/HAM) | Drug: Valproic acid, Drug: Corticosteroids, Drug: valproic acid plus corticosteroids | Neurological scales, Quality of life | University of Sao Paulo | Phase 3 | 60 | NCT00681980 (https://ClinicalTrials.gov/show/NCT00681980, Accessed on 22 March 2023) |
5. | Pilot Study of Combination Therapy With CHOP-Zenapax (CHOP-daclizumab) | Drug: CHOP-daclizumab | King’s College Hospital NHS Trust | Phase 4 | NCT01418430 (https://ClinicalTrials.gov/show/NCT01418430, Accessed on 22 March 2023) | ||
(iii) HPV | |||||||
1. | Impact of AV2 Antiviral Drug on the Treatment of HPV-associated Lesions of the Uterine Cervix | Drug: AV2, Drug: Placebo | Change of lesions, absence of HPV DNA, the correlation between change of lesions and change in HPV DNA, Change in HPV viral particle load | Jean-Pierre Van Geertruyden, University of Kinshasa, University Hospital, Antwerp, Universiteit Antwerpen | Phase 3 | 327 | NCT02346227 (https://ClinicalTrials.gov/show/NCT02346227, Accessed on 22 March 2023) |
2. | Recombinant Human Interferon a-2b Gel for HPV Gynaecological Infections | Drug: Yallaferon | The difference in hr-HPV DNA negative conversion rate, Secondary efficacy end points were the differences of single-type HPV infection, dual infection, and multiple infections in the sixth month between the two groups | Lee’s Pharmaceutical Limited | Phases 2 and 3 | 325 | NCT01824992 (https://ClinicalTrials.gov/show/NCT01824992, Accessed on 22 March 2023) |
3. | Study of the Efficacy and Safety of the Drug Ingaron (Interferon-gamma) in the Treatment of Anogenital Warts | Drug: Interferon-gamma human recombinant (IFN-G) | Recurrence of anogenital warts | SPP Pharmaclon Ltd. | Phase 3 | 30 | NCT05156541 (https://ClinicalTrials.gov/show/NCT05156541, Accessed on 22 March 2023) |
4. | Delivery, Uptake and Acceptability of HPV Vaccination in Tanzanian Girls | Biological: Gardasil HPV vaccine | Vaccine coverage by delivery strategy, Vaccine coverage (dose 2) by delivery strategy, Vaccine coverage (dose 1) by delivery strategy, Factors associated with refusal to vaccinate or to complete vaccination course, Identification of barriers to HPV vaccination, Estimation of the costs of introducing and scaling up HPV vaccines in schools | London School of Hygiene and Tropical Medicine, National Institute for Medical Research, Tanzania, Ocean Road Cancer Institute, Tanzania, Institut Catalan d’ Oncologia, Spain, Medical Research Council Social & Public Health Sciences Unit, UK, International Union Against Cancer, Switzerland | Phase 4 | 5532 | NCT01173900 (https://ClinicalTrials.gov/show/NCT01173900, Accessed on 22 March 2023) |
5. | Immunogenicity and Safety of Dengue Tetravalent Vaccine (TDV) and Recombinant 9-valent Human Papillomavirus Vaccine (9vHPV) in Participants Aged 9 to < 15 Years | Biological: 9vHPV vaccine, Biological: Dengue Tetravalent Vaccine (TDV) | Geometric Mean Titers (GMTs) for Human Papillomavirus (HPV) Types 6, 11, 16, 18, 31, 33, 45, 52, and 58, Percentage of Participants with Seropositivity for HPV Types 6, 11, 16, 18, 31, 33, 45, 52, and 58 as Measured by Immunoglobulin G Binding Assay (IgGBA) or Equivalent Assay, GMTs of Neutralizing Antibodies for Each of the 4 Dengue Serotypes, Percentage of Participants with Seropositivity for Each of the 4 Dengue Serotypes, Percentage of Participants with Seropositivity for Multiple (2, 3, or 4) Dengue Serotypes, Percentage of Participants with Solicited Local Reactions for 7 Days Following Vaccination by Severity, Percentage of Participants with Solicited Systemic Adverse Events (AEs) for 14 days Following Vaccination by Severity, Percentage of Participants with any Unsolicited AEs for 28 days Following Vaccination, Percentage of Participants with Serious Adverse Events (SAEs) | Takeda | Phase 3 | 618 | NCT04313244 (https://ClinicalTrials.gov/show/NCT04313244, Accessed on 22 March 2023) |
6. | Safety and Immunogenicity of Human Papillomavirus (HPV) Vaccine in Solid Organ Transplant Recipients | Biological: Human papillomavirus quadrivalent vaccine | The primary outcome will be a 2-fold rise in the type-specific HPV titer for at least one of the four serotypes contained in the vaccine at month 7, Vaccine adverse events, including episodes of rejection up to 1 year after study enrolment, Immunogenicity at 36 months postvaccination. | University of Alberta | Phase 3 | 50 | NCT00677677 (https://ClinicalTrials.gov/show/NCT00677677, Accessed on 22 March 2023) |
7. | Immunogenicity and Safety of a Quadrivalent Human Papillomavirus (HPV) Vaccine in Patients With SLE: a Controlled Study | Drug: human papillomavirus vaccination (Gardasil) | Antibody titers against 4 strains of human papillomavirus | Tuen Mun Hospital | Phase 4 | 100 | NCT00911521 (https://ClinicalTrials.gov/show/NCT00911521, Accessed on 22 March 2023) |
(iv) KSHV | |||||||
1. | Safety and Effectiveness of an Experimental Drug, IM862, in Treating Kaposi’s Sarcoma in AIDS Patients | Drug: IM862 | Cytran, NIH AIDS Clinical Trials Information Service | Phase 3 | 200 | NCT00002445 (https://ClinicalTrials.gov/show/NCT00002445, Accessed on 22 March 2023) | |
2. | Antiretroviral Therapy (ART) Alone or With Delayed Chemo Versus ART With Immediate Chemo for Limited AIDS-related Kaposi’s Sarcoma | Drug: efavirenz/emtricitabine/tenofovir disoproxil fumarate, Drug: etoposide | Kaposi Sarcoma (KS) Status at Week 48 Compared to Study Entry, KS Progressive Disease at Week 48 Compared to Study Entry | AIDS Clinical Trials Group, National Institute of Allergy, and Infectious Diseases (NIAID) | Phase 3 | 192 | NCT01352117 (https://ClinicalTrials.gov/show/NCT01352117, Accessed on 22 March 2023) |
3. | HIV/AIDS Kaposis Sarcoma: Comparison of Response to HAART vs HAART Plus CXT | Drug: Generic HAART Triomune: d4T, 3TC, NVP, Drug: Generic HAART Triomune: d4T, 3TC, NVP, and chemotherapy ABV | Clinical response of KS, Skin: tumor measurements of 5 indicator skin lesions. Assessment of KS as per AMC RKS 02 (www.amc.uab.edu), photography of indicator lesions with metric tape in the frame, Visceral: chest radiograph and endoscopy, where necessary, bronchoscopy, Safety and toxicity by DAIDS Toxicity criteria, Immunological and virological response to HAART as measured by CD4 and HIV-viral load, QOL by EORTC QLQ C30, Adherence | University of KwaZulu, AIDS Care Research in Africa, National Research Foundation, Singapore, AIDS Malignancy Consortium, Cipla Medpro, Dermatological Society of South Africa | Phase 4 | 112 | NCT00380770 (https://ClinicalTrials.gov/show/NCT00380770, Accessed on 22 March 2023) |
4. | Use of Stealth Liposomal Doxorubicin HCl (DOX-SL) in the Treatment of Moderate to Severe AIDS-Related Kaposi’s Sarcoma. | Drug: Doxorubicin hydrochloride (liposomal) | Sequus Pharmaceuticals, NIH AIDS Clinical Trials Information Service | Phase 3 | NCT00002147 (https://ClinicalTrials.gov/show/NCT00002147, Accessed on 22 March 2023) | ||
5. | A Pilot Study of the Effects of Highly Active Antiretroviral Therapy on Kaposi’s Sarcoma in Zimbabwe | Drug: abacavir/3TC/zidovudine, Drug: abacavir /3TC plus ritonavir boosted lopinavir | Compare effects of twice-daily all-(NRTI) antiretroviral regimen to a once-daily regimen of 2 NRTIs plus a protease inhibitor AIDS-KS subjects with good virologic suppression on all-NRTI regimen. | Parirenyatwa Hospital, University of Colorado, Denver, GlaxoSmithKline, Abbott | Phases 2 and 3 | 49 | NCT00834457 (https://ClinicalTrials.gov/show/NCT00834457, Accessed on 22 March 2023) |
6. | A Study of DOX-SL in the Treatment of AIDS-Related Kaposi’s Sarcoma | Drug: Doxorubicin hydrochloride (liposomal) | Sequus Pharmaceuticals, NIH AIDS Clinical Trials Information Service | Phase 3 | NCT00002319 (https://ClinicalTrials.gov/show/NCT00002319, Accessed on 22 March 2023) | ||
7. | Paclitaxel Compared with Doxorubicin in Treating Patients with Advanced AIDS-Related Kaposi’s Sarcoma | Drug: paclitaxel, Drug: pegylated liposomal doxorubicin hydrochloride, Other: laboratory biomarker analysis, Procedure: quality-of-life assessment | Progression-free survival, Patients’ health-related quality of life (QOL) in terms of change in pain score, e Antibody titers edema-related mobility, gastrointestinal (GI) symptoms and respiratory symptoms based on the total score from the Functional Assessment of HIV Infection (FAHI) v3 | National Cancer Institute (NCI) | Phase 3 | 240 | NCT00003350 (https://ClinicalTrials.gov/show/NCT00003350, Accessed on 22 March 2023) |
8. | Randomized, Comparative Trial of DOX-SL (Stealth Liposomal Doxorubicin Hydrochloride) Versus Bleomycin and Vincristine in the Treatment of AIDS-Related Kaposi’s Sarcoma | Drug: Doxorubicin hydrochloride (liposomal), Drug: Bleomycin sulfate, Drug: Vincristine sulfate | Sequus Pharmaceuticals, NIH AIDS Clinical Trials Information Service | Phase 3 | 220 | NCT00002105 (https://ClinicalTrials.gov/show/NCT00002105, Accessed on 22 March 2023) | |
9. | Doxorubicin in Treating Patients With AIDS-Related Kaposi’s Sarcoma | Drug: daunorubicin hydrochloride, Drug: pegylated liposomal doxorubicin hydrochloride | Roswell Park Cancer Institute | Phase 3 | NCT00002985 (https://ClinicalTrials.gov/show/NCT00000994, Accessed on 22 March 2023) | ||
10. | A Randomized Phase III Clinical Trial of Daunoxome Versus Combination Chemotherapy with Adriamycin/Bleomycin/Vincristine (ABV) in the Treatment of HIV-Associated Kaposi’s Sarcoma. | Drug: Daunorubicin (liposomal), Drug: Bleomycin sulfate, Drug: Vincristine sulfate, Drug: Doxorubicin hydrochloride | Nexstar Pharmaceuticals, NIH AIDS Clinical Trials Information Service | Phase 3 | NCT00002093 (https://ClinicalTrials.gov/show/NCT00000994, Accessed on 22 March 2023) | ||
11. | A Study of AZT in HIV-Infected Patients With AIDS-Related Kaposi’s Sarcoma | Drug: Zidovudine | National Institute of Allergy and Infectious Diseases (NIAID) | Phase 3 | 240 | NCT00000994 (https://ClinicalTrials.gov/show/NCT00000994, Accessed on 22 March 2023) | |
12. | Anti-Retrovirals for Kaposi’s Sarcoma | Drug: Lopinavir/ritonavir plus Emtricitabine/Tenofovir versus Efavirenz plus Emtricitabine/Tenofovir | Blinded assessment of the change in the burden of KS lesions, CD4+ T cell count and HIV plasma HIV RNA levels, KSHV DNA levels in saliva and blood, Humoral and cellular KSHV immune response markers, Quality-of-life assessment, Incidence of Kaposi’s sarcoma-associated Immune Reconstitution Inflammatory Syndrome (KS-IRIS) | University of California, San Francisco, National Institutes of Health (NIH), Gilead Sciences, Abbott, Merck Sharp & Dohme LLC | Phase 4 | 224 | NCT00444379 (https://ClinicalTrials.gov/show/NCT00444379, Accessed on 22 March 2023) |
(v) HCV | |||||||
1. | A Study to Evaluate the Efficacy and Safety of ABT-493/ABT-530 in Adults with Chronic Hepatitis C Virus (HCV) Genotype 2 Infection | Drug: ABT-493/ABT-530, Drug: Placebo for ABT-493/ABT-530 | Percentage of Participants with Sustained Virologic Response 12 Weeks Post-treatment (SVR12) in Arm A DB Active Drug Excluding Prior SOF + Ribavirin (RBV) | AbbVie | Phase 3 | 304 | NCT02640482 (https://ClinicalTrials.gov/show/NCT02640482, Accessed on 22 March 2023) |
2. | Treatment of Acute Hepatitis C Virus Infection with Pegylated Interferon in Injection Drug Users | Drug: Pegylated Interferon | The sustained viral response rate in the treatment group versus control (measured at Week 24), Adherence rate in the treatment group (measured at Week 24) | National Institute on Drug Abuse (NIDA), University of Washington | Phase 4 | 21 | NCT00194480 (https://ClinicalTrials.gov/show/NCT00194480, Accessed on 22 March 2023) |
3. | Drug Use & Infections in Vietnam - Hepatitis C (DRIVE-C) | Drug: Sofosbuvir 400 mg and Daclatasvir 60 mg, Drug: Sofosbuvir 400 mg and Daclatasvir 90 mg, Drug: Ribavirin, Drug: Sofosbuvir and Daclatasvir for 24 weeks | The proportion of all patients with the success of the model of care, the Proportion of patients with detectable HCV RNA | ANRS, Emerging Infectious Diseases | Phase 4 | 979 | NCT03537196 (https://ClinicalTrials.gov/show/NCT03537196, Accessed on 22 March 2023) |
4. | A Study to Evaluate the Efficacy and Safety of Three Experimental Drugs in Adults with Hepatitis C Virus Infection, Who Are Either Treatment-naive or Treatment-experienced in Brazil | Drug: ombitasvir/paritaprevir/ritonavir and dasabuvir, Drug: ribavirin | Percentage of Participants with Sustained Virologic Response 12 Weeks Post-treatment (SVR12), Change from Baseline to 12 Weeks After the Last Dose of Study Drug, (SF-36v2) Mental Component Summary (MCS) Scores: Change from Baseline to 12 Weeks After the Last Dose of Study Drug | AbbVie | Phase 3 | 222 | NCT02442271 (https://ClinicalTrials.gov/show/NCT02442271, Accessed on 22 March 2023) |
5. | Pegylated Interferon Plus Ribavirin in the Treatment of Active and Past Intravenous Drug Users Infected with Hepatitis C | Drug: pegylated interferon alfa-2a (Roche) and ribavirin | Sustained viral response | The University of Calgary, Canadian Institutes of Health Research (CIHR), Roche Pharma AG | Phase 4 | 66 | NCT00203606 (https://ClinicalTrials.gov/show/NCT01773070, Accessed on 22 March 2023) |
6. | A Follow-up Study Designed to Obtain Long-Term Data on Participants Who Either Achieved a Sustained Virologic Response or Did Not Achieve a Sustained Virologic Response in an AbbVie Sponsored Hepatitis C Study | Drug: ABT-450/ritonavir, Drug: ABT-333, Drug: ABT-267 | Percentage of Participants Who Experienced Relapse-12 overall With and Without New HCV Infection | AbbVie (prior sponsor, Abbott), AbbVie | Phase 3 | 478 | NCT01773070 (https://ClinicalTrials.gov/show/NCT01773070, Accessed on 22 March 2023) |
7. | Pilot Treatment as Prevention for HCV Among Persons Who Actively Inject Drugs | Other: modified directly observed therapy (mDOT), Other: unobserved dosing, Other: Motivational Interviewing-based counseling | Number of people who inject drugs (PWIDs) with HCV who were recruited and retained, Medication adherence to study drug, Challenges of medication adherence, SVR (end-of-treatment response), SOF/metabolite levels, HCV relapse and reinfection, Social and injector networks of participants | Phillip Coffin, MD, MIA, National Institute on Drug Abuse (NIDA), San Francisco Department of Public Health | Phase 4 | 31 | NCT02609893 (https://ClinicalTrials.gov/show/NCT02609893, Accessed on 22 March 2023) |
8. | Hepatitis C Treatment in PWIDs: MAT or Syringe Exchange Assisted-therapy vs Standard of Care | Drug: elbasvir-grazoprevir (50 mg/100 mg) | SVR 12, SVR 48, Discontinuation Rate or Lost to Follow Up, NS5A Resistance, Medication Adherence, Injection Drug Use Relapse (IDU) | Oregon Health and Science University | Phase 4 | 100 | NCT03093415 (https://ClinicalTrials.gov/show/NCT03093415, Accessed on 22 March 2023) |
9. | A Trial to Reduce Hepatitis C Among Injection Drug Users - 1 | Behavioral: Behavior Therapy | Hepatitis C seroconversion, Substance use | Butler Hospital, National Institute on Drug Abuse (NIDA) | Phase 3 | 277 | NCT00218192 (https://ClinicalTrials.gov/show/NCT00218192, Accessed on 22 March 2023) |
10. | A Study to Evaluate the Efficacy and Safety of Three Experimental Drugs Compared with Telaprevir (a Licensed Product) in People with Hepatitis C Virus Infection Who Have Not Had Treatment Before | Drug: ABT-450/r/ABT-267, ABT-333, Drug: Ribavirin, Drug: Telaprevir, Drug: Pegylated Interferon-alpha 2-a (PegIFN) | Percentage of Participants with Sustained Virologic Response 12 Weeks After Treatment (SVR12), Percentage of Participants with Sustained Virologic Response 24 Weeks After Treatment (SVR24) | AbbVie | Phase 3 | 311 | NCT01854697 (https://ClinicalTrials.gov/show/NCT01854697, Accessed on 22 March 2023) |
(vi) HBV | |||||||
1. | Effects of Telbivudine and Tenofovir Disoproxil Fumarate Treatment on the Hepatitis B Virus DNA Kinetics in CHB | Drug: Telbivudine, Drug: Tenofovir, Drug: Telbivudine plus tenofovir | Change in Hepatitis B Virus (HBV) Deoxyribonucleic Acid (DNA) Level from Baseline to Week 12 | Phase 3 | 83 | NCT00805675 (https://ClinicalTrials.gov/show/NCT00805675, Accessed on 22 March 2023) | |
2. | Effects of Telbivudine and Tenofovir Disoproxil Fumarate Treatment on the Hepatitis B Virus DNA Kinetics in CHB | Drug: Telbivudine, Drug: Tenofovir, Drug: Telbivudine plus tenofovir | Change in Hepatitis B Virus (HBV) Deoxyribonucleic Acid (DNA) Level from Baseline to Week 12 | Phase 3 | 109 | NCT00395018 (https://ClinicalTrials.gov/show/NCT00395018, Accessed on 22 March 2023) | |
3. | TDF VS LAM + ADV in LAM + ADV Treated LAM-resistant CHB Patients with Undetectable Hepatitis B Virus DNA | Drug: Lamivudine plus adefovir, Drug: Tenofovir | Percentage number of patients with virus reactivation, Virologic response, Antiviral resistance, Biochemical response, Serologic response, Safety assessment | Phase 4 | 171 | NCT01732367 (https://ClinicalTrials.gov/show/NCT01732367, Accessed on 22 March 2023) | |
4. | Evaluation of Tenofovir Disoproxil Fumarate in Adolescents with Chronic Hepatitis B Infection | Drug: Tenofovir disoproxil fumarate (TDF), Drug: Placebo | Percentage of Participants with HBV DNA < 400 copies/mL at Week 72 | Phase 3 | 106 | NCT00734162 (https://ClinicalTrials.gov/show/NCT00734162, Accessed on 22 March 2023) | |
5. | A Study of Maraviroc In HIV Co-Infected Subjects With Hepatitis C and Hepatitis B | Drug: Maraviroc, Drug: Placebo | Percentage of Participants with grade 3 and grade 4 Alanine Aminotransferase (ALT) Abnormalities at Week 48 | Phase 4 | 138 | NCT01327547 (https://ClinicalTrials.gov/show/NCT01327547, Accessed on 22 March 2023) | |
6. | Viral Kinetics Study of Telbivudine and Entecavir in Adults with Chronic Hepatitis B | Drug: Entecavir, Drug: Telbivudine | Change in Mean Hepatitis B Virus (HBV) DNA Levels, Change in Mean HBV DNA Level, The Area Under the Curve (AUC) of HBV DNA Change, Change in Alanine Aminotransferase (ALT) Levels, Characterization of Very Early Viral Kinetics: Estimation of Viral Clearance, Characterization of Very Early Viral Kinetics: Estimation of the Rate of Infected Cell Loss, Characterization of Very Early Viral Kinetics: Estimation of the Efficiency Factor of Blocking Virus Production, Number of Patients Who Are Polymerase Chain Reaction (PCR) Negative | Phase 3 | 44 | NCT00412529 (https://ClinicalTrials.gov/show/NCT03468907, Accessed on 22 March 2023) | |
(vii) EBV | |||||||
1. | Belatacept 3 Month Post Transplant Conversion Study | Drug: belatacept, Drug: Tacrolimus, Drug: MPA | Change in eGFR (MDRD) at 2 Years Post-transplant Compared to Baseline at Month 3 (Conversion), Acute Rejection, Graft Survival, Patient Survival | Lorenzo Gallon, Bristol-Myers Squibb, Northwestern University | Phase 4 | 28 | NCT02213068 (https://ClinicalTrials.gov/show/NCT02213068, Accessed on 22 March 2023) |
2. | A Trial of Adjuvant Chemotherapy in Nasopharyngeal Carcinoma Patients with Residual Epstein–Barr Virus (EBV) DNA Following Radiotherapy | Drug: Adjuvant chemotherapy (gemcitabine and cisplatin) | Relapse-free survival, Overall survival, Loco-regional control, Metastasis-free survival, Toxicity of adjuvant chemotherapy, Correlation of plasma EBV DNA and PET/CT scan with clinical course and outcome | Chinese University of Hong Kong, Hong Kong Nasopharyngeal Cancer Study Group Limited | Phase 3 | 104 | NCT00370890 (https://ClinicalTrials.gov/show/NCT00370890, Accessed on 22 March 2023) |
3. | A Phase III Trial Evaluating Chemotherapy and Immunotherapy for Advanced Nasopharyngeal Carcinoma (NPC) Patients | Biological: autologous EBV-specific Cytotoxic T cells, Drug: a combination of IV gemcitabine and IV carboplatin (AUC2) | Prolonging Overall Survival, Disease Progression, Overall Response Rate, Clinical Benefit Rate, and Quality of Life of patients | Tessa Therapeutics | Phase 3 | 330 | NCT02578641 (https://ClinicalTrials.gov/show/NCT02578641, Accessed on 22 March 2023) |
4. | Efficacy and Safety Study of Lenalidomide Plus R-CHOP Chemotherapy Versus Placebo Plus R-CHOP Chemotherapy in Untreated ABC Type Diffuse Large B-cell Lymphoma | Drug: lenalidomide, Drug: Placebo, Drug: Rituximab, Drug: Cyclophosphamide, Drug: Doxorubicin, Drug: prednisone, Drug: vincristine | Celgene | Phase 3 | 570 | NCT02285062 (https://ClinicalTrials.gov/show/NCT02285062, Accessed on 22 March 2023) | |
5. | Newly Diagnosed Mature B-ALL, Burkitt’s Lymphoma and Other High-grade Lymphoma in Adults | Drug: Adriamycin, Drug: Cyclophosphamide, Drug: Cytarabine, Drug: Dexamethasone/Prednisolone, Drug: VP16, Drug: Ifosfamide, Drug: Methotrexate, Drug: G-CSF, Drug: Rituximab, Drug: Vincristine/Vindesine, Procedure: Irradiation (in specific conditions) | Nicola Goekbuget, Goethe University | Phase 4 | 650 | NCT00199082 (https://ClinicalTrials.gov/show/NCT00199082, Accessed on 22 March 2023) | |
6. | Double Cord Versus Haploidentical (BMT CTN 1101) | Biological: Haploidentical Bone Marrow Transplant, Biological: Double Umbilical Cord Blood Transplant | Medical College of Wisconsin, National Heart, Lung, and Blood Institute (NHLBI), National Cancer Institute (NCI), Blood and Marrow Transplant Clinical Trials Network, National Marrow Donor Program | Phase 3 | 368 | NCT01597778 (https://ClinicalTrials.gov/show/NCT01597778, Accessed on 22 March 2023) | |
7. | Interleukin-2 or Observation Following Radiation Therapy, Combination Chemotherapy, and Peripheral Stem Cell Transplantation in Treating Patients with Recurrent Non-Hodgkin’s Lymphoma | Biological: aldesleukin, Biological: filgrastim, Drug: cyclophosphamide, Drug: etoposide, Radiation: radiation therapy, Procedure: peripheral blood stem cell transplantation, Procedure: bone marrow ablation with stem cell support | Overall survival, Disease-free survival, Frequency, and severity of toxicity associated with post-transplant aldesleukin therapy | National Cancer Institute (NCI) | Phase 3 | 206 | NCT00002649 (https://ClinicalTrials.gov/show/NCT01800071, Accessed on 22 March 2023) |
8. | A Phase Ib Trial of MVA-EBNA1/LMP2 Vaccine in Nasopharyngeal Carcinoma | Drug: MVA-EBNA1/LMP2 vaccine | Immune response to three cycles of MVA-EBNA1/LMP2 vaccine, Occurrence of adverse events defined according to NCI CTCAE version 4.02, Immune memory, and recall response to MVA-EBNA1/LMP2 vaccination, Measurement of EBV genome levels in plasma before, during and after vaccination, Tumor response as determined by Immune-Related Response Criteria (irRC) | Cancer Research UK | Phase 1 | 22 | NCT01800071 (https://ClinicalTrials.gov/show/NCT01800071, Accessed on 22 March 2023) |
(viii) HIV | |||||||
1. | Prospective Evaluation of Etravirine for HIV-infected Patients in Need of Lipid-lowering Drugs | Drug: stop statin and switch to an antiretroviral drug with less impact on lipid metabolism | The proportion of patients not qualifying anymore for statin treatment, fasting lipids changes | Calmy Alexandra, Janssen-Cilag A.G., Switzerland, University Hospital, Geneva | Phase 3 | 34 | NCT01543035 (https://ClinicalTrials.gov/show/NCT01543035, Accessed on 22 March 2023) |
2. | Behavioral Drug and HIV Risk Reduction counselling in Methadone Patients in China | Behavioral: Behavioral Drug and HIV Risk Reduction Counselling (BDRC), Behavioral: Drug counselling | Reductions of illicit opiate use, Reductions in HIV risk behaviors | Yale University, National Institute on Drug Abuse (NIDA) | Phase 3 | 45 | NCT00757744 (https://ClinicalTrials.gov/show/NCT00757744, Accessed on 22 March 2023) |
3. | Positive Change Agents Program-Tanzania (Evaluation) | Behavioral: Appreciative Inquiry Change Agents (CA) program (NAMWEZA) | HIV Testing or other related services for network members of HIV Positive change agents, Depressive symptoms | Harvard School of Public Health (HSPH), Harvard Medical School (HMS and HSDM), Muhimbili University of Health and Allied Sciences, Centres for Disease Control and Prevention | Phase 3 | 1046 | NCT01693458 (https://ClinicalTrials.gov/show/NCT01693458, Accessed on 22 March 2023) |
4. | Safety and Efficacy of Switching from Regimens of ABC/3TC + a third Agent to E/C/F/TAF Fixed-Dose Combination (FDC) in Virologically Suppressed HIV 1 Infected Adults | Drug: E/C/F/TAF, Drug: ABC/3TC, Drug: Third Antiretroviral Agent | Percentage of Participants Who Have HIV1 RNA < 50 Copies/mL as Defined by the FDA Snapshot Algorithm at Week 24, Change from Baseline in CD4+ Cell Count at Week 48 | Gilead Sciences | Phase 3 | 275 | NCT02605954 (https://ClinicalTrials.gov/show/NCT02605954, Accessed on 22 March 2023) |
5. | Using Drug Levels in the Blood to Guide Therapy in HIV Infected Patients Taking a Protease Inhibitor | Procedure: Therapeutic Drug Monitoring (TDM) | Change in log10 plasma HIV1 RNA concentration from Step 2 entry (Week 4) to Week 24 (20 weeks postrandomization), change in log10 plasma HIV1 RNA concentration from study entry to Week 24 (20 weeks post-randomization) | National Institute of Allergy and Infectious Diseases (NIAID) | Phase 3 | 360 | NCT00041769 (https://ClinicalTrials.gov/show/NCT00041769, Accessed on 22 March 2023) |
6. | Maraviroc is an Immunomodulatory Drug for Antiretroviral-treated HIV Infected Patients Exhibiting Immunologic Failure | Drug: Placebo, Drug: Maraviroc | Week 24 Change in Percentage of CD8+ T Cells That Co-express CD38 and HLA DR (Week 24 %CD38+HLA-DR+ CD8+ T Cells Minus Baseline %CD38+HLA-DR+ CD8+ T Cells), Change in CD4+ T Cell Count, Change in Ultrasensitive Plasma HIV RNA Level (Single Copy/mL Assay), Change in Brachial Artery Flow-mediated Dilatation (UCSF Site Only), Change in Gut-associated Lymphoid Tissue HIV RNA Level (UCSF Site Only) | University of California, San Francisco, Pfizer, amfAR, The Foundation for AIDS Research, Stanford University, Case Western Reserve University, Rush University | Phase 4 | 45 | NCT00735072 (https://ClinicalTrials.gov/show/NCT00735072, Accessed on 22 March 2023) |
7. | Anti-HIV Drug Regimens with or Without Protease Inhibitors and Drug Level Monitoring in HIV Infected Adolescents | Drug: Efavirenz + 2 NRTIs, Drug: Lopinavir/Ritonavir + 2 NRTIs, Procedure: Therapeutic Drug Monitoring | The proportion of patients achieving viral suppression (viral load less than 1,000 copies/mL) at Week 24 and maintaining suppression through Week 48 while remaining on study treatment, the proportion of patients achieving virologic suppression (viral load less than 1,000 copies/mL) at Week 24 and maintaining suppression through Week 96 while remaining on study treatment, CD4 (T helper cells), CD8 (cytotoxic T cells), naive CD4 T cells (CD62L/CD45RA/CD4), and activated CD8 T cells (HLA-DR/CD38/CD8), changes from baseline to Weeks 24, 48, and 96 for percentage and total number of CD19 (B cells), total T cells (CD3 T cells), CD4 (T helper cells), CD8 (cytotoxic T cells), naive CD4 T cells (CD62L/CD45RA/CD4), and activated CD8 T cells (HLA-DR/CD38/CD8) | National Institute of Allergy and Infectious Diseases (NIAID), Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) | Phase 3 | 240 | NCT00075907 (https://ClinicalTrials.gov/show/NCT00075907, Accessed on 22 March 2023) |
8. | Drug Interaction Study Between Lumefantrine and Lopinavir/Ritonavir | Drug: Lumefantrine - lopinavir/ritonavir drug interaction, Drug: Lumefantrine only arm | Makerere University, University of Liverpool | Phase 4 | 32 | NCT00619944 (https://ClinicalTrials.gov/show/NCT00619944, Accessed on 22 March 2023) | |
9. | Antidepressant Medication for Reducing HIV Risk Behavior in Depressed Intravenous Drug Users | Drug: Antidepressant Medication | Maintenance of HIV risk-free drug behavior (measured at Month 12), Reduction in depressive symptoms (measured at Month 12) | Butler Hospital, National Institute of Mental Health (NIMH) | Phase 3 | 265 | NCT00228007 (https://ClinicalTrials.gov/show/NCT00228007, Accessed on 22 March 2023) |
10. | Drug Interaction Study of Famotidine and Atazanavir with Ritonavir in HIV-Infected Patients | Drug: Atazanavir/Ritonavir, Drug: Atazanavir/Ritonavir + Famotidine, Drug: Atazanavir/Ritonavir + Tenofovir Disoproxil Fumarate + Famotidine | Bristol-Myers Squibb | Phase 4 | 40 | NCT00384904 (https://ClinicalTrials.gov/show/NCT00384904, Accessed on 22 March 2023) |
“Completed” status here means the study has ended and participants are no longer being examined or treated; that is, the last participant’s last visit has occurred.
5. Current Vaccines for Viral Oncogenesis Therapeutics
5.2.1. Mechanism of Cancer Vaccines
5.2.3. Current Vaccines and Their Effectiveness
S. No. | Viruses | Vaccines | References |
---|---|---|---|
1. | Hepatitis B virus (HBV) | Engerix-B and Recombivax HBHEPLISAV-B | (368) |
2. | Human Papilloma Virus (HPV) | Gardasil (a bivalent HPV vaccine), Cervarix | (368) |
3. | Kaposi sarcoma herpesvirus (KSHV/HHV-8) | Epitopes-based vaccines mRNA-based vaccines (Potential candidates) | (369,370) |
4. | Epstein–Barr virus (EBV/HHV-4) | EBV gp350-Ferritin nanoparticle vaccine (Under trials) | (371) |
5. | Merkel cell polyomavirus (MCPyV) | A VP1-target vaccine formulated with CRA (Potential candidate), Interleukin-12 (IL-12) Plasmid Vaccines | (372,373,374) |
6. | Hepatitis C virus (HCV) | New Hepatitis C Prophylactic Vaccine (Under trials) | (375) |
7. | Human T-cell leukemia/ Lymphotropic virus-1 (HTLV-1) | TAX protein-based epitopes vaccine peptide-based vaccine (HBZ peptide) (Potential candidates) | (376,377) |
8. | Human immunodeficiency virus (HIV) | BG505 MD39.3, BG505 MD39.3 gp151, and BG505 MD39.3 gp151 CD4KO HIV Trimer mRNA Vaccines | (378) |
6. Challenges Associated with Antivirals during Viral Oncogenesis
7. Future Perspectives and Concluding Remarks
Data Availability
Not Applicable.
Acknowledgments
Dr. Piyush Kumar Gupta is thankful to the Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, India, for providing the infrastructure and research facilities.
References
This article references 409 other publications.
- 1Anand, P.; Kunnumakara, A. B.; Sundaram, C.; Harikumar, K. B.; Tharakan, S. T.; Lai, O. S.; Sung, B.; Aggarwal, B. B. Cancer Is a Preventable Disease That Requires Major Lifestyle Changes. Pharm. Res. 2008, 25 (9), 2097– 2116, DOI: 10.1007/s11095-008-9661-9Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXps1ajtLw%253D&md5=9941527b4aebc3cbdc786b1f907d4b3cCancer is a Preventable Disease that Requires Major Lifestyle ChangesAnand, Preetha; Kunnumakkara, Ajaikumar B.; Sundaram, Chitra; Harikumar, Kuzhuvelil B.; Tharakan, Sheeja T.; Lai, Oiki S.; Sung, Bokyung; Aggarwal, Bharat B.Pharmaceutical Research (2008), 25 (9), 2097-2116CODEN: PHREEB; ISSN:0724-8741. (Springer)This year, more than 1 million Americans and more than 10 million people worldwide are expected to be diagnosed with cancer, a disease commonly believed to be preventable. Only 5-10% of all cancer cases can be attributed to genetic defects, whereas the remaining 90-95% have their roots in the environment and lifestyle. The lifestyle factors include cigarette smoking, diet (fried foods, red meat), alc., sun exposure, environmental pollutants, infections, stress, obesity, and phys. inactivity. The evidence indicates that of all cancer-related deaths, almost 25-30% are due to tobacco, as many as 30-35% are linked to diet, about 15-20% are due to infections, and the remaining percentage are due to other factors like radiation, stress, phys. activity, environmental pollutants etc. Therefore, cancer prevention requires smoking cessation, increased ingestion of fruits and vegetables, moderate use of alc., caloric restriction, exercise, avoidance of direct exposure to sunlight, minimal meat consumption, use of whole grains, use of vaccinations, and regular check-ups. In this review, we present evidence that inflammation is the link between the agents/factors that cause cancer and the agents that prevent it. In addn., we provide evidence that cancer is a preventable disease that requires major lifestyle changes.
- 2Mesri, E. A.; Feitelson, M. A.; Munger, K. Human Viral Oncogenesis: A Cancer Hallmarks Analysis. Cell Host Microbe 2014, 15 (3), 266– 282, DOI: 10.1016/j.chom.2014.02.011Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXks1Ons7o%253D&md5=554b50565ad47438a3add3683cb3457bHuman Viral Oncogenesis: A Cancer Hallmarks AnalysisMesri, Enrique A.; Feitelson, Mark A.; Munger, KarlCell Host & Microbe (2014), 15 (3), 266-282CODEN: CHMECB; ISSN:1931-3128. (Elsevier Inc.)A review. Approx. 12% of all human cancers are caused by oncoviruses. Human viral oncogenesis is complex, and only a small percentage of the infected individuals develop cancer, often many years to decades after the initial infection. This reflects the multistep nature of viral oncogenesis, host genetic variability, and the fact that viruses contribute to only a portion of the oncogenic events. In this review, the Hallmarks of Cancer framework of Hanahan and Weinberg (2000 and 2011) is used to dissect the viral, host, and environmental cofactors that contribute to the biol. of multistep oncogenesis mediated by established human oncoviruses. The viruses discussed include Epstein-Barr virus (EBV), high-risk human papillomaviruses (HPVs), hepatitis B and C viruses (HBV and HCV, resp.), human T cell lymphotropic virus-1 (HTLV-1), and Kaposi's sarcoma herpesvirus (KSHV).
- 3Mui, U. N.; Haley, C. T.; Tyring, S. K. Viral Oncology: Molecular Biology and Pathogenesis. J. Clin. Med. 2017, Vol. 6, Page 111 2017, 6 (12), 111, DOI: 10.3390/jcm6120111Google ScholarThere is no corresponding record for this reference.
- 4Bouvard, V.; Baan, R.; Straif, K.; Grosse, Y.; Secretan, B.; El Ghissassi, F.; Benbrahim-Tallaa, L.; Guha, N.; Freeman, C.; Galichet, L.; Cogliano, V. A Review of Human Carcinogens-Part B: Biological Agents. Lancet Oncol. 2009, 10 (4), 321– 322, DOI: 10.1016/S1470-2045(09)70096-8Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M3mt1KmtA%253D%253D&md5=d57408549b4ea6f939b52f3ce412a4efA review of human carcinogens--Part B: biological agentsBouvard Veronique; Baan Robert; Straif Kurt; Grosse Yann; Secretan Beatrice; El Ghissassi Fatiha; Benbrahim-Tallaa Lamia; Guha Neela; Freeman Crystal; Galichet Laurent; Cogliano VincentThe Lancet. Oncology (2009), 10 (4), 321-2 ISSN:.There is no expanded citation for this reference.
- 5De Martel, C.; Ferlay, J.; Franceschi, S.; Vignat, J.; Bray, F.; Forman, D.; Plummer, M. Chronic Disease Management in Ageing Populations. Lancet 2012, 379 (9829), 1851, DOI: 10.1016/S1470-2045(12)70137-7Google ScholarThere is no corresponding record for this reference.
- 6McFadden, K.; Luftig, M. A. Interplay between DNA Tumor Viruses and the Host DNA Damage Response. Curr. Top. Microbiol. Immunol. 2013, 371, 229– 257, DOI: 10.1007/978-3-642-37765-5_9Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjsl2qt70%253D&md5=5263ffc1337487f3e0716c088c113730Interplay Between DNA Tumor Viruses and the Host DNA Damage ResponseMcFadden, Karyn; Luftig, Micah A.Current Topics in Microbiology and Immunology (2013), 371 (Intrinsic Immunity), 229-257CODEN: CTMIA3; ISSN:2196-9965. (Springer GmbH)Viruses encounter many challenges within host cells in order to replicate their nucleic acid. In the case of DNA viruses, one challenge that must be overcome is recognition of viral DNA structures by the host DNA damage response (DDR) machinery. This is accomplished in elegant and unique ways by different viruses as each has specific needs and sensitivities dependent on its life cycle. In this review, we focus on three DNA tumor viruses and their interactions with the DDR. The viruses Epstein-Barr virus (EBV), Kaposi's sarcoma-assocd. herpesvirus (KSHV), and human papillomavirus (HPV) account for nearly all of the virus-assocd. human cancers worldwide. These viruses have also been excellent models for the study of oncogenic virus-mediated cell transformation. In this review, we will discuss how each of these viruses engage and subvert aspects of the host DDR. The first level of DDR engagement is a result of the genetic linkage between the oncogenic potential of these viruses and their ability to replicate. Namely, the promotion of cells from quiescence into the cell cycle to facilitate virus replication can be sensed through aberrant cellular DNA replication structures which activate the DDR and hinder cell transformation. DNA tumor viruses subvert this growth-suppressive DDR through changes in viral oncoprotein expression which ultimately facilitate virus replication. An addnl. level of DDR engagement is through direct detection of replicating viral DNA. These interactions parallel those obsd. in other DNA virus systems in that the need to subvert these intrinsic sensors of aberrant DNA structure in order to replicate must be in place. DNA tumor viruses are no exception. This review will cover the mol. features of DNA tumor virus interactions with the host DDR and the consequences for virus replication.
- 7Tornesello, M. L.; Annunziata, C.; Tornesello, A. L.; Buonaguro, L.; Buonaguro, F. M. Human Oncoviruses and P53 Tumor Suppressor Pathway Deregulation at the Origin of Human Cancers. Cancers 2018, Vol. 10, Page 213 2018, 10 (7), 213, DOI: 10.3390/cancers10070213Google ScholarThere is no corresponding record for this reference.
- 8Krump, N. A.; You, J. Molecular Mechanisms of Viral Oncogenesis in Humans. Nat. Rev. Microbiol. 2018 1611 2018, 16 (11), 684– 698, DOI: 10.1038/s41579-018-0064-6Google ScholarThere is no corresponding record for this reference.
- 9Lambert, P. F.; Sugden, B. Cancer Issue: Viruses and Human Cancer. Yale J. Biol. Med. 2006, 79 (3–4), 115, DOI: 10.1016/B978-0-323-47674-4.00012-8Google ScholarThere is no corresponding record for this reference.
- 10Ezirganli, S.; Kazancioglu, H. O.; Acar, A. H.; Ozdemir, H.; Kuzu, E.; Inan, D. S. Effects of Ankaferd Bloodstopper on Bone Healing in an Ovariectomized Osteoporotic Rat Model. Exp. Ther. Med. 2017, 13 (5), 1827– 1831, DOI: 10.3892/etm.2017.4166Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXpt1Oltr4%253D&md5=b9d6514e08379ab716515c7c385a89fbEffects of Ankaferd BloodStopper on bone healing in an ovariectomized osteoporotic rat modelEzirganli, Seref; Kazancioglu, Hakki Oguz; Acar, Ahmet Huseyin; Ozdemir, Hakan; Kuzu, Emre; Inan, Deniz SahinExperimental and Therapeutic Medicine (2017), 13 (5), 1827-1831CODEN: ETMXA2; ISSN:1792-1015. (Spandidos Publications Ltd.)Bone defects resulting from oncol. surgical resections, congenital facial anomalies, trauma or infection represent a significant and common clin. problem. The present study aimed to evaluate the effects of a com.-available medicinal plant ext. product, Ankaferd BloodStopper (ABS), on bone healing. The present study was performed on 24 female ovariectomized (OVX) rats. A defect on each rat calvarium was created using a trephine burr prior to the rats being divided into two groups. Defects were grafted with a gelatin sponge soaked with normal saline (control group) or plant ext. (exptl. group). Half of the animals were sacrificed after 2 wk and the others after 4 wk. In the control group, the defects were not filled with regenerated bone. By contrast, in the exptl. group, all defect areas had an increased amt. of regenerated bone and connective tissue. Osteoblastic activity appeared to be greater in the exptl. group however, osteoclastic activity was obsd. to be higher in the control group. At 2 and 4 wk, there was a significant difference in the amt. of newly regenerated bone obsd. in the exptl. group compared with the control group (P<0.05). Therefore, the results of the present study indicated that local ABS application had a pos. effect on bone healing in the OVX rat model.
- 11Papastergiou, V.; Lombardi, R.; MacDonald, D.; Tsochatzis, E. A. Global Epidemiology of Hepatitis B Virus (HBV) Infection. Curr. Hepat. Rep. 2015, 14 (3), 171– 178, DOI: 10.1007/s11901-015-0269-3Google ScholarThere is no corresponding record for this reference.
- 12Jiang, B.; Hildt, E. Intracellular Trafficking of HBV Particles. Cells 2020, Vol. 9, Page 2023 2020, 9 (9), 2023, DOI: 10.3390/cells9092023Google ScholarThere is no corresponding record for this reference.
- 13Liang, T. J. Hepatitis B: The Virus and Disease. Hepatology 2009, 49 (S5), S13– S21, DOI: 10.1002/hep.22881Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlvFKqsL4%253D&md5=b7851b7a1dadca4ce42dd2a1e05367aeHepatitis B: the virus and diseaseLiang, T. JakeHepatology (Hoboken, NJ, United States) (2009), 49 (5, Suppl.), S13-S21CODEN: HPTLD9; ISSN:0270-9139. (John Wiley & Sons, Inc.)A review. Hepatitis B virus (HBV) infects more than 300 million people worldwide and is a common cause of liver disease and liver cancer. HBV, a member of the Hepadnaviridae family, is a small DNA virus with unusual features similar to retroviruses. HBV replicates through an RNA intermediate and can integrate into the host genome. The unique features of the HBV replication cycle confer a distinct ability of the virus to persist in infected cells. Virol. and serol. assays have been developed for diagnosis of various forms of HBV-assocd. disease and for treatment of chronic hepatitis B infection. HBV infection leads to a wide spectrum of liver disease ranging from acute (including fulminant hepatic failure) to chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Acute HBV infection can be either asymptomatic or present with symptomatic acute hepatitis. Most adults infected with the virus recover, but 5%-10% are unable to clear the virus and become chronically infected. Many chronically infected persons have mild liver disease with little or no long-term morbidity or mortality. Other individuals with chronic HBV infection develop active disease, which can progress to cirrhosis and liver cancer. These patients require careful monitoring and warrant therapeutic intervention. Extrahepatic manifestations of HBV infection are rare but can be difficult to diagnose and manage. The challenges in the area of HBV-assocd. disease are the lack of knowledge in predicting outcome and progression of HBV infection and an unmet need to understand the mol., cellular, immunol., and genetic basis of various disease manifestations assocd. with HBV infection.
- 14Jiang, B.; Himmelsbach, K.; Ren, H.; Boller, K.; Hildt, E. Subviral Hepatitis B Virus Filaments, like Infectious Viral Particles, Are Released via Multivesicular Bodies. J. Virol. 2016, 90 (7), 3330– 3341, DOI: 10.1128/JVI.03109-15Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVygsLfF&md5=37bc9ef19a39871c378ada7ff0f4a3efSubviral hepatitis B virus filaments, like infectious viral particles, are released via multivesicular bodiesJiang, Bingfu; Himmelsbach, Kiyoshi; Ren, Huimei; Boller, Klaus; Hildt, EberhardJournal of Virology (2016), 90 (7), 3330-3341CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)In addn. to infectious viral particles, hepatitis B virus-replicating cells secrete large amts. of subviral particles assembled by the surface proteins, but lacking any capsid and genome. Subviral particles form spheres (22-nm particles) and filaments. Filaments contain a much larger amt. of the large surface protein (LHBs) compared to spheres. Spheres are released via the constitutive secretory pathway, while viral particles are ESCRT-dependently released via multivesicular bodies (MVBs). The interaction of virions with the ESCRT machinery is mediated by α-taxilin that connects the viral surface protein LHBs with the ESCRT component tsg101. Since filaments in contrast to spheres contain a significant amt. of LHBs, it is unclear whether filaments are released like spheres or like virions. To study the release of subviral particles in the absence of virion formation, a core-deficient HBV mutant was generated. Confocal microscopy, immune electron microscopy of ultrathin sections and isolation of MVBs revealed that filaments enter MVBs. Inhibition of MVB biogenesis by the small-mol. inhibitor U18666A or inhibition of ESCRT functionality by coexpression of transdominant neg. mutants (Vps4A, Vps4B, and CHMP3) abolishes the release of filaments while the secretion of spheres is not affected. These data indicate that in contrast to spheres which are secreted via the secretory pathway, filaments are released via ESCRT/MVB pathway like infectious viral particles.
- 15Cai, D.; Yan, R.; Xu, J. Z.; Zhang, H.; Shen, S.; Mitra, B.; Marchetti, A.; Kim, E. S.; Guo, H. Characterization of the Termini of Cytoplasmic Hepatitis B Virus Deproteinated Relaxed Circular DNA. J. Virol. 2020, 95 (1), e00922, DOI: 10.1128/JVI.00922-20Google ScholarThere is no corresponding record for this reference.
- 16Nassal, M. Hepatitis B Viruses: Reverse Transcription a Different Way. Virus Res. 2008, 134 (1–2), 235– 249, DOI: 10.1016/j.virusres.2007.12.024Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXlslWgu74%253D&md5=3c391d43f7ad0c406b412e8176df48d3Hepatitis B viruses: Reverse transcription a different wayNassal, MichaelVirus Research (2008), 134 (1-2), 235-249CODEN: VIREDF; ISSN:0168-1702. (Elsevier B.V.)A review. Hepatitis B virus (HBV), the causative agent of B-type hepatitis in humans, is the type member of the Hepadnaviridae, hepatotropic DNA viruses that replicate via reverse transcription. Beyond long-established differences to retroviruses in gene expression and overall replication strategy newer work has uncovered addnl. distinctions in the mechanism of reverse transcription per se. These include protein-priming by the unique extra terminal protein domain of the reverse transcriptase (RT) utilizing an RNA hairpin for de novo initiation of first strand DNA synthesis, and the strict dependence of this process on cellular chaperones. Recent in vitro reconstitution systems enabled first biochem. insights into this multifactorial reaction, complemented by high resoln. structural information on the RNA, though not yet the protein, level. Genetic approaches have revealed long-distance interactions in the nucleic acid templates as an important factor enabling the puzzling template switches required to produce the relaxed circular (RC) DNA found in infectious virions. Finally, the failure of even potent HBV RT inhibitors to eliminate nuclear covalently closed circular (ccc) DNA, the functional equiv. of integrated proviral DNA, has spurred a renewed interest in the mechanism of cccDNA generation. These new developments are in the focus of this review.
- 17Ji, J.; Xu, X.; Wu, Q.; Wang, X.; Li, W.; Yao, L.; Kan, Y.; Yuan, L.; Bi, Y.; Xie, Q. Simple and Visible Detection of Duck Hepatitis B Virus in Ducks and Geese Using Loop-Mediated Isothermal Amplification. Poult. Sci. 2020, 99 (2), 791– 796, DOI: 10.1016/j.psj.2019.12.024Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisFegs7bL&md5=eab334597a01c3e6d3bae1ddb5aa67c7Simple and visible detection of duck hepatitis B virus in ducks and geese using loop-mediated isothermal amplificationJi, Jun; Xu, Xin; Wu, Qianqian; Wang, Xueyu; Li, Wanyu; Yao, Lunguang; Kan, Yunchao; Yuan, Lu; Bi, Yingzuo; Xie, QingmeiPoultry Science (2020), 99 (2), 791-796CODEN: POSCAL; ISSN:1525-3171. (Elsevier Inc.)In this study, loop-mediated isothermal amplification (LAMP) was used to establish a rapid, specific, and visual detection method for duck hepatitis B virus (DHBV). The design and synthesis of 4 specific LAMP primers were based on the conserved gene region of the DHBV genome, and the optimum temp. and time of the LAMP reaction were 63°C and 50 min, resp. The LAMP assay was confirmed to be specific for DHBV detection and had the same sensitivity as the quant. PCR assay. A visual detection method for rapid detn. of results was developed using a color indicator contg. phenol red and cresol red. A color change was produced based on a pH change in the reaction system, indicating a pos. reaction. For the detection of samples from ducks and geese, the LAMP method has the advantages of simplicity, high sensitivity and specificity, good visibility, and low cost. Moreover, it is more practical and convenient than PCR-related assays for the clin. detection of DHBV.
- 18He, X.; Xu, H.; Wang, X.; Wu, J.; Niu, J.; Gao, P. Associations between the Single Nucleotide Polymorphisms of APOBEC3A, APOBEC3B and APOBEC3H, and Chronic Hepatitis B Progression and Hepatocellular Carcinoma in a Chinese Population. Mol. Med. Rep. 2019, 20 (3), 2177– 2188, DOI: 10.3892/mmr.2019.10455Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitVejtLjI&md5=58590b5de611ef4aae56ae30d091f815Associations between the single nucleotide polymorphisms of APOBEC3A, APOBEC3B and APOBEC3H, and chronic hepatitis B progression and hepatocellular carcinoma in a Chinese populationHe, Xiuting; Xu, Hongqin; Wang, Xiaomei; Wu, Jing; Niu, Junqi; Gao, PujunMolecular Medicine Reports (2019), 20 (3), 2177-2188CODEN: MMROA5; ISSN:1791-3004. (Spandidos Publications Ltd.)The present study examd. the relationships between the single nucleotide polymorphisms (SNPs) of three members of the apolipoprotein B mRNA-editing catalytic polypeptide-like 3 (A3) gene family, A3A, A3B and A3H, and hepatitis B virus (HBV) infection and hepatocellular carcinoma (HCC) in a Han Chinese population. A total of 654 patients were enrolled in the study between Jan. 2012 and July 2016, including 104 patients with chronic HBV infection (CHB), 265 patients with HBV-related liver cirrhosis and 285 patients with HBV-related HCC. A total of two A3A SNPs (rs7286317 and rs7290153), three A3B SNPs (rs2267398, rs2267401 and rs2076109), and five A3H SNPs (rs56695217, rs139302, rs139297, rs139316 and rs139292) were genotyped using a MassArray system. Statistical anal. and haplotype estn. were conducted using Haploview and Unphased software. No significant assocns. were obsd. between the A3A, A3B and A3H SNPs and the development of CHB and HCC. Haplotype anal. revealed that the mutant haplotypes C-T-A, C-T-G, T-G-G and T-T-G from the A3B SNPs rs2267398-rs2267401-rs2076109 carried a lower risk of HCC than the ref. haplotype. These findings suggested that there was no relationship between A3A, A3B and A3H SNPs and CHB progression or HCC development in the Han Chinese population.
- 19Kanda, T.; Goto, T.; Hirotsu, Y.; Moriyama, M.; Omata, M. Molecular Mechanisms Driving Progression of Liver Cirrhosis towards Hepatocellular Carcinoma in Chronic Hepatitis B and C Infections: A Review. Int. J. Mol. Sci. 2019, Vol. 20, Page 1358 2019, 20 (6), 1358, DOI: 10.3390/ijms20061358Google ScholarThere is no corresponding record for this reference.
- 20Slagle, B. L.; Bouchard, M. J. Role of HBx in Hepatitis B Virus Persistence and Its Therapeutic Implications. Curr. Opin. Virol. 2018, 30, 32– 38, DOI: 10.1016/j.coviro.2018.01.007Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXis1Ogu7Y%253D&md5=2fe685174a228e300cccfa766f66cd1aRole of HBx in hepatitis B virus persistence and its therapeutic implicationsSlagle, Betty L.; Bouchard, Michael J.Current Opinion in Virology (2018), 30 (), 32-38CODEN: COVUAF; ISSN:1879-6257. (Elsevier B.V.)A review. Chronic hepatitis B virus infection is a significant risk factor for cirrhosis and hepatocellular carcinoma. The HBx protein is required for virus replication, but the lack of robust infection models has hindered our understanding of HBx functions that could be targeted for antiviral purposes. We briefly review three properties of HBx: its binding to DDB1 and its regulation of cell survival and metab., to illustrate how a single viral protein can have multiple effects in a cell. We propose that different functions of HBx are needed, depending on the changing hepatocyte environment encountered during a chronic virus infection, and that these functions might serve as novel therapeutic targets for inhibiting hepatitis B virus replication and the development of assocd. diseases.
- 21Kattan, S. W.; Nafie, M. S.; Elmgeed, G. A.; Alelwani, W.; Badar, M.; Tantawy, M. A. Molecular Docking, Anti-Proliferative Activity and Induction of Apoptosis in Human Liver Cancer Cells Treated with Androstane Derivatives: Implication of PI3K/AKT/MTOR Pathway. J. Steroid Biochem. Mol. Biol. 2020, 198, 105604, DOI: 10.1016/j.jsbmb.2020.105604Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisV2ksbw%253D&md5=b6c2b349a55e0d48a14a5c8c22849856Molecular docking, anti-proliferative activity and induction of apoptosis in human liver cancer cells treated with androstane derivatives: Implication of PI3K/AKT/mTOR pathwayKattan, Shahad W.; Nafie, Mohamed S.; Elmgeed, Gamal A.; Alelwani, Walla; Badar, Muhammad; Tantawy, Mohamed A.Journal of Steroid Biochemistry and Molecular Biology (2020), 198 (), 105604CODEN: JSBBEZ; ISSN:0960-0760. (Elsevier Ltd.)Worldwide, cancer is still an area with high unmet medical need. Lead optimization efforts towards structure-based drug design were employed to discover newly synthesized hetero-steroid derivs. with promising anticancer effects against hepatocellular carcinoma (HCC). The aim of our study is to evaluate the anti-proliferative activity and the mechanism, a dual PI3K/mTOR inhibitor, and mechanism of action of a series of heterocylic androstane derivs. as anti-HCC agent. The cytotoxic effects of different heterocylic androstanes and 5FU as single agents, were assessed against both HepG2 cells and Non-malignant MDCK cell line to assess the toxicity. Then the underlying mechanism of compd. 4 as most promising compd. was evaluated using mol. docking, MTT assay, cell cycle anal., DNA fragmentation, and real-time PCR. The results of MTT assay showed potential cytotoxic effect for compd. 4 and 5 against liver cancer cell line with IC50 value 39.81 and 57.54 μM, resp. Inhibition of the PI3K/AKT/mTOR pathway was achieved by compd. 4, which was documented by mol. docking and augmented by gene expression anal. Detailed mechanism revealed that compd. 4 induced cell cycle arrest, DNA fragmentation, and induction of apoptosis by inhibition of anti-apoptotic genes, and upregulation of apoptotic genes. Our results shed a light on aminopyrazoloandrostane deriv. 4 as an inhibitor of the PI3K/AKT/mTOR pathway, which might be acting as promising anti-liver cancer agent. Our data support further investigation of agents targeting the PI3K/AKT/mTOR.
- 22Zhou, S.; Li, Y.; Gao, J.; Wang, Y.; Ma, X.; Ding, H.; Li, X.; Sun, S. Novel Protein Kinase C Phosphorylated Kinase Inhibitor-Matrine Suppresses Replication of Hepatitis B Virus via Modulating the Mitogen-Activated Protein Kinase Signal. https://doi.org/10.1080/21655979.2021.2024957 2022, 13 (2), 2851– 2865, DOI: 10.1080/21655979.2021.2024957Google ScholarThere is no corresponding record for this reference.
- 23Bender, D.; Hildt, E. Effect of Hepatitis Viruses on the Nrf2/Keap1-Signaling Pathway and Its Impact on Viral Replication and Pathogenesis. Int. J. Mol. Sci. 2019, Vol. 20, Page 4659 2019, 20 (18), 4659, DOI: 10.3390/ijms20184659Google ScholarThere is no corresponding record for this reference.
- 24Parker, P. J.; Brown, S. J.; Calleja, V.; Chakravarty, P.; Cobbaut, M.; Linch, M.; Marshall, J. J. T.; Martini, S.; McDonald, N. Q.; Soliman, T.; Watson, L. Equivocal, Explicit and Emergent Actions of PKC Isoforms in Cancer. Nat. Rev. Cancer 2020 211 2021, 21 (1), 51– 63, DOI: 10.1038/s41568-020-00310-4Google ScholarThere is no corresponding record for this reference.
- 25Yang, S.; Liu, Y.; Feng, X.; Wang, X.; Wu, M.; Gong, L.; Shu, B.; lu, Q.; Dong, J. HBx Acts as an Oncogene and Promotes the Invasion and Metastasis of Hepatocellular Carcinoma Both in Vivo and Vitro. Dig. Liver Dis. 2021, 53 (3), 360– 366, DOI: 10.1016/j.dld.2020.10.007Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitFCltLvM&md5=3f5c29e50843b6c4cbc032fcabcbee4bHBx acts as an oncogene and promotes the invasion and metastasis of hepatocellular carcinoma both in vivo and vitroYang, Shizhong; Liu, Yanfeng; Feng, Xiaobin; Wang, Xiaojuan; Wu, Meilong; Gong, Lei; Shu, Bin; Qian, lu; Dong, JiahongDigestive and Liver Disease (2021), 53 (3), 360-366CODEN: DLDIFK; ISSN:1590-8658. (Elsevier B.V.)Hepatitis B virus X (HBx) has been reported to be closely related to hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). This study aimed to detect the expression pattern of HBx and explore whether HBx protein can promote HCC invasion and metastasis both in vivo and vitro.HBx expression was detected in HCC tissues via immunochem. A recombinant adenovirus vector contg. the HBx gene was constructed and transfected into the HCC cell line SMMC-7721. Wound healing, transwell migration, and invasion assays were performed to evaluate migration and invasion potentials. A splenic implant tumor nude mice model was established to confirm its invasion and metastatic abilities in vivo.The pos. rate of HBx in HCC tissues was 67.89%. HBx overexpression significantly promoted the migration and invasion abilities of SMMC-7721 cells in vitro. The tumor model showed that splenic implant tumor vol. and no. of liver metastatic tumor nodes were significantly larger and higher in the HBx overexpression group than in the control group.HBx is highly expressed in HCC tissues and promotes HCC invasion and metastasis both in vivo and vitro with oncogene activity, thereby suggesting that HBx can serve as a novel therapeutic target in HCC.
- 26Zhu, H.; Wang, Y.; Chen, J.; Cheng, G.; Xue, J. Transgenic Mice Expressing Hepatitis B Virus X Protein Are More Susceptible to Carcinogen Induced Hepatocarcinogenesis. Exp. Mol. Pathol. 2004, 76 (1), 44– 50, DOI: 10.1016/j.yexmp.2003.09.001Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXltVCqsQ%253D%253D&md5=c8b9a129eb472f34e610d3d43cb454c6Transgenic mice expressing hepatitis B virus X protein are more susceptible to carcinogen induced hepatocarcinogenesisZhu, Huanzhang; Wang, Yun; Chen, Jianquan; Cheng, Guoxiang; Xue, JinglunExperimental and Molecular Pathology (2004), 76 (1), 44-50CODEN: EXMPA6; ISSN:0014-4800. (Elsevier Science)The hepatitis B virus X (HBx) protein is thought to be implicated in the development of human hepatocellular carcinoma (HCC), but its exact function remains controversial. To investigate whether the expression of the HBx gene alone can induce HCC on an inbred C57BL/6 strain that displays a lower spontaneous rate of liver cancer, and to det. if HBx transgenic mice are more susceptible to the effects of hepatocarcinogens, C57-TgN (HBx) X transgenic mice were bred with normal C57BL/6 mice strain. The F1 mice (about 50% HBx pos. and 50% HBx neg.) were treated with a single dose of diethylnitrosamine (DEN) at 7 days of age, or were untreated. Mice were killed at appropriate time points and were analyzed for histol. change in the liver. The expression of HBx protein were examd. by using immunohistochem. staining. Glycogen storage foci were examd. by using periodic acid-Schiff (PAS) staining. In HBx transgenic mice untreated with DEN, HBx expression and glycogen storage foci were always obsd. in the liver after 8 wk, but not obvious histol. pathol. changes. Histol. examn. of liver tissue confirmed that DEN-treated HBx mice developed approx. twice as many focal lesions of basophilic hepatocytes as treated wild-type littermates. Hepatocellular adenomas and carcinomas were also more frequent in DEN-treated HBx-pos. than HBx-neg. mice. Taken together, our results suggest that HBx gene expression alone is not sufficient for carcinogenesis, but may act as a promoter for malignant transformation.
- 27Bouchard, M. J.; Wang, L. H.; Schneider, R. J. Calcium Signaling by HBx Protein in Hepatitis B Virus DNA Replication. Science (80-.). 2001, 294 (5550), 2376– 2378, DOI: 10.1126/science.294.5550.2376Google ScholarThere is no corresponding record for this reference.
- 28Burchell, A. N.; Winer, R. L.; de Sanjosé, S.; Franco, E. L. Epidemiology and Transmission Dynamics of Genital HPV Infection. In Vaccine; Elsevier, 2006; Vol. 24, pp S52- S61. DOI: 10.1016/J.VACCINE.2006.05.031 .Google ScholarThere is no corresponding record for this reference.
- 29Scott-Wittenborn, N.; Fakhry, C. Epidemiology of HPV Related Malignancies. Semin. Radiat. Oncol. 2021, 31 (4), 286– 296, DOI: 10.1016/j.semradonc.2021.04.001Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB2crgvFCrsA%253D%253D&md5=c02ab2a4c081b7b090da25fa1b2134e5Epidemiology of HPV Related MalignanciesScott-Wittenborn Nicholas; Fakhry CaroleSeminars in radiation oncology (2021), 31 (4), 286-296 ISSN:.Human papillomavirus is the most common sexually transmitted infection in the world and had been linked to both anogenital and oropharyngeal cancers. It causes nearly 100% of cervical cancers and an increasing portion of oropharyngeal cancers. The geographical burden of cervical HPV infection and associated cancers is not uniform and is mainly found in low middle income countries in South America, Africa, and Asia. However, HPV-positive oropharyngeal cancer is rapidly becoming more prevalent in high middle income countries. With the development of vaccines which prevent HPV infection, the World Health Organization has designated the extirpation of HPV and its associated cancers a priority. Countries that have implemented adequate vaccine programs have shown a decrease in HPV prevalence. Understanding the epidemiology of HPV and its associated cancers is fundamental in improving vaccine programs and other health programs.
- 30Dahlström, L. A.; Andersson, K.; Luostarinen, T.; Thoresen, S.; Ögmundsdottír, H.; Tryggvadottír, L.; Wiklund, F.; Skare, G. B.; Eklund, C.; Sjölin, K.; Jellum, E.; Koskela, P.; Wadell, G.; Lehtinen, M.; Dillner, J. Prospective Seroepidemiologic Study of Human Papillomavirus and Other Risk Factors in Cervical Cancer. Cancer Epidemiol. Biomarkers Prev. 2011, 20 (12), 2541– 2550, DOI: 10.1158/1055-9965.EPI-11-0761Google ScholarThere is no corresponding record for this reference.
- 31Bravo, I. G.; de Sanjosé, S.; Gottschling, M. The Clinical Importance of Understanding the Evolution of Papillomaviruses. Trends Microbiol. 2010, 18 (10), 432– 438, DOI: 10.1016/j.tim.2010.07.008Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1Wlt77F&md5=74a64d1409e72b76464fbe5e71fa5d89The clinical importance of understanding the evolution of papillomavirusesBravo, Ignacio G.; de Sanjose, Silvia; Gottschling, MarcTrends in Microbiology (2010), 18 (10), 432-438CODEN: TRMIEA; ISSN:0966-842X. (Elsevier Ltd.)A review. A significant fraction of human cancers is assocd. with infections by different papillomaviruses (PVs). In other vertebrates, the presence of specific PVs is also assocd. with different neoplasias. The popular view of PVs conceives them to be largely static and relies on generalized assumptions that have rarely been rigorously tested such as: virus-host codivergence, strict tissue tropism and host-specificity, their very low mutation rate and the absence of recombination. Here, we want to stress the need and the medical importance of understanding the evolutionary history and present-day dynamics of PVs. Understanding the way that PV genomes have evolved will clarify the link between a given genotype and the phenotypic and clin. outcome of the corresponding viral infection.
- 32Berman, B.; Amini, S.; Maderal, A. Condyloma Acuminata. Treat. Ski. Dis. Compr. Ther. Strateg. 2018, 288– 290, DOI: 10.1016/B978-0-7020-6912-3.00089-6Google ScholarThere is no corresponding record for this reference.
- 33Rose, A. B. Introns as Gene Regulators: A Brick on the Accelerator. Front. Genet. 2019, 9 (FEB), 672, DOI: 10.3389/fgene.2018.00672Google ScholarThere is no corresponding record for this reference.
- 34Schiller, J. T.; Lowy, D. R. Understanding and Learning from the Success of Prophylactic Human Papillomavirus Vaccines. Nat. Rev. Microbiol. 2012, 10 (10), 681– 692, DOI: 10.1038/nrmicro2872Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xhtlajs7zK&md5=f2940838733b63ae532a18aebb3d1f87Understanding and learning from the success of prophylactic human papillomavirus vaccinesSchiller, John T.; Lowy, Douglas R.Nature Reviews Microbiology (2012), 10 (10), 681-692CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)A review. An estd. 5% of human cancers are caused by human papillomavirus (HPV) infections, and most of these cancers are of the cervix. Two prophylactic HPV vaccines that target the two most oncogenic virus types, HPV16 and HPV18, are now com. available. In controlled clin. trials, the vaccines proved to be effective at preventing incident anogenital infection and the assocd. neoplastic disease that is induced by these virus types. Here, we highlight the specific aspects of HPV biol. and vaccine compn. that are likely to contribute to the efficacy of these vaccines, and we discuss how these particular features might or might not be relevant for the development of effective vaccines against other sexually transmitted viruses such as HIV and herpes simplex virus (HSV).
- 35Yan, X.; Yu, Z.; Zhang, P.; Battisti, A. J.; Holdaway, H. A.; Chipman, P. R.; Bajaj, C.; Bergoin, M.; Rossmann, M. G.; Baker, T. S. The Capsid Proteins of a Large, Icosahedral DsDNA Virus. J. Mol. Biol. 2009, 385 (4), 1287, DOI: 10.1016/j.jmb.2008.11.002Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXntFemtA%253D%253D&md5=33f09fc2484563d7e564b5206f4520edThe Capsid Proteins of a Large, Icosahedral dsDNA VirusYan, Xiaodong; Yu, Zeyun; Zhang, Ping; Battisti, Anthony J.; Holdaway, Heather A.; Chipman, Paul R.; Bajaj, Chandrajit; Bergoin, Max; Rossmann, Michael G.; Baker, Timothy S.Journal of Molecular Biology (2009), 385 (4), 1287-1299CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)Chilo iridescent virus (CIV) is a large (∼ 1850 Å diam.) insect virus with an icosahedral, T = 147 capsid, a double-stranded DNA (dsDNA) genome, and an internal lipid membrane. The structure of CIV was detd. to 13 Å resoln. by means of cryoelectron microscopy (cryoEM) and three-dimensional image reconstruction. A homol. model of P50, the CIV major capsid protein (MCP), was built based on its amino acid sequence and the structure of the homologous Paramecium bursaria chlorella virus 1 Vp54 MCP. This model was fitted into the cryoEM d. for each of the 25 trimeric CIV capsomers per icosahedral asym. unit. A difference map, in which the fitted CIV MCP capsomers were subtracted from the CIV cryoEM reconstruction, showed that there are at least three different types of minor capsid proteins assocd. with the capsomers outside the lipid membrane. "Finger" proteins are situated at many, but not all, of the spaces between three adjacent capsomers within each trisymmetron, and "zip" proteins are situated between sets of three adjacent capsomers at the boundary between neighboring trisymmetrons and pentasymmetrons. Based on the results of segmentation and d. correlations, there are at least eight finger proteins and three dimeric and two monomeric zip proteins in one asym. unit of the CIV capsid. These minor proteins appear to stabilize the virus by acting as intercapsomer crosslinks. One transmembrane "anchor" protein per icosahedral asym. unit, which extends from beneath one of the capsomers in the pentasymmetron to the internal leaflet of the lipid membrane, may provide addnl. stabilization for the capsid. These results are consistent with the observations for other large, icosahedral dsDNA viruses that also utilize minor capsid proteins for stabilization and for detg. their assembly.
- 36Sapp, M.; Bienkowska-Haba, M. Viral Entry Mechanisms: Human Papillomavirus and a Long Journey from Extracellular Matrix to the Nucleus. FEBS J. 2009, 276 (24), 7206– 7216, DOI: 10.1111/j.1742-4658.2009.07400.xGoogle Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFGqtr3I&md5=5c8a0c61a260a4afaf1286048460e6eeViral entry mechanisms: human papillomavirus and a long journey from extracellular matrix to the nucleusSapp, Martin; Bienkowska-Haba, MalgorzataFEBS Journal (2009), 276 (24), 7206-7216CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)A review. Papillomaviruses are epitheliotropic non-enveloped double-stranded DNA viruses, whose replication is strictly dependent on the terminally differentiating tissue of the epidermis. They induce self-limiting benign tumors of skin and mucosa, which may progress to malignancy (e.g. cervical carcinoma). Prior to entry into basal cells, virions attach to heparan sulfate moieties of the basement membrane. This triggers conformational changes, which affect both capsid proteins, L1 and L2, and such changes are a prerequisite for interaction with the elusive uptake receptor. These processes are very slow, resulting in an uptake half-time of up to 14 h. This minireview summarizes recent advances in our understanding of cell surface events, internalization and the subsequent intracellular trafficking of papillomaviruses.
- 37Rubio, I.; Seitz, H.; Canali, E.; Sehr, P.; Bolchi, A.; Tommasino, M.; Ottonello, S.; Müller, M. The N-Terminal Region of the Human Papillomavirus L2 Protein Contains Overlapping Binding Sites for Neutralizing, Cross-Neutralizing and Non-Neutralizing Antibodies. Virology 2011, 409 (2), 348– 359, DOI: 10.1016/j.virol.2010.10.017Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsF2isL%252FL&md5=66cbba5c32ff30f5874eb73a41f613d0The N-terminal region of the human papillomavirus L2 protein contains overlapping binding sites for neutralizing, cross-neutralizing and non-neutralizing antibodiesRubio, Ivonne; Seitz, Hanna; Canali, Elena; Sehr, Peter; Bolchi, Angelo; Tommasino, Massimo; Ottonello, Simone; Mueller, MartinVirology (2011), 409 (2), 348-359CODEN: VIRLAX; ISSN:0042-6822. (Elsevier B.V.)The N-terminal region of the human papillomavirus (HPV) L2 protein has been shown to contain immune epitopes able to induce the prodn. of neutralizing and cross-neutralizing antibodies. Using bacterial thioredoxin as a scaffold, we managed to enhance the immunogenicity of putative L2 neutralizing epitopes, but only a minor fraction of the resulting immune responses was found to be neutralizing. To det. the recognition patterns for non-neutralizing, neutralizing and cross-neutralizing antibodies, we isolated and characterized a panel of 46 monoclonal antibodies directed against different HPV16 L2 epitopes. Four of such antibodies proved to be neutralizing, and two of them, both targeting the amino acid (aa) 20-38 region of L2, were found to cross-neutralize a broad range of papillomaviruses. The epitopes recognized by neutralizing and cross-neutralizing antibodies were mapped at high resoln. and were found to be characterized by distinct recognition patterns. Even in the case of the L2 20-38 epitope, cross-neutralization of HPV31 pseudovirions proved to be extremely inefficient, and this was found to be primarily due to the lack of a proline residue at position 30. HPV16 specific amino acids in this region also appear to be responsible for the lack of cross-neutralizing activity, thus suggesting a potential immune escape mechanism. For the aa 71-80 region, instead, the data indicate that restriction of neutralization to HPV16 is due to sequence (or structural) differences laying outside of the epitope. Besides providing new insights on the mol. bases of L2-mediated immune reactivity, the present data may pave the way to novel vaccination approaches specifically evoking cross-neutralizing antibody responses.
- 38Kines, R. C.; Thompson, C. D.; Lowy, D. R.; Schiller, J. T.; Day, P. M. The Initial Steps Leading to Papillomavirus Infection Occur on the Basement Membrane Prior to Cell Surface Binding. Proc. Natl. Acad. Sci. U. S. A. 2009, 106 (48), 20458– 20463, DOI: 10.1073/pnas.0908502106Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjtFWitg%253D%253D&md5=fd0a2d49924f15ed9bfd1ce2f739c16aThe initial steps leading to papillomavirus infection occur on the basement membrane prior to cell surface bindingKines, Rhonda C.; Thompson, Cynthia D.; Lowy, Douglas R.; Schiller, John T.; Day, Patricia M.Proceedings of the National Academy of Sciences of the United States of America (2009), 106 (48), 20458-20463, S20458/1-S20458/5CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Using a murine challenge model, we previously detd. that human papillomavirus (HPV) pseudovirions initially bind preferentially to the cervicovaginal basement membrane (BM) at sites of trauma. We now report that the capsids undergo a conformational change while bound to the BM that results in L2 cleavage by a proprotein convertase (PC), furin, and/or PC5/6, followed by the exposure of an N-terminal cross-neutralization L2 epitope and transfer of the capsids to the epithelial cell surface. Prevention of this exposure by PC inhibition results in detachment of the pseudovirions from the BM and their eventual loss from the tissue, thereby preventing infection. Pseudovirions whose L2 had been precleaved by furin can bypass the PC inhibition of binding and infectivity. Cleavage of heparan sulfate proteoglycans (HSPG) with heparinase III prevented infection and BM binding by the precleaved pseudovirions, but did not prevent them from binding robustly to cell surfaces. These results indicate that the infectious process has evolved so that the initial steps take place on the BM, in contrast to the typical viral infection that is initiated by binding to the cell surface. The data are consistent with a dynamic model of in vivo HPV infection in which a conformational change and PC cleavage on the BM allows transfer of virions from HSPG attachment factors to an LI-specific receptor on basal keratinocytes migrating into the site of trauma.
- 39Wang, J. W.; Roden, R. B. S. L2, the Minor Capsid Protein of Papillomavirus. Virology 2013, 445 (1–2), 175– 186, DOI: 10.1016/j.virol.2013.04.017Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXot1KrtLk%253D&md5=b518366ee9b84e91de7ef1e7496ad43bL2, the minor capsid protein of papillomavirusWang, Joshua W.; Roden, Richard B. S.Virology (2013), 445 (1-2), 175-186CODEN: VIRLAX; ISSN:0042-6822. (Elsevier B.V.)A review. The capsid protein L2 plays major roles in both papillomavirus assembly and the infectious process. While L1 forms the majority of the capsid and can self-assemble into empty virus-like particles (VLPs), L2 is a minor capsid component and lacks the capacity to form VLPs. However, L2 co-assembles with L1 into VLPs, enhancing their assembly. L2 also facilitates encapsidation of the ∼8 kbp circular and nucleosome-bound viral genome during assembly of the non-enveloped T=7d virions in the nucleus of terminally differentiated epithelial cells, although, like L1, L2 is not detectably expressed in infected basal cells. With respect to infection, L2 is not required for particles to bind to and enter cells. However L2 must be cleaved by furin for endosome escape. L2 then travels with the viral genome to the nucleus, wherein it accumulates at ND-10 domains. Here, we provide an overview of the biol. of L2.
- 40Münger, K.; Baldwin, A.; Edwards, K. M.; Hayakawa, H.; Nguyen, C. L.; Owens, M.; Grace, M.; Huh, K. Mechanisms of Human Papillomavirus-Induced Oncogenesis. J. Virol. 2004, 78 (21), 11451, DOI: 10.1128/JVI.78.21.11451-11460.2004Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptVKmur4%253D&md5=3f018e3e8e692de3638bfe23a426f036Mechanisms of human papillomavirus-induced oncogenesisMuenger, Karl; Baldwin, Amy; Edwards, Kirsten M.; Hayakawa, Hiroyuki; Nguyen, Christine L.; Owens, Michael; Grace, Miranda; Huh, KyungWonJournal of Virology (2004), 78 (21), 11451-11460CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)A review. The role of E6 and E7 as oncoproteins of HPV in human oncogenesis is discussed.
- 41Middleton, K.; Peh, W.; Southern, S.; Griffin, H.; Sotlar, K.; Nakahara, T.; El-Sherif, A.; Morris, L.; Seth, R.; Hibma, M.; Jenkins, D.; Lambert, P.; Coleman, N.; Doorbar, J. Organization of Human Papillomavirus Productive Cycle during Neoplastic Progression Provides a Basis for Selection of Diagnostic Markers. J. Virol. 2003, 77 (19), 10186– 10201, DOI: 10.1128/JVI.77.19.10186-10201.2003Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXnsFyms7w%253D&md5=a275189dd55496aaafb427668841e234Organization of human papillomavirus productive cycle during neoplastic progression provides a basis for selection of diagnostic markersMiddleton, Kate; Peh, Woei; Southern, Shirley; Griffin, Heather; Sotlar, Karl; Nakahara, Tomomi; El-Sherif, Amira; Morris, Lesley; Seth, Rashmi; Hibma, Merilyn; Jenkins, David; Lambert, Paul; Coleman, Nicholas; Doorbar, JohnJournal of Virology (2003), 77 (19), 10186-10201CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)The productive cycle of human papillomaviruses (HPVs) can be divided into discrete phases. Cell proliferation and episomal maintenance in the lower epithelial layers are followed by genome amplification and the expression of capsid proteins. These events, which occur in all productive infections, can be distinguished by using antibodies to viral gene products or to surrogate markers of their expression. Here we have compared precancerous lesions caused by HPV type 16 (HPV16) with lesions caused by HPV types that are not generally assocd. with human cancer. These include HPV2 and HPV11, which are related to HPV16 (supergroup A), as well as HPV1 and HPV65, which are evolutionarily divergent (supergroups E and B). HPV16-induced low-grade squamous intraepithelial lesions (CIN1) are productive infections which resemble those caused by other HPV types. During progression to cancer, however, the activation of late events is delayed, and the thickness of the proliferative compartment is progressively increased. In many HPV16-induced high-grade squamous intraepithelial lesions (CIN3), late events are restricted to small areas close to the epithelial surface. Such heterogeneity in the organization of the productive cycle was seen only in lesions caused by HPV16 and was not apparent when lesions caused by other HPV types were compared. By contrast, the order in which events in the productive cycle were initiated was invariant and did not depend on the infecting HPV type or the severity of disease. The distribution of viral gene products in the infected cervix depends on the extent to which the virus can complete its productive cycle, which in turn reflects the severity of cervical neoplasia. It appears from our work that the presence of such proteins in cells at the epithelial surface allows the severity of the underlying disease to be predicted and that markers of viral gene expression may improve cervical screening.
- 42Isaacson Wechsler, E.; Wang, Q.; Roberts, I.; Pagliarulo, E.; Jackson, D.; Untersperger, C.; Coleman, N.; Griffin, H.; Doorbar, J. Reconstruction of Human Papillomavirus Type 16-Mediated Early-Stage Neoplasia Implicates E6/E7 Deregulation and the Loss of Contact Inhibition in Neoplastic Progression. J. Virol. 2012, 86 (11), 6358– 6364, DOI: 10.1128/JVI.07069-11Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38rgsVGgug%253D%253D&md5=c2f6c00bcd1a0ca78799595630253d19Reconstruction of human papillomavirus type 16-mediated early-stage neoplasia implicates E6/E7 deregulation and the loss of contact inhibition in neoplastic progressionIsaacson Wechsler Erin; Wang Qian; Roberts Ian; Pagliarulo Emilio; Jackson Deborah; Untersperger Christina; Coleman Nick; Griffin Heather; Doorbar JohnJournal of virology (2012), 86 (11), 6358-64 ISSN:.Infection with human papillomavirus type 16 (HPV-16) can lead to low- or high-grade squamous intraepithelial lesions (LSIL or HSIL). Here we show that these in vivo disease states can be replicated in raft cultures of early-pass HPV-16 episomal cell lines, at both the level of pathology and the level of viral gene expression. A reduced responsiveness to cell-cell contact inhibition and an increase in E6/E7 activity correlated closely with phenotype. Similar deregulation is likely to underlie the appearance of LSIL or HSIL soon after infection.
- 43Mittal, S.; Banks, L. Molecular Mechanisms Underlying Human Papillomavirus E6 and E7 Oncoprotein-Induced Cell Transformation. Mutat. Res. Rev. Mutat. Res. 2017, 772, 23– 35, DOI: 10.1016/j.mrrev.2016.08.001Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhtlyjtb3L&md5=22b10bb984bd87a3f626bf6fe9d2b45dMolecular mechanisms underlying human papillomavirus E6 and E7 oncoprotein-induced cell transformationMittal, Suruchi; Banks, LawrenceMutation Research, Reviews in Mutation Research (2017), 772 (), 23-35CODEN: MRRRFK; ISSN:1383-5742. (Elsevier B.V.)Human papillomaviruses (HPVs) are the causative agents of 5% of all human cancers, with cervical cancer being the most important. Two viral oncoproteins, E6 and E7, are essential for the development and maintenance of malignancy. Both proteins function by targeting crit. pathways that are essential for maintaining cellular homeostasis. As a consequence of these activities, this produces an environment that is favorable for the normal viral life cycle, but when perturbed, can result in the initiation of changes to the host cell, which ultimately results in the development of a malignancy. In this review we discuss the role of these different functions of the viral oncoproteins during the viral life cycle and carcinogenesis, with an emphasis on how induction of DNA damage by the viral oncoproteins, in conjunction with the stem like nature of the target cells, can ultimately result in the development of cancer.
- 44Doorbar, J.; Egawa, N.; Griffin, H.; Kranjec, C.; Murakami, I. Human Papillomavirus Molecular Biology and Disease Association. Rev. Med. Virol. 2015, 25 (S1), 2– 23, DOI: 10.1002/rmv.1822Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXjvVOltbc%253D&md5=49f907ac48e8670df0009d74baeaa7afHuman papillomavirus molecular biology and disease associationDoorbar, John; Egawa, Nagayasu; Griffin, Heather; Kranjec, Christian; Murakami, IsaoReviews in Medical Virology (2015), 25 (S1), 2-23CODEN: RMVIEW; ISSN:1052-9276. (John Wiley & Sons Ltd.)Summary : Human papillomaviruses (HPVs) have evolved over millions of years to propagate themselves in a range of different animal species including humans. Viruses that have co-evolved slowly in this way typically cause chronic inapparent infections, with virion prodn. in the absence of apparent disease. This is the case for many Beta and Gamma HPV types. The Alpha papillomavirus types have however evolved immunoevasion strategies that allow them to cause persistent visible papillomas. These viruses activate the cell cycle as the infected epithelial cell differentiates in order to create a replication competent environment that allows viral genome amplification and packaging into infectious particles. This is mediated by the viral E6, E7, and E5 proteins. High-risk E6 and E7 proteins differ from their low-risk counterparts however in being able to drive cell cycle entry in the upper epithelial layers and also to stimulate cell proliferation in the basal and parabasal layers. Deregulated expression of these cell cycle regulators underlies neoplasia and the eventual progression to cancer in individuals who cannot resolve high-risk HPV infection. Most work to date has focused on the study of high-risk HPV types such as HPV 16 and 18, which has led to an understanding of the mol. pathways subverted by these viruses. Such approaches will lead to the development of better strategies for disease treatment, including targeted antivirals and immunotherapeutics. Priorities are now focused toward understanding HPV neoplasias at sites other than the cervix (e.g. tonsils, other transformation zones) and toward understanding the mechanisms by which low-risk HPV types can sometimes give rise to papillomatosis and under certain situations even cancers. Copyright © 2015 John Wiley & Sons, Ltd.
- 45Galloway, D. A.; Laimins, L. A. Human Papillomaviruses: Shared and Distinct Pathways for Pathogenesis. Curr. Opin. Virol. 2015, 14, 87– 92, DOI: 10.1016/j.coviro.2015.09.001Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFShsbvM&md5=4185bbe221deee14497f3cae31366672Human papillomaviruses: shared and distinct pathways for pathogenesisGalloway, Denise A.; Laimins, Laimonis A.Current Opinion in Virology (2015), 14 (), 87-92CODEN: COVUAF; ISSN:1879-6257. (Elsevier B. V.)A review. Over 200 types of human papillomaviruses (HPV) have been identified that infect epithelial cells at different anat. locations. HPVs are grouped into five genera with the alpha and beta viruses being the most commonly studied. Members of the alpha HPV genus infect genital epithelia and are the causative agents of many anogenital cancers. Beta HPVs infect cutaneous epithelia and have been suggested as co-factors in the development of non-melanoma skin cancers. Recent studies have shown that activation of DNA damage pathways is important for the productive life cycle of the alpha HPVs while the beta viruses suppress their activation. These differences likely contribute to the varying types of lesions and malignancies that are assocd. with these viruses.
- 46Albrecht, D.; Meyer, T.; Lorenzen, T.; Stoehr, A.; Arndt, R.; Plettenberg, A. Epidemiology of HHV-8 Infection in HIV-Positive Patients with and without Kaposi Sarcoma: Diagnostic Relevance of Serology and PCR. J. Clin. Virol. 2004, 30 (2), 145– 149, DOI: 10.1016/j.jcv.2003.09.017Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2c3isl2rsg%253D%253D&md5=fa6feb57cc659855b4f4147dd4657ad5Epidemiology of HHV-8 infection in HIV-positive patients with and without Kaposi sarcoma: diagnostic relevance of serology and PCRAlbrecht Dirk; Meyer Thomas; Lorenzen Thore; Stoehr Albrecht; Arndt Ruediger; Plettenberg AndreasJournal of clinical virology : the official publication of the Pan American Society for Clinical Virology (2004), 30 (2), 145-9 ISSN:1386-6532.BACKGROUND: Kaposi's sarcoma (KS) remains the most common neoplasm in HIV-infected patients. Human herpesvirus 8 (HHV-8) infection is etiologically associated with KS. Diagnostic procedures with regard to HHV-8 infection are not routinely performed in HIV-infected patients; diagnostic and prognostic value of HHV-8 serology or PCR are unknown in this setting. Epidemiological data concerning HHV-8 infection of HIV-infected patients in Germany are rare. OBJECTIVES: To assess prevalence of HHV-8 infection in a cohort of HIV-infected patients with and without KS in Germany and to correlate this to manifestations and clinical course of KS. STUDY DESIGN: HHV-8 serology was performed in 483 patients in routine care for HIV-infection in northern Germany. HHV-8 DNA was analyzed by PCR in peripheral blood mononuclear cells (PBMC) of 293 patients; in a subgroup multiple samples were analyzed. History and manifestations of KS were recorded. RESULTS: HHV-8 antibodies were detected using IFT in 91% of 33 patients with KS and 52% of 398 patients without KS. In 36 of 293 (12.3%) patients HHV-8 DNA was detected in PBMC. In general, HHV-8 DNA was not continuously detected when multiple samples from the same patient were analyzed. Patients with KS history were more likely to be PCR positive than those without (45.5% versus 7.8%). In patients with active KS HHV-8 DNA was detected more frequently than in patients with disease remission. HHV-8 DNA was not detected in serologically negative patients. However, three patients with KS history in full remission for several years were seronegative. CONCLUSIONS: HIV-infected patients were frequently found to be positive for HHV-8 antibodies. The number of patients positive for viral DNA in PBMC was much smaller. Single PCR-examinations were of little value for prognosis, but repeated detection of HHV-8 DNA represents an increased risk of disease activity.
- 47Rohner, E.; Wyss, N.; Trelle, S.; Mbulaiteye, S. M.; Egger, M.; Novak, U.; Zwahlen, M.; Bohlius, J. HHV-8 Seroprevalence: A Global View. Syst. Rev. 2014, 3 (1), 1– 7, DOI: 10.1186/2046-4053-3-11Google ScholarThere is no corresponding record for this reference.
- 48Rewane, A; Tadi, P. Herpes virus type 8. In: StatPearls. StatPearls Publishing: Treasure Island (FL); 2022. pp. 1– 8. https://www.ncbi.nlm.nih.gov/books/NBK556023/.Google ScholarThere is no corresponding record for this reference.
- 49Dittmer, D. P.; Damania, B. Kaposi Sarcoma-Associated Herpesvirus: Immunobiology, Oncogenesis, and Therapy. J. Clin. Invest. 2016, 126 (9), 3165– 3175, DOI: 10.1172/JCI84418Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2szmvFyqtA%253D%253D&md5=2c9694bffbda1d72aaeafa30b5b7968aKaposi sarcoma-associated herpesvirus: immunobiology, oncogenesis, and therapyDittmer Dirk P; Damania BlossomThe Journal of clinical investigation (2016), 126 (9), 3165-75 ISSN:.Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is the etiologic agent underlying Kaposi sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. This human gammaherpesvirus was discovered in 1994 by Drs. Yuan Chang and Patrick Moore. Today, there are over five thousand publications on KSHV and its associated malignancies. In this article, we review recent and ongoing developments in the KSHV field, including molecular mechanisms of KSHV pathogenesis, clinical aspects of KSHV-associated diseases, and current treatments for cancers associated with this virus.
- 50Purushothaman, P.; Uppal, T.; Verma, S. C. Molecular Biology of KSHV Lytic Reactivation. Viruses 2015, 7 (1), 116, DOI: 10.3390/v7010116Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXislWqsrc%253D&md5=1200b2d4f766b2727214182c2aa79ad8Molecular biology of KSHV lytic reactivationPurushothaman, Pravinkumar; Uppal, Timsy; Verma, Subhash C.Viruses (2015), 7 (1), 116-153/1-116-153/38, 38 pp.CODEN: VIRUBR; ISSN:1999-4915. (MDPI AG)A review. Kaposi's sarcoma-assocd. herpesvirus (KSHV) primarily persists as a latent episome in infected cells. During latent infection, only a limited no. of viral genes are expressed that help to maintain the viral episome and prevent lytic reactivation. The latent KSHV genome persists as a highly ordered chromatin structure with bivalent chromatin marks at the promoter-regulatory region of the major immediate-early gene promoter. Various stimuli can induce chromatin modifications to an active euchromatic epigenetic mark, leading to the expression of genes required for the transition from the latent to the lytic phase of KSHV life cycle. Enhanced replication and transcription activator (RTA) gene expression triggers a cascade of events, resulting in the modulation of various cellular pathways to support viral DNA synthesis. RTA also binds to the origin of lytic DNA replication to recruit viral, as well as cellular, proteins for the initiation of the lytic DNA replication of KSHV. In this review we will discuss some of the pivotal genetic and epigenetic factors that control KSHV reactivation from the transcriptionally restricted latent program.
- 51Cesarman, E.; Damania, B.; Krown, S. E.; Martin, J.; Bower, M.; Whitby, D. Kaposi Sarcoma. Nat. Rev. Dis. Prim. 2019 51 2019, 5 (1), 1– 21, DOI: 10.1038/s41572-019-0060-9Google ScholarThere is no corresponding record for this reference.
- 52Cousins, E.; Nicholas, J. Molecular Biology of Human Herpesvirus 8: Novel Functions and Virus-Host Interactions Implicated in Viral Pathogenesis and Replication. Recent Results Cancer Res. 2014, 193, 227, DOI: 10.1007/978-3-642-38965-8_13Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXosVOqtLo%253D&md5=5078fe376e966e9bda5c713a07f777afMolecular Biology of Human Herpesvirus 8: Novel Functions and Virus-Host Interactions Implicated in Viral Pathogenesis and ReplicationCousins, Emily; Nicholas, JohnRecent Results in Cancer Research (2014), 193 (Viruses and Human Cancer), 227-268CODEN: RRCRBU; ISSN:2197-6767. (Springer GmbH)Human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma-assocd. herpesvirus (KSHV), is the second identified human gammaherpesvirus. Like its relative Epstein-Barr virus, HHV-8 is linked to B-cell tumors, specifically primary effusion lymphoma and multicentric Castleman's disease, in addn. to endothelial-derived KS. HHV-8 is unusual in its possession of a plethora of "accessory" genes and encoded proteins in addn. to the core, conserved herpesvirus and gammaherpesvirus genes that are necessary for basic biol. functions of these viruses. The HHV-8 accessory proteins specify not only activities deducible from their cellular protein homologies but also novel, unsuspected activities that have revealed new mechanisms of virus-host interaction that serve virus replication or latency and may contribute to the development and progression of virus-assocd. neoplasia. These proteins include viral interleukin-6 (vIL-6), viral chemokines (vCCLs), viral G protein-coupled receptor (vGPCR), viral interferon regulatory factors (vIRFs), and viral antiapoptotic proteins homologous to FLICE (FADD-like IL-1β converting enzyme)-inhibitory protein (FLIP) and survivin. Other HHV-8 proteins, such as signaling membrane receptors encoded by open reading frames K1 and K15, also interact with host mechanisms in unique ways and have been implicated in viral pathogenesis. Addnl., a set of micro-RNAs encoded by HHV-8 appear to modulate expression of multiple host proteins to provide conditions conducive to virus persistence within the host and could also contribute to HHV-8-induced neoplasia. Here, we review the mol. biol. underlying these novel virus-host interactions and their potential roles in both virus biol. and virus-assocd. disease.
- 53Angius, F.; Ingianni, A.; Pompei, R. Human Herpesvirus 8 and Host-Cell Interaction: Long-Lasting Physiological Modifications, Inflammation and Related Chronic Diseases. Microorganisms 2020, 8 (3), 388, DOI: 10.3390/microorganisms8030388Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVWjtr7N&md5=0dbc4fa238d6831cc492999fd650ca77Human herpesvirus 8 and host-cell interaction: long-lasting physiological modifications, inflammation and related chronic diseasesAngius, Fabrizio; Ingianni, Angela; Pompei, RaffaelloMicroorganisms (2020), 8 (3), 388CODEN: MICRKN; ISSN:2076-2607. (MDPI AG)Oncogenic and latent-persistent viruses belonging to both DNA and RNA groups are known to cause serious metab. alterations. Among these, the Human Herpesvirus 8 (HHV8) infection induces stable modifications in biochem. and cellular metab., which in turn affect its own pathol. properties. HHV8 enhances the expression of insulin receptors, supports the accumulation of neutral lipids in cytoplasmic lipid droplets and induces alterations in both triglycerides and cholesterol metab. in endothelial cells. In addn., HHV8 is also known to modify immune response and cytokine prodn. with implications for cell oxidative status (i.e., reactive oxygen species activation). This review underlines the recent findings regarding the role of latent and persistent HHV8 viral infection in host physiol. and pathogenesis.
- 54Cannon, M.; Philpott, N. J.; Cesarman, E. The Kaposi’s Sarcoma-Associated Herpesvirus G Protein-Coupled Receptor Has Broad Signaling Effects in Primary Effusion Lymphoma Cells. J. Virol. 2003, 77 (1), 57, DOI: 10.1128/JVI.77.1.57-67.2003Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXkt1Sg&md5=12febc1c85e92e66e24996561f7ea5ebThe Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor has broad signaling effects in primary effusion lymphoma cellsCannon, Mark; Philpott, Nicola J.; Cesarman, EthelJournal of Virology (2003), 77 (1), 57-67CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Kaposi's sarcoma-assocd. herpesvirus [KSHV/human herpesvirus 8 (HHV-8)] is a gamma-2-herpesvirus responsible for Kaposi's sarcoma as well as primary effusion lymphoma (PEL). KSHV is a lymphotropic virus that has pirated many mammalian genes involved in inflammation, cell cycle control, and angiogenesis. Among these is the early lytic viral G protein-coupled receptor (vGPCR), a homolog of the human interleukin-8 (IL-8) receptor. When expressed, vGPCR is constitutively active and can signal via mitogen- and stress-activated kinases. In certain models it activates the transcriptional potential of NF-κB and activator protein 1 (AP-1) and induces vascular endothelial growth factor (VEGF) prodn. Despite its importance to the pathogenesis of all KSHV-mediated disease, little is known about vGPCR activity in hematopoietic cells. To study the signaling potential and downstream effects of vGPCR in such cells, the authors have developed PEL cell lines that express vGPCR under the control of an inducible promoter. The sequences required for tetracycline-mediated induction were cloned into a plasmid contg. adeno-assocd. virus type 2 elements to enhance integration efficiency. This novel plasmid permitted studies of vGPCR activity in naturally infected KSHV-pos. lymphocytes. The authors show that vGPCR activates ERK-2 and p38 in PEL cells. In addn., it increases the transcription of reporter genes under the control of AP-1, NF-κB, CREB, and NFAT, a Ca2+-dependent transcription factor important to KSHV lytic gene expression. VGPCR also increases the transcription of KSHV open reading frames 50 and 57, thereby displaying broad potential to affect viral transcription patterns. Finally, vGPCR signaling results in increased PEL cell elaboration of KSHV vIL-6 and VEGF, growth factors involved in KSHV-mediated disease pathogenesis.
- 55Brinkmann, M. M.; Pietrek, M.; Dittrich-Breiholz, O.; Kracht, M.; Schulz, T. F. Modulation of Host Gene Expression by the K15 Protein of Kaposi’s Sarcoma-Associated Herpesvirus. J. Virol. 2007, 81 (1), 42, DOI: 10.1128/JVI.00648-06Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtlCgu73L&md5=f7fb535acada42a1ca5a4f5ef83d6823Modulation of host gene expression by the K15 protein of Kaposi's sarcoma-associated herpesvirusBrinkmann, Melanie M.; Pietrek, Marcel; Dittrich-Breiholz, Oliver; Kracht, Michael; Schulz, Thomas F.Journal of Virology (2007), 81 (1), 42-58CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Kaposi's sarcoma-assocd. herpesvirus (KSHV) contains several open reading frames (ORFs) encoding proteins capable of initiating signal transduction pathways. Among them is the K15 ORF, which consists of eight exons encoding a protein with 12 predicted transmembrane domains and a cytoplasmic C terminus. When transiently expressed, the 8-exon K15 transcript gives rise to a protein with an apparent mol. mass of 45 kDa. K15 interacts with cellular proteins, TRAF (tumor necrosis factor receptor-assocd. factor) and Src kinases, and activates AP-1, NF-κB, and the mitogen-activated protein kinases (MAPKs) c-jun-N-terminal kinase and extracellular signal-regulated kinase. This signaling activity of K15 is related to phosphorylation of Y481 of the K15 SH2-B motif Y481 EEV. In this study we demonstrate the expression of an endogenous 45-kDa K15 protein in KSHV BAC36-infected epithelial cells. This endogenous K15 protein shows the same intracellular localization as transiently expressed K15, and expression kinetic studies suggest it to be a lytic gene. We have further detd. the downstream target genes of K15 signaling using DNA oligonucleotide microarrays. We demonstrate that K15 is capable of inducing expression of multiple cytokines and chemokines, including interleukin-8 (IL-8), IL-6, CCL20, CCL2, CXCL3, and IL-1α/β, as well as expression of Dscr1 and Cox-2. In epithelial cells, K15-induced upregulation of most genes was dependent on phosphorylation of Y481, whereas in endothelial cells mutation of Y481 did not result in a complete loss of Dscr1 and Cox-2 expression and NFAT-activity. Our study establishes K15 as one of the KSHV lytic genes that are inducing expression of multiple cytokines, which have been shown to play an important role in KSHV-assocd. pathogenesis.
- 56Caselli, E.; Fiorentini, S.; Amici, C.; Di Luca, D.; Caruso, A.; Santoro, M. G. Human Herpesvirus 8 Acute Infection of Endothelial Cells Induces Monocyte Chemoattractant Protein 1-Dependent Capillary-like Structure Formation: Role of the IKK/NF-KappaB Pathway. Blood 2007, 109 (7), 2718– 2726, DOI: 10.1182/blood-2006-03-012500Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjvVynsr4%253D&md5=df1a8e1f979aeefb39a3cfecd8643996Human herpesvirus 8 acute infection of endothelial cells induces monocyte chemoattractant protein 1-dependent capillary-like structure formation: role of the IKK/NF-κB pathwayCaselli, Elisabetta; Fiorentini, Simona; Amici, Carla; Di Luca, Dario; Caruso, Arnaldo; Santoro, M. GabriellaBlood (2007), 109 (7), 2718-2726CODEN: BLOOAW; ISSN:0006-4971. (American Society of Hematology)Human herpesvirus 8 (HHV-8) is considered the causative agent of Kaposi sarcoma, a highly vascularized neoplasm characterized by spindle-shaped cells of endothelial origin and inflammatory cell infiltration. The cell transforming ability of HHV-8 has been assocd. with the activation of NF-κB, a nuclear factor playing a pivotal role in promoting inflammation and cell proliferation; however, little is known about NF-κB activation during acute HHV-8 infection. In the present study, we used a recently established in vitro model of HHV-8 acute productive infection in endothelial cells to investigate the effect of HHV-8 on NF-κB activity and function. HHV-8 rapidly and potently induced NF-κB activity in endothelial cells via stimulation of the IκB kinase (IKK). Following IKK activation, HHV-8 selectively triggered the prodn. of high levels of monocyte chemoattractant protein 1 (MCP-1), whereas it did not affect the expression of other NF-κB-dependent proinflammatory proteins, including TNF-α, IL-8, and RANTES. Deletion of NF-κB-binding sites in the MCP-1 enhancer resulted in significant inhibition of HHV-8-induced transcription. Furthermore, MCP-1 prodn. was accompanied by virus-induced capillary-like structure formation at early stages of infection. The results suggest that HHV-8-induced MCP-1 may play an important role in promoting inflammation and pathogenic angiogenesis typical of HHV-8-assocd. lesions.
- 57Jham, B. C.; Montaner, S. The Kaposi’s Sarcoma-Associated Herpesvirus G Protein Coupled Receptor: Lessons on Dysregulated Angiogenesis from a Viral Oncogene. J. Cell. Biochem. 2010, 110 (1), 1, DOI: 10.1002/jcb.22524Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXltVGitrc%253D&md5=725db5d8fc308d90cc16fbc4b80caa62The Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor: lessons on dysregulated angiogenesis from a viral oncogeneJham, Bruno C.; Montaner, SilviaJournal of Cellular Biochemistry (2010), 110 (1), 1-9CODEN: JCEBD5; ISSN:0730-2312. (Wiley-Liss, Inc.)A review. Tumor viruses can induce cell transformation by overcoming cellular defense mechanisms and promoting the ungoverned proliferation of infected cells. To this end, functionally related viral oncogenes have evolved in disparate viruses to over-ride key proliferative and survival intracellular pathways, thus assuring efficient viral replication and contributing to tumor formation. Indeed, the study of viral oncogenes was a powerful tool for disclosing fundamental insights into these basic cellular processes. In this regard, the Kaposi's sarcoma-assocd. herpesvirus (KSHV or HHV8), the etiol. agent of Kaposi's sarcoma (KS), is an exemplary model of an oncogenic virus that includes within its genome several homologues of cellular genes implicated in the regulation of cell proliferation and apoptosis. However, emerging evidence now points to a single KSHV gene, ORF74, encoding for the viral G protein-coupled receptor (vGPCR), as essential for KS development. Expressed in only a fraction of cells within KS lesions, this viral receptor induces tumorigenesis through both autocrine and paracrine mechanisms. Indeed, work from several labs. has demonstrated that vGPCR can promote cell proliferation, enhance cell survival, modulate cell migration, stimulate angiogenesis, and recruit inflammatory cells, both in expressing cells, as well as in neighboring (bystander) cells. Examn. of this powerful viral oncogene may expose novel targets for the treatment of patients with KS and could ultimately provide a unique perspective into how GPCRs, and specifically chemokine receptors, contribute to angiogenesis and tumorigenesis.
- 58Verma, S. C.; Lan, K.; Robertson, E. Structure and Function of Latency-Associated Nuclear Antigen. Curr. Top. Microbiol. Immunol. 2007, 312, 101, DOI: 10.1007/978-3-540-34344-8_4Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlagur%252FF&md5=e974f5ab3c0068a87356f78d438d7f2fStructure and function of latency-associated nuclear antigenVerma, S. C.; Lan, K.; Robertson, E.Current Topics in Microbiology and Immunology (2007), 312 (Kaposi Sarcoma Herpesvirus), 101-136CODEN: CTMIA3; ISSN:0070-217X. (Springer GmbH)A review. Latency-assocd. nuclear antigen (LANA) encoded by open reading frame 73 (ORF73) is the major latent protein expressed in all forms of KSHV-assocd. malignancies. LANA is a large (222-234 kDa) nuclear protein that interacts with various cellular as well as viral proteins. LANA has been classified as an oncogenic protein as it dysregulates various cellular pathways including tumor suppressor pathways assocd. with pRb and p53 and can transform primary rat embryo fibroblasts in cooperation with the cellular oncogene Hras. It assocs. with GSK-3β, an important modulator of Wnt signaling pathway leading to the accumulation of cytoplasmic β-catenin, which upregulates Tcf/Lef regulated genes after entering into the nucleus. LANA also blocks the expression of RTA, the reactivation transcriptional activator, which is crit. for the latency to lytic switch, and thus helps in maintaining viral latency. LANA tethers the viral episomal DNA to the host chromosomes by directly binding to its cognate binding sequence within the TR region of the genome through its C terminus and to the nucleosomes through the N terminus of the mol. Tethering to the host chromosomes helps in efficient partitioning of the viral episomes in the dividing cells. Disruptions of LANA expression led to redn. in the episomal copies of the viral DNA, supporting its role in persistence of the viral DNA. The functions known so far suggest that LANA is a key player in KSHV-mediated pathogenesis.
- 59Jarrett, A. F.; Armstrong, A. A.; Alexander, E. Epidemiology of EBV and Hodgkin’s Lymphoma. Ann. Oncol. 1996, 7 (SUPPL. 4), S5– S10, DOI: 10.1093/annonc/7.suppl_4.S5Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28vjtlCitw%253D%253D&md5=d36d53ffb750f23ed1a66914bbb7fb82Epidemiology of EBV and Hodgkin's lymphomaJarrett A F; Armstrong A A; Alexander EAnnals of oncology : official journal of the European Society for Medical Oncology (1996), 7 Suppl 4 (), 5-10 ISSN:0923-7534.The incidence of Hodgkin's disease shows marked heterogeneity with respect to age, gender, race, geographical locale, social class and histological subtype. The Epstein-Barr virus (EBV) is associated with a proportion of cases, approximately 40% in developed countries, and there is a body of evidence which suggests that mixed cellularity Hodgkin's disease is more likely to be EBV-associated than nodular sclerosis Hodgkin's disease. This study investigates the relationship between EBV and both age and histological subtype. We confirm the above subtype distribution and that childhood and older adult cases are more likely to be EBV-associated than young adult cases. Young adult nodular sclerosis cases are rarely EBV-associated providing further evidence that this is a separate disease entity. The results of this study support the multiple aetiology hypothesis which suggests that Hodgkin's disease in different age groups has different aetiologies. The epidemiology of Hodgkin's disease and studies investigating the distribution of EBV-associated cases are reviewed.
- 60Swerdlow, S. H.; Campo, E.; Pileri, S. A.; Harris, N. L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G. A.; Zelenetz, A. D.; Jaffe, E. S. The 2016 Revision of the World Health Organization Classification of Lymphoid Neoplasms. Blood 2016, 127 (20), 2375– 2390, DOI: 10.1182/blood-2016-01-643569Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXjs1ans74%253D&md5=3605196d3e7b33bbad6f780f3a0e06b2The 2016 revision of the World Health Organization classification of lymphoid neoplasmsSwerdlow, Steven H.; Campo, Elias; Pileri, Stefano A.; Harris, Nancy Lee; Stein, Harald; Siebert, Reiner; Advani, Ranjana; Ghielmini, Michele; Salles, Gilles A.; Zelenetz, Andrew D.; Jaffe, Elaine S.Blood (2016), 127 (20), 2375-2390CODEN: BLOOAW; ISSN:1528-0020. (American Society of Hematology)A revision of the nearly 8-yr-old World Health Organization classification of the lymphoid neoplasms and the accompanying monograph is being published. It reflects a consensus among hematopathologists, geneticists, and clinicians regarding both updates to current entities as well as the addn. of a limited no. of new provisional entities. The revision clarifies the diagnosis and management of lesions at the very early stages of lymphomagenesis, refines the diagnostic criteria for some entities, details the expanding genetic/mol. landscape of numerous lymphoid neoplasms and their clin. correlates, and refers to investigations leading to more targeted therapeutic strategies. The major changes are reviewed with an emphasis on the most important advances in our understanding that impact our diagnostic approach, clin. expectations, and therapeutic strategies for the lymphoid neoplasms.
- 61Hjalgrim, H.; Friborg, J.; Melbye, M. The Epidemiology of EBV and Its Association with Malignant Disease. Hum. Herpesviruses Biol. Ther. Immunoprophyl. 2007, 929– 959, DOI: 10.1017/CBO9780511545313.054Google ScholarThere is no corresponding record for this reference.
- 62Aalto, S. M.; Juvonen, E.; Tarkkanen, J.; Volin, L.; Haario, H.; Ruutu, T.; Hedman, K. Epstein-Barr Viral Load and Disease Prediction in a Large Cohort of Allogeneic Stem Cell Transplant Recipients. Clin. Infect. Dis. 2007, 45 (10), 1305– 1309, DOI: 10.1086/522531Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2snksVWitg%253D%253D&md5=c371ae17ddaedc8d956de51ff8c2b85cEpstein-Barr viral load and disease prediction in a large cohort of allogeneic stem cell transplant recipientsAalto S M; Juvonen E; Tarkkanen J; Volin L; Haario H; Ruutu T; Hedman KClinical infectious diseases : an official publication of the Infectious Diseases Society of America (2007), 45 (10), 1305-9 ISSN:.BACKGROUND: We wanted to determine the clinical significance and predictability of Epstein-Barr virus (EBV) infections among a large cohort of recipients of allogeneic, unselected stem cell transplants. METHODS: During 1988-1999, a total of 5479 consecutive serum samples obtained during 406 transplantations performed in Helsinki, Finland, were retrospectively analyzed by quantitative polymerase chain reaction for the presence of EBV DNA. RESULTS: Overall, EBV DNA was noted in at least 1 serum sample for 57 patients (14.0%), of whom 22 (5.4%) were found to have progressively increasing and ultimately high (>50,000 copies/mL) EBV DNA levels (median level, 179,000 copies/mL). In addition, 16 patients (4.0%) had low EBV DNA levels (median level, 3260 copies/mL) in isolated sera before death. Among the transplant recipients who survived, transient EBV DNAemia (median level, 3110 copies/mL), which apparently corresponded to asymptomatic EBV infection, was noted in 19 patients (4.7%). CONCLUSIONS: Low-level EBV DNA positivity in serum occurs relatively frequently after stem cell transplantation and may subside without specific treatment. However, high EBV DNA levels (i.e., >50,000 copies/mL) are strong predictors for the development of posttransplantation lymphoproliferative disease, are not spontaneously reversible, and should be treated immediately. If the EBV DNA level is >or=50,000 copies/mL, the patient can be classified as having life-threatening EBV infection.
- 63Dunmire, S. K.; Grimm, J. M.; Schmeling, D. O.; Balfour, H. H.; Hogquist, K. A. The Incubation Period of Primary Epstein-Barr Virus Infection: Viral Dynamics and Immunologic Events. PLoS Pathog. 2015, 11 (12), e1005286, DOI: 10.1371/journal.ppat.1005286Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFSktbw%253D&md5=d191dd6b3d95be64b81478acc210bcfcThe incubation period of primary epstein-barr virus infection: viral dynamics and immunologic eventsDunmire, Samantha K.; Grimm, Jennifer M.; Schmeling, David O.; Balfour, Henry H., Jr.; Hogquist, Kristin A.PLoS Pathogens (2015), 11 (12), e1005286/1-e1005286/18CODEN: PPLACN; ISSN:1553-7374. (Public Library of Science)Epstein-Barr virus (EBV) is a human herpesvirus that causes acute infectious mononucleosis and is assocd. with cancer and autoimmune disease. While many studies have been performed examg. acute disease in adults following primary infection, little is known about the virol. and immunol. events during EBV's lengthy 6 wk incubation period owing to the challenge of collecting samples from this stage of infection. We conducted a prospective study in college students with special emphasis on frequent screening to capture blood and oral wash samples during the incubation period. Here we describe the viral dissemination and immune response in the 6 wk prior to onset of acute infectious mononucleosis symptoms. While virus is presumed to be present in the oral cavity from time of transmission, we did not detect viral genomes in the oral wash until one week before symptom onset, at which time viral genomes were present in high copy nos., suggesting loss of initial viral replication control. In contrast, using a sensitive nested PCR method, we detected viral genomes at low levels in blood about 3 wk before symptoms. However, high levels of EBV in the blood were only obsd. close to symptom onset-coincident with or just after increased viral detection in the oral cavity. These data imply that B cells are the major reservoir of virus in the oral cavity prior to infectious mononucleosis. The early presence of viral genomes in the blood, even at low levels, correlated with a striking decrease in the no. of circulating plasmacytoid dendritic cells well before symptom onset, which remained depressed throughout convalescence. On the other hand, natural killer cells expanded only after symptom onset. Likewise, CD4+ Foxp3+ regulatory T cells decreased two fold, but only after symptom onset. We obsd. no substantial virus specific CD8 T cell expansion during the incubation period, although polyclonal CD8 activation was detected in concert with viral genomes increasing in the blood and oral cavity, possibly due to a systemic type I interferon response. This study provides the first description of events during the incubation period of natural EBV infection in humans and definitive data upon which to formulate theories of viral control and disease pathogenesis.
- 64Balfour, H. H.; Dunmire, S. K.; Hogquist, K. A. Infectious Mononucleosis. Clin. Transl. Immunol. 2015, 4 (2), e33 DOI: 10.1038/cti.2015.1Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXktVWjsrg%253D&md5=e6c3e6d3d0e9eae954d312c5238cc66cInfectious mononucleosisBalfour, Henry H., Jr.; Dunmire, Samantha K.; Hogquist, Kristin A.Clinical and Translational Immunology (2015), 4 (), e33CODEN: CTILBR; ISSN:2050-0068. (NPG Nature Asia-Pacific)Infectious mononucleosis is a clin. entity characterized by pharyngitis, cervical lymph node enlargement, fatigue and fever, which results most often from a primary Epstein-Barr virus (EBV) infection. EBV, a lymphocrytovirus and a member of the γ-herpesvirus family, infects at least 90% of the population worldwide, the majority of whom have no recognizable illness. The virus is spread by intimate oral contact among adolescents, but how preadolescents acquire the virus is not known. During the incubation period of approx. 6 wk, viral replication first occurs in the oropharynx followed by viremia as early as 2 wk before onset of illness. The acute illness is marked by high viral loads in both the oral cavity and blood accompanied by the prodn. of IgM antibodies against EBV viral capsid antigen and an extraordinary expansion of CD8+ T lymphocytes directed against EBV-infected B cells. During convalescence, CD8+ T cells return to normal levels and antibodies develop against EBV nuclear antigen-1. A typical clin. picture in an adolescent or young adult with a pos. heterophile test is usually sufficient to make the diagnosis of infectious mononucleosis, but heterophile antibodies are not specific and do not develop in some patients esp. young children. EBV-specific antibody profiles are the best choice for staging EBV infection. In addn. to causing acute illness, long-term consequences are linked to infectious mononucleosis, esp. Hodgkin lymphoma and multiple sclerosis. There is no licensed vaccine for prevention and no specific approved treatment. Future research goals are development of an EBV vaccine, understanding the risk factors for severity of the acute illness and likelihood of developing cancer or autoimmune diseases, and discovering anti-EBV drugs to treat infectious mononucleosis and other EBV-spurred diseases.
- 65Pannone, G.; Zamparese, R.; Pace, M.; Pedicillo, M. C.; Cagiano, S.; Somma, P.; Errico, M. E.; Donofrio, V.; Franco, R.; De Chiara, A.; Aquino, G.; Bucci, P.; Bucci, E.; Santoro, A.; Bufo, P. The Role of EBV in the Pathogenesis of Burkitt’s Lymphoma: An Italian Hospital Based Survey. Infect. Agent. Cancer 2014, 9 (1), 34, DOI: 10.1186/1750-9378-9-34Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmsVCjtw%253D%253D&md5=694ac0af2e67db0a7751da372c4edec2The role of EBV in the pathogenesis of Burkitt's Lymphoma: an Italian hospital based surveyPannone, Giuseppe; Zamparese, Rosanna; Pace, Mirella; Pedicillo, Maria Carmela; Cagiano, Simona; Somma, Pasquale; Errico, Maria Elena; Donofrio, Vittoria; Franco, Renato; De Chiara, Annarosaria; Aquino, Gabriella; Bucci, Paolo; Bucci, Eduardo; Santoro, Angela; Bufo, PantaleoInfectious Agents and Cancer (2014), 9 (), 34/1-34/11, 11 pp.CODEN: IACNB4; ISSN:1750-9378. (BioMed Central Ltd.)The exact worldwide incidence of Burkitt's lymphoma is not known. There are three distinct clin. variants of Burkitt's lymphoma, each manifesting differences in epidemiol., clin. presentation, morphol., biol. and genetic features: the endemic (African), the sporadic (non-endemic), and the immunodeficiency-assocd. form. In particular, we reported data regarding Burkitt's lymphoma incidence in the world and across different European countries. Finally, we described clinic-pathol. data of 48 Burkitt's lymphomas occurred in Italy from 2003 to 2013, in 4 different hospitals, two of which located in east side, and the other ones located in the west-coast. Forty Burkitt's lymphomas occurs in children (age range 3-12), and 8 were adulthood Burkitt's lymphomas (age range 18-87). In the pediatric group the Male:Female ratio (M:F) was of 4:1, whereas the group of the adult patients has a M:F of 1:1.67. Immunohistochem. detection of Latent Membrane Protein 1 (LMP1) expression and Epstein-Barr virus Encoded RNA (EBER) In Situ Hybridization (ISH) procedures have been performed. Lymphocyte B monoclonal spread has been demonstrated using a Polymerase Chain Reaction (PCR) based method to amplify Fragment Restriction FR1, FR2 and FR3 Ig heavy chains DNA fragments. Only 38 cases out of 48 were analyzed for LMP-1 showing various percentage of stained cells in 47.4% of the patients. Considering ISH for EBER detection results: - 1 out 2 (50%) adult analyzed cases was pos., with 50% of stained tumor cells (this patient was a 22 years old female, coming from Napoli); - 15 out 24 (62.5%) children analyzed Burkitt's lymphomas resulted as pos. for EBER; - the overall positivity has been obsd. in 16/26 Burkitt's lymphomas (61.53%). - Finally, EBV has been detected in children and adult patients, one of them with deregulation of the oncogene c-MYC by chromosomal translocation.
- 66Pavlovic, A.; Glavina Durdov, M.; Capkun, V.; Jakelic Pitesa, J.; Bozic Sakic, M. Classical Hodgkin Lymphoma with Positive Epstein-Barr Virus Status Is Associated with More FOXP3 Regulatory T Cells. Med. Sci. Monit. 2016, 22, 2340, DOI: 10.12659/MSM.896629Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosVSnsrY%253D&md5=1a0ad7543b7053f7868ed5f231094a91Classical Hodgkin lymphoma with positive epstein-barr virus status is associated with more FOXP3 regulatory T cellsPavlovic, Antonia; Durdov, Merica Glavina; Capkun, Vesna; Pitesa, Jasminka Jakelic; Sakic, Maja BozicMedical Science Monitor (2016), 22 (), 2340-2346CODEN: MSMOFR; ISSN:1643-3750. (International Scientific Information, Inc.)Background: Classical Hodgkin lymphoma (cHL) is characterized by sparse malignant Hodgkin and Reed-Sternberg cells dispersed in an inflammatory microenvironment. Immune evasion of malignant cells is partially due to the existence of a subpopulation of immunosuppressive regulatory T cells (Treg). The aim of this study was to analyze T cell compn. in cHL with special emphasis on Treg in regard to Epstein-Barr virus (EBV) status, subtype, and patient age. Material/Methods: The study included 102 patients with cHL diagnosed during a 12-yr period. EBV status of cHL was assessed immunohistochem. using antibodies directed to the EBV- encoded LMP1. To define T lymphocyte populations, slides were double-stained with FOXP3 for Treg, and CD4 or CD8 for T cells. In each case the no. of single- and/or double-pos. cells was counted on an image analyzer in 10 high-power fields. Statistical anal. was performed and differences were considered significant at P<0.05. Results: EBV-pos. status of cHL was confirmed in 30 (29%) cases, mainly in patients older than 54 years and in mixed cellularity subtype. In EBV-pos. cHL, higher nos. of CD8+ cells were found. In cHL with pos. EBV status, more FOXP3+ Treg were found, as well as higher nos. of FOXP3+CD4+ Treg compared with EBV-neg. cHL. The no. of CD4+ cells decreased with age. The frequency of FOXP3+CD8+ Treg was variable, without a statistically significant assocn. with age or EBV status. Conclusions: EBV status has an impact on compn. of T cell populations in the cHL microenvironment.
- 67Carbone, A.; Gloghini, A.; Dotti, G. EBV-Associated Lymphoproliferative Disorders: Classification and Treatment. Oncologist 2008, 13 (5), 577– 585, DOI: 10.1634/theoncologist.2008-0036Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1czltFaqtg%253D%253D&md5=bc426b942d555393aab2dcb70d9ee95aEBV-associated lymphoproliferative disorders: classification and treatmentCarbone Antonino; Gloghini Annunziata; Dotti GiampietroThe oncologist (2008), 13 (5), 577-85 ISSN:1083-7159.Since its discovery as the first human tumor virus, Epstein-Barr virus (EBV) has been implicated in the development of a wide range of B-cell lymphoproliferative disorders, including Burkitt's lymphoma, classic Hodgkin's lymphoma, and lymphomas arising in immunocompromised individuals (post-transplant and HIV-associated lymphoproliferative disorders). T-cell lymphoproliferative disorders that have been reported to be EBV associated include a subset of peripheral T-cell lymphomas, angioimmunoblastic T-cell lymphoma, extranodal nasal type natural killer/T-cell lymphoma, and other rare histotypes. EBV encodes a series of products interacting with or exhibiting homology to a wide variety of antiapoptotic molecules, cytokines, and signal transducers, hence promoting EBV infection, immortalization, and transformation. However, the exact mechanism by which EBV promotes oncogenesis is an area of active debate. The focus of this review is on the pathology, diagnosis, classification, and pathogenesis of EBV-associated lymphomas. Recent advances in EBV cell-based immunotherapy, which is beginning to show promise in the treatment of EBV-related disorders, are discussed.
- 68Cai, Q.; Chen, K.; Young, K. H. Epstein-Barr Virus-Positive T/NK-Cell Lymphoproliferative Disorders. Exp. Mol. Med. 2015, 47 (1), e133, DOI: 10.1038/emm.2014.105Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVartrw%253D&md5=3bf293fdd2fe625051ab52609b21b03fEpstein-Barr virus-positive T/NK-cell lymphoproliferative disordersCai, Qingqing; Chen, Kailin; Young, Ken H.Experimental & Molecular Medicine (2015), 47 (1), e133CODEN: EMMEF3; ISSN:2092-6413. (NPG Nature Asia-Pacific)A review. Epstein-Barr virus, a ubiquitous human herpesvirus, can induce both lytic and latent infections that result in a variety of human diseases, including lymphoproliferative disorders. The oncogenic potential of Epstein-Barr virus is related to its ability to infect and transform B lymphocytes into continuously proliferating lymphoblastoid cells. However, Epstein-Barr virus has also been implicated in the development of T/natural killer cell lymphoproliferative diseases. Epstein-Barr virus encodes a series of products that mimic several growth, transcription and anti-apoptotic factors, thus usurping control of pathways that regulate diverse homeostatic cellular functions and the microenvironment. However, the exact mechanism by which Epstein-Barr virus promotes oncogenesis and inflammatory lesion development remains unclear. Epstein-Barr virus-assocd. T/natural killer cell lymphoproliferative diseases often have overlapping clin. symptoms as well as histol. and immunophenotypic features because both lymphoid cell types derive from a common precursor. Accurate classification of Epstein-Barr virus-assocd. T/natural killer cell lymphoproliferative diseases is a prerequisite for appropriate clin. management. Currently, the treatment of most T/natural killer cell lymphoproliferative diseases is less than satisfactory. Novel and targeted therapies are strongly required to satisfy clin. demands. This review describes our current knowledge of the genetics, oncogenesis, biol., diagnosis and treatment of Epstein-Barr virus-assocd. T/natural killer cell lymphoproliferative diseases.
- 69Jácome, A. A. dos A.; de Lima, E. M.; Kazzi, A. I.; Chaves, G. F.; de Mendonça, D. C.; Maciel, M. M.; dos Santos, J. S. Epstein-Barr Virus-Positive Gastric Cancer: A Distinct Molecular Subtype of the Disease?. Rev. Soc. Bras. Med. Trop. 2016, 49 (2), 150– 157, DOI: 10.1590/0037-8682-0270-2015Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2s%252FgvFKgsw%253D%253D&md5=4564912c8c5ace704fa3e6c95708a626Epstein-Barr virus-positive gastric cancer: a distinct molecular subtype of the disease?Jacome Alexandre Andrade Dos Anjos; Lima Enaldo Melo de; Kazzi Ana Izabela; Chaves Gabriela Freitas; Mendonca Diego Cavalheiro de; Maciel Marina Mara; Santos Jose Sebastiao DosRevista da Sociedade Brasileira de Medicina Tropical (2016), 49 (2), 150-7 ISSN:.Approximately 90% of the world population is infected by Epstein-Barr virus (EBV). Usually, it infects B lymphocytes, predisposing them to malignant transformation. Infection of epithelial cells occurs rarely, and it is estimated that about to 10% of gastric cancer patients harbor EBV in their malignant cells. Given that gastric cancer is the third leading cause of cancer-related mortality worldwide, with a global annual incidence of over 950,000 cases, EBV-positive gastric cancer is the largest group of EBV-associated malignancies. Based on gene expression profile studies, gastric cancer was recently categorized into four subtypes; EBV-positive, microsatellite unstable, genomically stable and chromosomal instability. Together with previous studies, this report provided a more detailed molecular characterization of gastric cancer, demonstrating that EBV-positive gastric cancer is a distinct molecular subtype of the disease, with unique genetic and epigenetic abnormalities, reflected in a specific phenotype. The recognition of characteristic molecular alterations in gastric cancer allows the identification of molecular pathways involved in cell proliferation and survival, with the potential to identify therapeutic targets. These findings highlight the enormous heterogeneity of gastric cancer, and the complex interplay between genetic and epigenetic alterations in the disease, and provide a roadmap to implementation of genome-guided personalized therapy in gastric cancer. The present review discusses the initial studies describing EBV-positive gastric cancer as a distinct clinical entity, presents recently described genetic and epigenetic alterations, and considers potential therapeutic insights derived from the recognition of this new molecular subtype of gastric adenocarcinoma.
- 70Petrara, M. R.; Giunco, S.; Serraino, D.; Dolcetti, R.; De Rossi, A. Post-Transplant Lymphoproliferative Disorders: From Epidemiology to Pathogenesis-Driven Treatment. Cancer Lett. 2015, 369 (1), 37– 44, DOI: 10.1016/j.canlet.2015.08.007Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtlGku7zO&md5=fe946d61d717effb1341753581de6376Post-transplant lymphoproliferative disorders: From epidemiology to pathogenesis-driven treatmentPetrara, Maria Raffaella; Giunco, Silvia; Serraino, Diego; Dolcetti, Riccardo; De Rossi, AnitaCancer Letters (New York, NY, United States) (2015), 369 (1), 37-44CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)Post-transplant lymphoproliferative disorders (PTLDs) represent the most severe complication of both solid organ and hematopoietic stem cell transplantation. The Epstein-Barr Virus (EBV) is the main driver of PTLD, particularly those occurring early after transplantation. EBV-driven malignancies are assocd. with selective expression of latent viral proteins, but uncontrolled lytic replication may favor early phases of cell transformation. Besides immunodepression, persistent immune activation and chronic inflammation play an important role in both virus reactivation and expansion of EBV-infected B cells. EBV-induced immortalization requires the expression of telomerase. TERT, the rate-limiting component of the telomerase complex, is central in the switch from the lytic to the latent viral program, and TERT inhibition induces the EBV lytic cycle and cell death. Immunotherapy and combination of EBV lytic cycle inducers with antiviral drugs are promising strategies to improve the treatment of PTLD patients. This review is aimed at providing an update on the intriguing assocn. between EBV and PTLD, mainly focusing on cases arising after kidney and liver transplantation, which account for the vast majority of transplants.
- 71Jaffe, E. S.; Nicolae, A.; Pittaluga, S. Peripheral T-Cell and NK-Cell Lymphomas in the WHO Classification: Pearls and Pitfalls. Mod. Pathol. 2013 261 2013, 26 (1), S71– S87, DOI: 10.1038/modpathol.2012.181Google ScholarThere is no corresponding record for this reference.
- 72Tse, E.; Kwong, Y. L. The Diagnosis and Management of NK/T-Cell Lymphomas. J. Hematol. Oncol. 2017, 10 (1), 1– 13, DOI: 10.1186/s13045-017-0452-9Google ScholarThere is no corresponding record for this reference.
- 73Sivachandran, N.; Dawson, C. W.; Young, L. S.; Liu, F.-F.; Middeldorp, J.; Frappier, L. Contributions of the Epstein-Barr Virus EBNA1 Protein to Gastric Carcinoma. J. Virol. 2012, 86 (1), 60– 68, DOI: 10.1128/JVI.05623-11Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmsVKluro%253D&md5=48ef3aefc38d5b22ead7a42f2d98a3c2Contributions of the Epstein-Barr virus EBNA1 protein to gastric carcinomaSivachandran, Nirojini; Dawson, Christopher W.; Young, Lawrence S.; Liu, Fei-Fei; Middeldorp, Jaap; Frappier, LoriJournal of Virology (2012), 86 (1), 60-68CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Approx. 10% of gastric carcinomas (GC) are comprised of cells latently infected with Epstein-Barr virus (EBV); however, the mechanism by which EBV contributes to the development of this malignancy is unclear. We have investigated the cellular effects of the only EBV nuclear protein expressed in GC, EBNA1, focusing on promyelocytic leukemia (PML) nuclear bodies (NBs), which play important roles in apoptosis, p53 activation, and tumor suppression. AGS GC cells infected with EBV were found to contain fewer PML NBs and less PML protein than the parental EBV-neg. AGS cells, and these levels were restored by silencing EBNA1. Conversely, EBNA1 expression was sufficient to induce the loss of PML NBs and proteins in AGS cells. Consistent with PML functions, EBNA1 expression decreased p53 activation and apoptosis in response to DNA damage and resulted in increased cell survival. In addn., EBNA1 mutants unable to bind CK2 kinase or ubiquitin-specific protease 7 had decreased ability to induce PML loss and to interfere with p53 activation. PML levels in EBV-pos. and EBV-neg. GC biopsy specimens were then compared by immunohistochem. Consistent with the results in the AGS cells, EBV-pos. tumors had significantly lower PML levels than EBV-neg. tumors. The results indicate that EBV infection of GC cells leads to loss of PML NBs through the action of EBNA1, resulting in impaired responses to DNA damage and promotion of cell survival. Therefore, PML disruption by EBNA1 is one mechanism by which EBV may contribute to the development of gastric cancer.
- 74Chang, M. S.; Kim, D. H.; Roh, J. K.; Middeldorp, J. M.; Kim, Y. S.; Kim, S.; Han, S.; Kim, C. W.; Lee, B. L.; Kim, W. H.; Woo, J. H. Epstein-Barr Virus-Encoded BARF1 Promotes Proliferation of Gastric Carcinoma Cells through Regulation of NF-ΚB. J. Virol. 2013, 87 (19), 10515– 10523, DOI: 10.1128/JVI.00955-13Google Scholar74https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVygtLzI&md5=68480afa3d2c157ebc6b10da9a6c8101Epstein-Barr virus-encoded BARF1 promotes proliferation of gastric carcinoma cells through regulation of NF-κBChang, Mee Soo; Kim, Dong Ha; Roh, Jin Kyung; Middeldorp, Jaap M.; Kim, Yang Soo; Kim, Sunghan; Han, Seungbong; Kim, Chul Woo; Lee, Byung Lan; Kim, Woo Ho; Woo, Jun HeeJournal of Virology (2013), 87 (19), 10515-10523CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)In Epstein-Barr virus (EBV)-infected gastric carcinoma, EBV-encoded BARF1 has been hypothesized to function as an oncogene. To evaluate cellular changes induced by BARF1, we isolated the full-length BARF1 gene from gastric carcinoma cells that were naturally infected with EBV and transfected BARF1 into EBV-neg. gastric carcinoma cells. BARF1 protein was primarily secreted into culture supernatant and only marginally detectable within cells. Compared with gastric carcinoma cells contg. empty vector, BARF1-expressing gastric carcinoma cells exhibited increased cell proliferation (P < 0.05). There were no significant differences in apoptosis, invasion, or migration between BARF1-expressing gastric carcinoma cells and empty vector-transfected cells. BARF1-expressing gastric carcinoma cells demonstrated increased nuclear expression of nuclear factor kappa B (NF-κB) RelA protein and increased NF-κB-dependent cyclin D1. The expression of p21WAF1 was diminished by BARF1 transfection and increased by NF-κB inhibition. Proliferation of naturally EBV-infected gastric carcinoma cells was suppressed by BARF1 small interfering RNA (siRNA) (P < 0.05). Immunohistochem. anal. of 120 human gastric carcinoma tissues demonstrated increased expression of cyclin D1 and reduced expression of p21WAF1 in EBV-pos. samples vs. EBV-neg. gastric carcinomas (P < 0.05). In conclusion, the secreted BARF1 may stimulate proliferation of EBV-infected gastric carcinoma cells via upregulation of NF-κB/cyclin D1 and redn. of the cell cycle inhibitor p21WAF1, thereby facilitating EBV-induced cancer progression.
- 75Breda, E.; Catarino, R. J. F.; Azevedo, I.; Lobão, M.; Monteiro, E.; Medeiros, R. Epstein-Barr Virus Detection in Nasopharyngeal Carcinoma: Implications in a Low-Risk Area. Braz. J. Otorhinolaryngol. 2010, 76 (3), 310– 315, DOI: 10.1590/S1808-86942010000300007Google Scholar75https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cnpvFarsw%253D%253D&md5=b344cdf57ed536ee2facc8811680824bEpstein-Barr virus detection in nasopharyngeal carcinoma: implications in a low-risk areaBreda Eduardo; Catarino Raquel Jorge Ferreira; Azevedo Isabel; Lobao Marisa; Monteiro Eurico; Medeiros RuiBrazilian journal of otorhinolaryngology (2010), 76 (3), 310-5 ISSN:.UNLABELLED: Several studies have been published concerning Epstein-barr virus (EBV) infection and nasopharyngeal cancer (NPC) development. The incidences of histological types are different according to endemic or non-endemic regions. Latent EBV infection is found in almost all cases of NPC in endemic regions, but normally absent in type I carcinomas, more common in non-endemic regions. AIM: The purpose of this hospital-based study was to analyze the presence of EBV in nasopharyngeal tumor tissues and in peripheral blood of nasopharyngeal cancer patients and healthy individuals, in a low risk, non-endemic area. METHODS: EBV detection in samples of nasopharyngeal cancer patients and healthy individuals. RESULTS: This study indicates that the frequency of EBV positive cases in peripheral blood is higher in advanced tumor stages. CONCLUSIONS: The incidence rates of NPC have a distinct distribution. Since the prevalence of this disease is low in occidental countries, little is known about the biology of these tumors in non-endemic areas. We observed statistically significant differences in EBV detection between the NPC patient group and the control group. This study may help to understand the biological mechanisms of NPC and the correlation of EBV infection with this disease, in a low risk, non-endemic region.
- 76Chu, E. A.; Wu, J. M.; Tunkel, D. E.; Ishman, S. L. Nasopharyngeal Carcinoma: The Role of the Epstein-Barr Virus. Medscape J. Med. 2008, 10 (7), 165Google Scholar76https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1crotVylug%253D%253D&md5=fd1cba411c6d6efeac8fabbc4f817afaNasopharyngeal carcinoma: the role of the Epstein-Barr virusChu Eugene A; Wu Julie M; Tunkel David E; Ishman Stacey LMedscape journal of medicine (2008), 10 (7), 165 ISSN:.Nasopharyngeal carcinoma (NPC) arises from the epithelium of the nasopharynx and is a rare tumor in most parts of the world, including the United States. Neck swelling, nasal obstruction, and epistaxis are the most common presenting symptoms. The etiology of NPC is multifactorial and includes genetic susceptibility, exposure to carcinogens, and prior infection with the Epstein-Barr virus (EBV). We report a case of a 16-year-old African-American male who presented with hemoptysis and a 3-month history of a neck mass. Diagnostic evaluation identified a nasopharyngeal mass that upon biopsy was shown to be an undifferentiated nasopharyngeal carcinoma with immunohistochemical stains markedly positive for EBV. Recent studies have further elucidated the role of EBV in the pathogenesis of NPC and have demonstrated the utility of EBV studies in staging, prognosis, and post-therapeutic monitoring.
- 77Shinozaki-Ushiku, A.; Kunita, A.; Fukayama, M. Update on Epstein-Barr Virus and Gastric Cancer (Review). Int. J. Oncol. 2015, 46 (4), 1421– 1434, DOI: 10.3892/ijo.2015.2856Google Scholar77https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFGnurfO&md5=c9b0cabb6c7d355837cf0a1485bdd1faUpdate on Epstein-Barr virus and gastric cancer (Review)Shinozaki-Ushiku, Aya; Kunita, Akiko; Fukayama, MasashiInternational Journal of Oncology (2015), 46 (4), 1421-1434CODEN: IJONES; ISSN:1791-2423. (Spandidos Publications Ltd.)Epstein-Barr virus-assocd. gastric carcinoma (EBVaGC) is a distinct subtype that accounts for nearly 10% of gastric carcinomas. EBVaGC is defined by monoclonal proliferation of carcinoma cells with latent EBV infection, as demonstrated by EBV-encoded small RNA (EBER) in situ hybridization. EBVaGC has characteristic clinicopathol. features, including predominance among males, a proximal location in the stomach, lymphoepithelioma-like histol. and a favorable prognosis. EBVaGC belongs to latency type I or II, in which EBERs, EBNA-1, BARTs, LMP-2A and BART miRNAs are expressed. Previous studies have shown that some EBV latent genes have oncogenic properties. Recent advances in genome-wide and comprehensive mol. analyses have demonstrated that both genetic and epigenetic changes contribute to EBVaGC carcinogenesis. Genetic changes that are characteristic of EBVaGC include frequent mutations in PIK3CA and ARID1A and amplification of JAK2 and PD-L1/L2. Global CpG island hypermethylation, which induces epigenetic silencing of tumor suppressor genes, is also a unique feature of EBVaGC and is considered to be crucial for its carcinogenesis. Furthermore, post-transcriptional gene expression regulation by cellular and/or EBV-derived microRNAs has attracted considerable attention. These abnormalities result in significant alterations in gene expression related to cell proliferation, apoptosis, migration and immune signaling pathways. In the present review we highlight the latest findings on EBVaGC from clinicopathol. and mol. perspectives to provide a better understanding of EBV involvement in gastric carcinogenesis.
- 78Fukuda, M.; Ikuta, K.; Yanagihara, K.; Tajima, M.; Kuratsune, H.; Kurata, T.; Sairenji, T. Effect of Transforming Growth Factor-Beta1 on the Cell Growth and Epstein-Barr Virus Reactivation in EBV-Infected Epithelial Cell Lines. Virology 2001, 288 (1), 109– 118, DOI: 10.1006/viro.2001.1071Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXmsFyhsL0%253D&md5=390f28e848bb8eeabf5336376a655650Effect of Transforming Growth Factor-β1 on the Cell Growth and Epstein-Barr Virus Reactivation in EBV-Infected Epithelial Cell LinesFukuda, Makoto; Ikuta, Kazufumi; Yanagihara, Kazuyoshi; Tajima, Masako; Kuratsune, Hirohiko; Kurata, Takeshi; Sairenji, TakeshiVirology (2001), 288 (1), 109-118CODEN: VIRLAX; ISSN:0042-6822. (Academic Press)Transforming growth factor (TGF)-β1 is a multi-functional cytokine that plays important roles in regulating cell growth and differentiation in many biol. systems. In this study, we found that gastric tissue-derived Epstein-Barr virus (EBV)-infected epithelial cell lines GT38 and GT39 had resistance to TGF-β1-mediated growth inhibition and apoptosis compared to a TGF-β1-susceptible gastric carcinoma cell line HSC-39. However, TGF-β1 partially induced EBV reactivation in GT38 and GT39 cells, as shown by the induction of EBV immediate-early BZLF1 RNA and its protein product ZEBRA and early antigen-D. The expressions of TGF-β receptor I and II were detected in GT38 and GT39 cells by Northern and Western blot analyses. Both cell lines spontaneously produced the TGF-β1, which was sufficient for inhibiting cell growth of HSC-39 cells. Taken together, these data suggest that TGF-β1 may be a key factor for EBV reactivation and selective growth of EBV-infected epithelial cells in vivo. (c) 2001 Academic Press.
- 79Hino, R.; Uozaki, H.; Inoue, Y.; Shintani, Y.; Ushiku, T.; Sakatani, T.; Takada, K.; Fukayama, M. Survival Advantage of EBV-Associated Gastric Carcinoma: Survivin up-Regulation by Viral Latent Membrane Protein 2A. Cancer Res. 2008, 68 (5), 1427– 1435, DOI: 10.1158/0008-5472.CAN-07-3027Google Scholar79https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXislagurY%253D&md5=d7225624e1760002c672df66469dabd9Survival Advantage of EBV-Associated Gastric Carcinoma: Survivin Up-regulation by Viral Latent Membrane Protein 2AHino, Rumi; Uozaki, Hiroshi; Inoue, Yoko; Shintani, Yukako; Ushiku, Tetsuo; Sakatani, Takashi; Takada, Kenzo; Fukayama, MasashiCancer Research (2008), 68 (5), 1427-1435CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)EBV-assocd. gastric carcinoma is a distinct subset of gastric carcinoma infected with EBV, which shows latency I type expression of EBV latent genes (EBNA1, EBER, BARF0, and LMP2A). To clarify the role of EBV in this type of gastric carcinoma, the cell biol. characteristics (growth, apoptosis, and migration) were evaluated in gastric carcinoma cell lines (MKN-1, TMK1, MKN-74 and MKN-7) with and without infection of recombinant EBV harboring the neomycin resistance gene. The infection reiterated the latency I type infection, and the only difference obsd. in EBV-infected gastric carcinoma cell lines was the resistance to serum deprivation-induced apoptosis. Comparative analyses of transcripts of apoptosis-assocd. genes in MKN-1 and EBV-MKN-1 and subsequent quant. reverse transcription-PCR anal. showed up-regulation of the cellular survivin gene in EBV-infected gastric carcinoma cell lines. Small interfering RNA-mediated knockdown of survivin increased apoptosis in EBV-MKN-1 to the level of the original MKN-1 cells. Transfection of EBV-latent genes into MKN-1 showed that LMP2A, but not EBNA1, EBER, or BARF0, up-regulated survivin gene expression. LMP2A-mediated survivin up-regulation in gastric carcinoma cells was inhibited with a nuclear factor-κB (NF-κB) inhibitor, Bay 11-7082. In parallel with these findings in vitro, survivin expression was frequent in carcinoma tissues of gastric carcinoma by immunohistochem., and significantly more in EBV-assocd. gastric carcinoma (12 of 13) than in EBV-neg. gastric carcinoma in the advanced stage (P = 0.0307). Thus, EBV uses its latent protein, LMP2A, to activate the NF-κB-survivin pathway to rescue EBV-infected epithelial cells from serum deprivation, and up-regulation of survivin may play a role in the progression of this specific type of gastric carcinoma infected with EBV.
- 80Liu, X.; Gao, Y.; Luo, B.; Zhao, Y. Construction and Antiapoptosis Activities of Recombinant Adenoviral Expression Vector Carrying EBV Latent Membrane Protein 2A. Gastroenterol. Res. Pract. 2011, 2011, 1, DOI: 10.1155/2011/182832Google ScholarThere is no corresponding record for this reference.
- 81Seeger, C.; Mason, W. S. Hepatitis B Virus Biology. Microbiol. Mol. Biol. Rev. 2000, 64 (1), 51– 68, DOI: 10.1128/MMBR.64.1.51-68.2000Google Scholar81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXitFygsr4%253D&md5=7882f1b23edb2f85b95e9ac953dbb731Hepatitis B virus biologySeeger, Christoph; Mason, William S.Microbiology and Molecular Biology Reviews (2000), 64 (1), 51-68CODEN: MMBRF7; ISSN:1092-2172. (American Society for Microbiology)A review with 255 refs.
- 82Woodman, C. B. J.; Collins, S. I.; Young, L. S. The Natural History of Cervical HPV Infection: Unresolved Issues. Nat. Rev. Cancer 2007, 7 (1), 11– 22, DOI: 10.1038/nrc2050Google Scholar82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtlCht7bN&md5=351ca65cb5ac1484ebc7f203d86c03c9The natural history of cervical HPV infection: unresolved issuesWoodman, Ciaran B. J.; Collins, Stuart I.; Young, Lawrence S.Nature Reviews Cancer (2007), 7 (1), 11-22CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)The identification of high-risk human papillomavirus (HPV) types as a necessary cause of cervical cancer offers the prospect of effective primary prevention and the possibility of improving the efficiency of cervical screening programs. However, for these opportunities to be realized, a more complete understanding of the natural history of HPV infection, and its relationship to the development of epithelial abnormalities of the cervix, is required. We discuss areas of uncertainty, and their possible effect on disease prevention strategies.
- 83Doorbar, J. Molecular Biology of Human Papillomavirus Infection and Cervical Cancer. Clin. Sci. (Lond). 2006, 110 (5), 525– 541, DOI: 10.1042/CS20050369Google Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD287pt1eqtA%253D%253D&md5=4cc03f6d2ee0a591a673b01627c63672Molecular biology of human papillomavirus infection and cervical cancerDoorbar JohnClinical science (London, England : 1979) (2006), 110 (5), 525-41 ISSN:0143-5221.HPVs (human papillomaviruses) infect epithelial cells and cause a variety of lesions ranging from common warts/verrucas to cervical neoplasia and cancer. Over 100 different HPV types have been identified so far, with a subset of these being classified as high risk. High-risk HPV DNA is found in almost all cervical cancers (>99.7%), with HPV16 being the most prevalent type in both low-grade disease and cervical neoplasia. Productive infection by high-risk HPV types is manifest as cervical flat warts or condyloma that shed infectious virions from their surface. Viral genomes are maintained as episomes in the basal layer, with viral gene expression being tightly controlled as the infected cells move towards the epithelial surface. The pattern of viral gene expression in low-grade cervical lesions resembles that seen in productive warts caused by other HPV types. High-grade neoplasia represents an abortive infection in which viral gene expression becomes deregulated, and the normal life cycle of the virus cannot be completed. Most cervical cancers arise within the cervical transformation zone at the squamous/columnar junction, and it has been suggested that this is a site where productive infection may be inefficiently supported. The high-risk E6 and E7 proteins drive cell proliferation through their association with PDZ domain proteins and Rb (retinoblastoma), and contribute to neoplastic progression, whereas E6-mediated p53 degradation prevents the normal repair of chance mutations in the cellular genome. Cancers usually arise in individuals who fail to resolve their infection and who retain oncogene expression for years or decades. In most individuals, immune regression eventually leads to clearance of the virus, or to its maintenance in a latent or asymptomatic state in the basal cells.
- 84Mesri, E. A.; Cesarman, E.; Boshoff, C. Kaposi’s Sarcoma and Its Associated Herpesvirus. Nat. Rev. Cancer 2010, 10 (10), 707– 719, DOI: 10.1038/nrc2888Google Scholar84https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtF2mtrjO&md5=746af8c2eca5fb9f7853c6b0f9b9d0fbKaposi's sarcoma and its associated herpesvirusMesri, Enrique A.; Cesarman, Ethel; Boshoff, ChrisNature Reviews Cancer (2010), 10 (10), 707-719CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Kaposi's sarcoma (KS) is the most common cancer in HIV-infected untreated individuals. Kaposi's sarcoma-assocd. herpesvirus (KSHV; also known as human herpesvirus 8 (HHV8)) is the infectious cause of this neoplasm. In this Review we describe the epidemiol. of KS and KSHV, and the insights into the remarkable mechanisms through which KSHV can induce KS that have been gained in the past 16 years. KSHV latent transcripts, such as latency-assocd. nuclear antigen (LANA), viral cyclin, viral FLIP and viral-encoded microRNAs, drive cell proliferation and prevent apoptosis, whereas KSHV lytic proteins, such as viral G protein-coupled receptor, K1 and virally encoded cytokines (viral interleukin-6 and viral chemokines) further contribute to the unique angioproliferative and inflammatory KS lesions through a mechanism called paracrine neoplasia.
- 85Young, L. S.; Rickinson, A. B. Epstein-Barr Virus: 40 Years On. Nat. Rev. Cancer 2004, 4 (10), 757– 768, DOI: 10.1038/nrc1452Google Scholar85https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXos1ymu78%253D&md5=8e74eae45a3ca16243922a65a5ecddd0Epstein-Barr virus: 40 years onYoung, Lawrence S.; Rickinson, Alan B.Nature Reviews Cancer (2004), 4 (10), 757-768CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. Epstein-Barr virus (EBV) was discovered 40 yr ago from examg. electron micrographs of cells cultured from Burkitt's lymphoma, a childhood tumor that is common in sub-Saharan Africa, where its unusual geog. distribution - which matches that of holoendemic malaria - indicated a viral etiol. However, far from showing a restricted distribution, EBV - a γ-herpesvirus - was found to be widespread in all human populations and to persist in the vast majority of individuals as a lifelong, asymptomatic infection of the B-lymphocyte pool. Despite such ubiquity, the link between EBV and 'endemic' Burkitt's lymphoma proved consistent and became the first of an unexpectedly wide range of assocns. discovered between this virus and tumors.
- 86Shi, G.; Suzuki, T. Molecular Basis of Encapsidation of Hepatitis C Virus Genome. Front. Microbiol. 2018, 9 (MAR), 396, DOI: 10.3389/fmicb.2018.00396Google Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MnjsFymtg%253D%253D&md5=263bdc99fec0cbff02dec071bf73f1f8Molecular Basis of Encapsidation of Hepatitis C Virus GenomeShi Guoli; Suzuki TetsuroFrontiers in microbiology (2018), 9 (), 396 ISSN:1664-302X.Hepatitis C virus (HCV), a major etiologic agent of human liver diseases, is a positive-sense single-stranded RNA virus and is classified in the Flaviviridae family. Although research findings for the assembly of HCV particles are accumulating due to development of HCV cell culture system, the mechanism(s) by which the HCV genome becomes encapsidated remains largely unclear. In general, viral RNA represents only a small fraction of the RNA molecules in the cells infected with RNA viruses, but the viral genomic RNA is considered to selectively packaged into virions. It was recently demonstrated that HCV RNAs containing 3' end of the genome are selectively incorporated into virus particles during the assembly process and the 3' untranslated region functions as a cis-acting element for RNA packaging. Here, we discuss the molecular basis of RNA encapsidation of HCV and classical flaviviruses, contrast with the packaging mechanism of HIV-1.
- 87Kannian, P.; Green, P. L. Human T Lymphotropic Virus Type 1 (HTLV-1): Molecular Biology and Oncogenesis. Viruses 2010, 2 (9), 2037, DOI: 10.3390/v2092037Google Scholar87https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXht1eksbrL&md5=46d1e0e5945a2246719b1d4108676c8cHuman T lymphotropic virus type 1 (HTLV-1): molecular biology and oncogenesisKannian, Priya; Green, Patrick L.Viruses (2010), 2 (), 2037-2077CODEN: VIRUBR; ISSN:1999-4915. (Molecular Diversity Preservation International)A review. Human T lymphotropic viruses (HTLVs) are complex deltaretroviruses that do not contain a proto-oncogene in their genome, yet are capable of transforming primary T lymphocytes both in vitro and in vivo. There are four known strains of HTLV including HTLV type 1 (HTLV-1), HTLV-2, HTLV-3 and HTLV-4. HTLV-1 is primarily assocd. with adult T cell leukemia (ATL) and HTLV-1-assocd. myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-2 is rarely pathogenic and is sporadically assocd. with neurol. disorders. There have been no diseases assocd. with HTLV-3 or HTLV-4 to date. Due to the difference in the disease manifestation between HTLV-1 and HTLV-2, a clear understanding of their individual pathobiologies and the role of various viral proteins in transformation should provide insights into better prognosis and prevention strategies. In this review, we aim to summarize the data accumulated so far in the transformation and pathogenesis of HTLV-1, focusing on the viral Tax and HBZ and citing appropriate comparisons to HTLV-2.
- 88Stevenson, M. HIV-1 Pathogenesis. Nat. Med. 2003, 9 (7), 853– 860, DOI: 10.1038/nm0703-853Google Scholar88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXkvFKqurg%253D&md5=075f6a49140f0eac9270d6ce0eb6ce9fHIV-1 pathogenesisStevenson, MarioNature Medicine (New York, NY, United States) (2003), 9 (7), 853-860CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)A review. Despite considerable advances in HIV science in the past 20 yr, the reason why HIV-1 infection is pathogenic is still debated and the goal of eradicating HIV-1 infection remains elusive. A deeper understanding of the interplay between HIV-1 and its host and why simian immunodeficiency virus (SIV) is nonpathogenic in some natural hosts may provide a few answers. Topics discussed include virus-host interactions at the mol. level; and elements of viral pathogenicity.
- 89Mui, U. N.; Haley, C. T.; Tyring, S. K. Viral Oncology: Molecular Biology and Pathogenesis. J. Clin. Med. 2017, 6 (12), 111, DOI: 10.3390/jcm6120111Google Scholar89https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmtVKru7Y%253D&md5=ce4b7919e386d30cd757d1cc11fc642aViral oncology: molecular biology and pathogenesisMui, Uyen Ngoc; Haley, Christopher T.; Tyring, Stephen K.Journal of Clinical Medicine (2017), 6 (12), 111/1-111/58CODEN: JCMOHK; ISSN:2077-0383. (MDPI AG)Oncoviruses are implicated in approx. 12% of all human cancers. A large no. of the world's population harbors at least one of these oncoviruses, but only a small proportion of these individuals go on to develop cancer. The interplay between host and viral factors is a complex process that works together to create a microenvironment conducive to oncogenesis. In this review, the mol. biol. and oncogenic pathways of established human oncoviruses will be discussed. Currently, there are seven recognized human oncoviruses, which include Epstein-Barr Virus (EBV), Human Papillomavirus (HPV), Hepatitis B and C viruses (HBV and HCV), Human T-cell lymphotropic virus-1 (HTLV-1), Human Herpesvirus-8 (HHV-8), and Merkel Cell Polyomavirus (MCPyV). Available and emerging therapies for these oncoviruses will be mentioned.
- 90Shuda, M.; Feng, H.; Kwun, H. J.; Rosen, S. T.; Gjoerup, O.; Moore, P. S.; Chang, Y. T Antigen Mutations Are a Human Tumor-Specific Signature for Merkel Cell Polyomavirus. Proc. Natl. Acad. Sci. U. S. A. 2008, 105 (42), 16272– 16277, DOI: 10.1073/pnas.0806526105Google Scholar90https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht12qtLrL&md5=422db1a8e1304be44b2fcb8fa0dc04beT antigen mutations are a human tumor-specific signature for Merkel cell polyomavirusShuda, Masahiro; Feng, Huichen; Kwun, Hyun Jin; Rosen, Steven T.; Gjoerup, Ole; Moore, Patrick S.; Chang, YuanProceedings of the National Academy of Sciences of the United States of America (2008), 105 (42), 16272-16277CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Merkel cell polyomavirus (MCV) is a virus discovered in our lab. at the University of Pittsburgh that is monoclonally integrated into the genome of ≈80% of human Merkel cell carcinomas (MCCs). Transcript mapping was performed to show that MCV expresses transcripts in MCCs similar to large T (LT), small T (ST), and 17kT transcripts of SV40. Nine MCC tumor-derived LT genomic sequences have been examd., and all were found to harbor mutations prematurely truncating the MCV LT helicase. In contrast, four presumed episomal viruses from nontumor sources did not possess this T antigen signature mutation. Using coimmunopptn. and origin replication assays, we show that tumor-derived virus mutations do not affect retinoblastoma tumor suppressor protein (Rb) binding by LT but do eliminate viral DNA replication capacity. Identification of an MCC cell line (MKL-1) having monoclonal MCV integration and the signature LT mutation allowed us to functionally test both tumor-derived and wild type (WT) T antigens. Only WT LT expression activates replication of integrated MCV DNA in MKL-1 cells. Our findings suggest that MCV-pos. MCC tumor cells undergo selection for LT mutations to prevent autoactivation of integrated virus replication that would be detrimental to cell survival. Because these mutations render the virus replication-incompetent, MCV is not a "passenger virus" that secondarily infects MCC tumors.
- 91Bodaghi, S.; Comoli, P.; Bösch, R.; Azzi, A.; Gosert, R.; Leuenberger, D.; Ginevri, F.; Hirsch, H. H. Antibody Responses to Recombinant Polyomavirus BK Large T and VP1 Proteins in Young Kidney Transplant Patients. J. Clin. Microbiol. 2009, 47 (8), 2577– 2585, DOI: 10.1128/JCM.00030-09Google Scholar91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVyhurnI&md5=c0e8adf72266554fdf560be8cd816057Antibody responses to recombinant polyomavirus BK large T and VP1 proteins in young kidney transplant patientsBodaghi, Sohrab; Comoli, Patrizia; Bosch, Robert; Azzi, Alberta; Gosert, Rainer; Leuenberger, David; Ginevri, Fabrizio; Hirsch, Hans H.Journal of Clinical Microbiology (2009), 47 (8), 2577-2585CODEN: JCMIDW; ISSN:0095-1137. (American Society for Microbiology)BK virus (BKV)-specific immunity is crit. for polyomavirus-assocd. nephropathy, but antibody responses are incompletely defined. We compared the hemagglutination inhibition assay (HIA) with IgG enzyme immunoassays (EIA) to BKV proteins expressed in baculovirus-infected insect cells. N-terminal, internal, and C-terminal domains of the BKV large T antigen (BKLT) were fused to glutathione S-transferase (GST), yielding GST-BKLTD1, GST-BKLTD2, and GST-BKLTD3, resp. The BKV capsid VP1 was expressed as a GST fusion (BKVP1) or as a native VP1 assembled into viruslike particles (BKVLP). We tested 422 sera from 28 healthy donors (HD), 99 dialysis patients (DP; median age, 15 years; range, 3 to 32 years), and 46 age-matched kidney transplant patients (KTP; median age, 15 years; range, 2 to 33 years). In HD, HIA and BKVLP EIA both yielded a 91.7% seroreactivity, whereas all other EIA responses were lower (BKVP1, 83.3%; BKLTD1, 25%; BKLTD2, 29%; BKLTD3, 40%). HIA titers significantly correlated with EIA levels for BKVLP, BKVP1, and BKLTD1 but not for BKLTD2 or BKLTD3, which were barely above the cutoff. In DP, the seroreactivities of HIA, BKVLP, and BKLTD1 were lower than that in HD (63.6%, 86.9%, and 10.1%, resp.) and they had lower titers (P < 0.001). In KTP, seropositivities for BKVLP, BKVP1, and BKLTD1 were 78%, 50%, and 17%, resp., but anti-BKVLP levels increased significantly in KTP with viruria and viremia, whereas anti-BKLTD1 levels increased after clearing sustained BKV viremia. In conclusion, anti-BKVLP is equiv. to HIA in HD but is more sensitive to det. the BKV serostatus in DP and KTP. In KTP, anti-BKVLP responds to recent BKV viruria and viremia, whereas anti-BKLTD1 may indicate emerging BKV-specific immune control.
- 92Tolstov, Y. L.; Pastrana, D. V.; Feng, H.; Becker, J. C.; Jenkins, F. J.; Moschos, S.; Chang, Y.; Buck, C. B.; Moore, P. S. Human Merkel Cell Polyomavirus Infection II. MCV Is a Common Human Infection That Can Be Detected by Conformational Capsid Epitope Immunoassays. Int. J. Cancer 2009, 125 (6), 1250, DOI: 10.1002/ijc.24509Google Scholar92https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXovVeltrg%253D&md5=ca2d0159574e54bb81192a3f093373b8Human Merkel cell polyomavirus infection II. MCV is a common human infection that can be detected by conformational capsid epitope immunoassaysTolstov, Yanis L.; Pastrana, Diana V.; Feng, Huichen; Becker, Juergen C.; Jenkins, Frank J.; Moschos, Stergios; Chang, Yuan; Buck, Christopher B.; Moore, Patrick S.International Journal of Cancer (2009), 125 (6), 1250-1256CODEN: IJCNAW; ISSN:0020-7136. (Wiley-Liss, Inc.)Merkel cell polyomavirus (MCV) is a newly-discovered human tumor virus found in ∼80% of Merkel cell carcinoma (MCC). The rate of MCV infection among persons without MCC is unknown. We developed a MCV virus-like particle (VLP) enzyme-linked immunoassay (EIA) that does not cross-react with human BK or murine polyomaviruses. Peptide mapping of the MCV VP1 gene and immunoblotting with denatured MCV VLP are less sensitive than the MCV EIA in detecting MCV antibodies suggesting antibody reactivity in this assay primarily targets conformational but not linear epitopes. Among MCC patients, all 21 (100%) patients tested with MCV-pos. tumors had high serum MCV IgG but not high MCV IgM levels. Only 3 of 6 (50%) MCC patients with MCV-neg. tumors were pos. for MCV antibodies. Sera from most adults, including 107 of 166 (64%) blood donors, 63 of 100 (63%) com. donors and 37 of 50 (74%) systemic lupus erythematosus patients, show evidence for prior MCV exposure. Age-specific MCV prevalence was detd. by examg. a cross-sectional distribution of 150 Langerhans cell histiocytosis (an unrelated neoplasm) patient sera. MCV prevalence increases from 50% among children age 15 years or younger to 80% among persons older than 50 years. We did not find evidence for vertical transmission among infants. Although past exposure to MCV is common among all adult groups, MCC patients have a markedly elevated MCV IgG response compared with control patients. Our study demonstrates that MCV is a widespread but previously unrecognized human infection. © 2009 UICC.
- 93Pastrana, D. V.; Tolstov, Y. L.; Becker, J. C.; Moore, P. S.; Chang, Y.; Buck, C. B. Quantitation of Human Seroresponsiveness to Merkel Cell Polyomavirus. PLoS Pathog. 2009, 5 (9), e1000578, DOI: 10.1371/journal.ppat.1000578Google ScholarThere is no corresponding record for this reference.
- 94Arora, R.; Chang, Y.; Moore, P. S. MCV and Merkel Cell Carcinoma: A Molecular Success Story. Curr. Opin. Virol. 2012, 2 (4), 489– 498, DOI: 10.1016/j.coviro.2012.05.007Google Scholar94https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtF2rur3N&md5=329b88f4e96ca333e68c409b18fbe41eMCV and Merkel cell carcinoma: a molecular success storyArora, Reety; Chang, Yuan; Moore, Patrick S.Current Opinion in Virology (2012), 2 (4), 489-498CODEN: COVUAF; ISSN:1879-6257. (Elsevier B. V.)A review. Merkel cell polyomavirus (MCV), discovered in 2008, is clonally integrated in ∼80% Merkel cell carcinoma (MCC). MCV is a common skin flora and initiates cancer in susceptible hosts only after it acquires a precise set of mutations that render it replication incompetent. Both MCV large and small T proteins promote cancer cell survival and proliferation. Large T targets pocket proteins regulating cell cycle transit while small T activates cap-dependent translation crit. for cancer cell growth. These findings already have led to new diagnostics and clin. trials to target MCV-induced survivin and to promote antitumor immunity. In four years, the cause, diagnosis and therapy for an intractable cancer has been changed due to the mol. discovery of MCV.
- 95Lewis, J. S.; Duncavage, E.; Klonowski, P. W. Oral Cavity Neuroendocrine Carcinoma: A Comparison Study with Cutaneous Merkel Cell Carcinoma and Other Mucosal Head and Neck Neuroendocrine Carcinomas. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2010, 110 (2), 209– 217, DOI: 10.1016/j.tripleo.2010.04.007Google Scholar95https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cjhvF2hsQ%253D%253D&md5=069a330cac006e60a0332d0c79df49c9Oral cavity neuroendocrine carcinoma: a comparison study with cutaneous Merkel cell carcinoma and other mucosal head and neck neuroendocrine carcinomasLewis James S Jr; Duncavage Eric; Klonowski Paul WOral surgery, oral medicine, oral pathology, oral radiology, and endodontics (2010), 110 (2), 209-17 ISSN:.OBJECTIVES: Published cases of oral high-grade neuroendocrine carcinoma (HGNEC) variably call the tumors Merkel cell carcinoma (MCC) or small-cell/high-grade neuroendocrine carcinoma. We studied cases of cutaneous MCC and mucosal HGNEC to better distinguish them and to better define oral cases. STUDY DESIGN: Twelve cutaneous MCC and 14 mucosal HGNEC cases were identified. We reviewed the hematoxylin-eosin (H&E) morphology and performed immunohistochemistry for cytokeratin 20 (CK20), thyroid transcription factor 1 (TTF-1), p63, neurofilament (NF), and achaete-scute homolog 1 (MASH-1). We also performed polymerase chain reaction (PCR) for the Merkel cell polyomavirus (MCPyV). RESULTS: By morphology and immunohistochemistry, MCC and HGNEC showed many differences. CK20, NF, and TTF-1 stains were the most discriminatory. MASH-1 was expressed by both MCC and HGNEC. MCPyV was present in MCC and absent in all HGNEC. The 2 oral cavity mucosal carcinomas segregated into MCC and HGNEC types, the former having the H&E nuclear morphology and classic dot-like perinuclear CK20 staining typically associated with MCC. CONCLUSIONS: Oral cavity neuroendocrine carcinoma can be segregated into MCC and small-cell/HGNEC types by morphology and CK20 immunohistochemistry. MCPyV was present by PCR in cutaneous MCC but was not found in mucosal HGNEC.
- 96Paik, J. Y.; Hall, G.; Clarkson, A.; Lee, L.; Toon, C.; Colebatch, A.; Chou, A.; Gill, A. J. Immunohistochemistry for Merkel Cell Polyomavirus Is Highly Specific but Not Sensitive for the Diagnosis of Merkel Cell Carcinoma in the Australian Population. Hum. Pathol. 2011, 42 (10), 1385– 1390, DOI: 10.1016/j.humpath.2010.12.013Google Scholar96https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3Mfmslaktg%253D%253D&md5=48a95673c3f3fbf569a9d39c8901c867Immunohistochemistry for Merkel cell polyomavirus is highly specific but not sensitive for the diagnosis of Merkel cell carcinoma in the Australian populationPaik Julie Y; Hall Geoffrey; Clarkson Adele; Lee Lianne; Toon Christopher; Colebatch Andrew; Chou Angela; Gill Anthony JHuman pathology (2011), 42 (10), 1385-90 ISSN:.Recent studies have demonstrated a high frequency of detection of Merkel cell polyomavirus in Merkel cell carcinoma. However, most of these studies are from European or North American centers that have relatively low sun exposure and may have a higher incidence of virus-driven oncogenesis compared with the highly sun-exposed but predominantly fair-skinned Australian population. We performed immunohistochemistry for Merkel cell polyomavirus on 104 cases of Merkel cell carcinoma and 74 cases of noncutaneous small cell-undifferentiated carcinoma from 3 major Australian centers. Nineteen (18.3%) cases of Merkel cell carcinoma showed positive staining for Merkel cell polyomavirus versus 1 (1.3%) of small cell-undifferentiated carcinoma. All 15 cases (14.3%) of Merkel cell carcinoma with areas of mixed squamous differentiation showed negative staining. We found positive staining in only 3 (7.7%) of 39 Merkel cell carcinoma from the head and neck (the most sun-exposed area) versus 16 (24.6%) of 65 of tumors from other sites (P < .05). Our findings support the concept of a Merkel cell polyomavirus-driven and a non-Merkel cell polyomavirus-driven (primarily sun-dependent) pathway in Merkel cell carcinoma carcinogenesis, with the latter being significantly more frequent in Australia and in mixed squamous-Merkel cell carcinoma (which is also more frequent in Australia). Although immunohistochemistry for Merkel cell polyomavirus seems to be highly specific in all populations, the low incidence of Merkel cell polyomavirus-positive Merkel cell carcinoma in a highly sun-exposed population limits its diagnostic utility in this setting.
- 97Lemos, B. D.; Storer, B. E.; Iyer, J. G.; Phillips, J. L.; Bichakjian, C. K.; Fang, L. C.; Johnson, T. M.; Liegeois-Kwon, N. J.; Otley, C. C.; Paulson, K. G.; Ross, M. I.; Yu, S. S.; Zeitouni, N. C.; Byrd, D. R.; Sondak, V. K.; Gershenwald, J. E.; Sober, A. J.; Nghiem, P. Pathologic Nodal Evaluation Improves Prognostic Accuracy in Merkel Cell Carcinoma: Analysis of 5823 Cases as the Basis of the First Consensus Staging System. J. Am. Acad. Dermatol. 2010, 63 (5), 751– 761, DOI: 10.1016/j.jaad.2010.02.056Google Scholar97https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3cfpsl2huw%253D%253D&md5=cdb038bc6bd1b2a34aee1d5d104fecd1Pathologic nodal evaluation improves prognostic accuracy in Merkel cell carcinoma: analysis of 5823 cases as the basis of the first consensus staging systemLemos Bianca D; Storer Barry E; Iyer Jayasri G; Phillips Jerri Linn; Bichakjian Christopher K; Fang L Christine; Johnson Timothy M; Liegeois-Kwon Nanette J; Otley Clark C; Paulson Kelly G; Ross Merrick I; Yu Siegrid S; Zeitouni Nathalie C; Byrd David R; Sondak Vernon K; Gershenwald Jeffrey E; Sober Arthur J; Nghiem PaulJournal of the American Academy of Dermatology (2010), 63 (5), 751-61 ISSN:.BACKGROUND: The management of Merkel cell carcinoma (MCC) has been complicated by a lack of detailed prognostic data and by the presence of conflicting staging systems. OBJECTIVE: We sought to determine the prognostic significance of tumor size, clinical versus pathologic nodal evaluation, and extent of disease at presentation and thereby derive the first consensus staging/prognostic system for MCC. METHODS: A total of 5823 prospectively enrolled MCC cases from the National Cancer Data Base had follow-up data (median 64 months) and were used for prognostic analyses. RESULTS: At 5 years, overall survival was 40% and relative survival (compared with age- and sex-matched population data) was 54%. Among all MCC cases, 66% presented with local, 27% with nodal, and 7% with distant metastatic disease. For cases presenting with local disease only, smaller tumor size was associated with better survival (stage I, ≤2 cm, 66% relative survival at 5 years; stage II, >2 cm, 51%; P < .0001). Patients with clinically local-only disease and pathologically proven negative nodes had better outcome (76% at 5 years) than those who only underwent clinical nodal evaluation (59%, P < .0001). LIMITATIONS: The National Cancer Data Base does not capture disease-specific survival. Overall survival for patients with MCC was therefore used to calculate relative survival based on matched population data. CONCLUSION: Although the majority (68%) of patients with MCC in this nationwide cohort did not undergo pathologic nodal evaluation, this procedure may be indicated in many cases as it improves prognostic accuracy and has important treatment implications for those found to have microscopic nodal involvement.
- 98Deneve, J. L.; Messina, J. L.; Marzban, S. S.; Gonzalez, R. J.; Walls, B. M.; Fisher, K. J.; Chen, Y. A.; Cruse, C. W.; Sondak, V. K.; Zager, J. S. Merkel Cell Carcinoma of Unknown Primary Origin. Ann. Surg. Oncol. 2012, 19 (7), 2360– 2366, DOI: 10.1245/s10434-011-2213-2Google Scholar98https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38vhtVelsA%253D%253D&md5=2239b307f9f9e3c32457eede2e843e19Merkel cell carcinoma of unknown primary originDeneve Jeremiah L; Messina Jane L; Marzban Suroosh S; Gonzalez Ricardo J; Walls Brooke M; Fisher Kate J; Chen Y Ann; Cruse C Wayne; Sondak Vernon K; Zager Jonathan SAnnals of surgical oncology (2012), 19 (7), 2360-6 ISSN:.BACKGROUND: Merkel cell carcinoma (MCC) is a rare neuroendocrine tumor of the skin. MCC from an unknown primary origin (MCCUP) can present a diagnostic and therapeutic challenge. We describe our single-institution experience with the diagnosis and management of MCCUP presenting as metastases to lymph nodes. METHODS: After institutional review board approval, our institutional database spanning the years 1998-2010 was queried for patients with MCCUP. Clinicopathologic variables and outcomes were assessed. RESULTS: From a database of 321 patients with MCC, 38 (12%) were identified as having nodal MCCUP. Median age was 67 years, and 79% were men. Nodal basins involved at presentation were cervical (58%), axillary/epitrochlear (21%), or inguinal/iliac (21%). CK20 staining was positive in 93% of tumors tested, and all were negative for thyroid transcription factor-1. Twenty-nine patients (76%) underwent complete regional lymph node dissection (LND): 3 had LND alone, ten had LND and adjuvant radiotherapy, and 16 underwent LND followed by chemoradiotherapy. Definitive chemoradiotherapy without surgery was provided to six patients (16%), while radiotherapy alone was provided to three (8%). Recurrence was observed in 34% of patients. Median recurrence-free survival was 35 months. Ten patients (26%) died, five of disease and five of other causes. The median overall survival was 104 months. CONCLUSIONS: Nodal MCCUP is a rare disease affecting primarily elderly white men. Recurrence is observed in approximately one-third of patients, with a 104 month median overall survival after a multimodal treatment approach consisting of surgery along with adjuvant chemotherapy and radiotherapy in the majority of patients.
- 99Feng, H.; Shuda, M.; Chang, Y.; Moore, P. S. Clonal Integration of a Polyomavirus in Human Merkel Cell Carcinoma. Science 2008, 319 (5866), 1096– 1100, DOI: 10.1126/science.1152586Google Scholar99https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXit1yhsbY%253D&md5=79c4e37f638b771e22a19c78a41a0002Clonal Integration of a Polyomavirus in Human Merkel Cell CarcinomaFeng, Huichen; Shuda, Masahiro; Chang, Yuan; Moore, Patrick S.Science (Washington, DC, United States) (2008), 319 (5866), 1096-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Merkel cell carcinoma (MCC) is a rare but aggressive human skin cancer that typically affects elderly and immunosuppressed individuals, a feature suggestive of an infectious origin. The authors studied MCC samples by digital transcriptome subtraction and detected a fusion transcript between a previously undescribed virus T antigen and a human receptor tyrosine phosphatase. Further investigation led to identification and sequence anal. of the 5387-base-pair genome of a previously unknown polyomavirus that we call Merkel cell polyomavirus (MCV or MCPyV). MCV sequences were detected in 8 of 10 (80%) MCC tumors but only 5 of 59 (8%) control tissues from various body sites and 4 of 25 (16%) control skin tissues. In six of eight MCV-pos. MCCs, viral DNA was integrated within the tumor genome in a clonal pattern, suggesting that MCV infection and integration preceded clonal expansion of the tumor cells. Thus, MCV may be a contributing factor in the pathogenesis of MCC.
- 100Kassem, A.; Schöpflin, A.; Diaz, C.; Weyers, W.; Stickeler, E.; Werner, M.; Zur Hausen, A. Frequent Detection of Merkel Cell Polyomavirus in Human Merkel Cell Carcinomas and Identification of a Unique Deletion in the VP1 Gene. Cancer Res. 2008, 68 (13), 5009– 5013, DOI: 10.1158/0008-5472.CAN-08-0949Google Scholar100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXnvFOqt70%253D&md5=76d0f05b0574f9c9d59b2b08b9d92cb4Frequent detection of Merkel cell polyomavirus in human merkel cell carcinomas and identification of a unique deletion in the VP1 geneKassem, Ahmad; Schoepflin, Anja; Diaz, Carlos; Weyers, Wolfgang; Stickeler, Elmar; Werner, Martin; zur Hausen, AxelCancer Research (2008), 68 (13), 5009-5013CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Merkel cell carcinoma (MCC) is a rare but very aggressive human malignancy of the elderly or immunosuppressed patients. Recently, the clonal integration of a new human polyoma virus, which was termed Merkel cell polyomavirus (MCPyV), has been reported in 8 of 10 MCC patients. In the present study, we studied the formalin-fixed and paraffin-embedded tissue specimens of 39 MCC for the presence of MCPyV by PCR. We applied four different primer sets directed against the large T antigen and the VP1 gene of MCPyV. We were able to detect MCPyV in 77% (n = 30) of MCC as confirmed by sequence analyses of the PCR products. Sequence analyses showed only minor nucleotide changes compared with the previously published MCC sequences. In addn., one patient revealed the amplification of two PCR products using PCR primers directed against the VP1 gene. Sequence analyses confirmed the presence of the expected 351-bp PCR product and in addn. a second PCR product of 261 bp contg. a unique 90-bp deletion in the VP1 gene, which will lead to a predicted loss of 28 amino acids. The unique 90-bp deletion within the VP1 gene possibly is a result of incomplete viral integration of MCPyV in the MCC. The presence of MCPyV in the majority of MCC tissue specimens in our study strongly underlines a possible role for MCPyV as an etiol. agent in the carcinogenesis of MCC. [Cancer Res 2008;68(13):5009-13].
- 101Garneski, K. M.; Warcola, A. H.; Feng, Q.; Kiviat, N. B.; Leonard, J. H.; Nghiem, P. Merkel Cell Polyomavirus Is More Frequently Present in North American than Australian Merkel Cell Carcinoma Tumors. J. Invest. Dermatol. 2009, 129 (1), 246– 248, DOI: 10.1038/jid.2008.229Google Scholar101https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVymu77N&md5=3f2fdf306343f24becee55ed77c0d70cMerkel Cell Polyomavirus Is More Frequently Present in North American than Australian Merkel Cell Carcinoma TumorsGarneski, Kelly M.; Warcola, Ashley H.; Feng, Qinghua; Kiviat, Nancy B.; Leonard, J. Helen; Nghiem, PaulJournal of Investigative Dermatology (2009), 129 (1), 246-248CODEN: JIDEAE; ISSN:0022-202X. (Nature Publishing Group)The authors used quant. real-time PCR to detect the presence or absence of MCPyV DNA in 37 MCC tumor specimens originating from 37 patients in North America and Australia. It was found that 16 of 37 MCC tumor tissues were pos. for MCPyV DNA (43 %). Interestingly, the proportion of tumors pos. for MCPyV was much higher in tumors originating from North America than those originating from Australia. These data further present the interesting observation that North American MCC tumors are more frequently MCPyV pos. than Australian MCC tumors.
- 102Busam, K. J.; Jungbluth, A. A.; Rekthman, N.; Coit, D.; Pulitzer, M.; Bini, J.; Arora, R.; Hanson, N. C.; Tassello, J. A.; Frosina, D.; Moore, P.; Chang, Y. Merkel Cell Polyomavirus Expression in Merkel Cell Carcinomas and Its Absence in Combined Tumors and Pulmonary Neuroendocrine Carcinomas. Am. J. Surg. Pathol. 2009, 33 (9), 1378– 1385, DOI: 10.1097/PAS.0b013e3181aa30a5Google Scholar102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1Mrnsl2htw%253D%253D&md5=b298ece63e8807c3a7035d3e8ca1f4e4Merkel cell polyomavirus expression in merkel cell carcinomas and its absence in combined tumors and pulmonary neuroendocrine carcinomasBusam Klaus J; Jungbluth Achim A; Rekthman Natasha; Coit Daniel; Pulitzer Melissa; Bini Jason; Arora Reety; Hanson Nicole C; Tassello Jodie A; Frosina Denise; Moore Patrick; Chang YuanThe American journal of surgical pathology (2009), 33 (9), 1378-85 ISSN:.Merkel cell carcinoma (MCC) is the eponym for primary cutaneous neuroendocrine carcinoma. Recently, a new polyoma virus has been identified that is clonally integrated in the genome of the majority of MCCs, with truncating mutations in the viral large T antigen gene. We examined the presence of Merkel cell polyomavirus (MCV) in a set of 17 frozen tumor samples by quantitative polymerase chain reaction; 15 of them (88%) were positive. Sections from corresponding archival material were analyzed by immunohistochemistry (IHC) with the novel monoclonal antibody CM2B4, generated against a predicted antigenic epitope on the MCV T antigen, and tested for the expression of cytokeratin 20 (CK20). Sufficient archival material for IHC was available in only 15 of the 17 cases whose frozen tissue samples had been studied by polymerase chain reaction. Of the 15 tumors analyzed immunohistochemically, 10 (67%) showed positive labeling with CM2B4, 14 (93%) expressed CK20. A tissue microarray of 36 MCCs, 7 combined squamous and neuroendocrine carcinomas of the skin, and 26 pulmonary neuroendocrine carcinomas were also examined by IHC. Of the 36 MCCs assembled on a microarray, 32 (89%) tumors expressed CK20, and 27 (75%) were immunoreactive with CM2B4. The skin tumors with a combined squamous and neuroendocrine phenotype and all pulmonary neuroendocrine carcinomas failed to react with CM2B4. Our study shows that CM2B4 is a useful reagent for the diagnosis of MCC. It labels the majority of MCCs, but fails to react with pulmonary neuroendocrine carcinomas. We also found that neuroendocrine carcinomas of the skin arising in association with a squamous cell carcinoma seem to be independent of MCV.
- 103Kwun, H. J.; Guastafierro, A.; Shuda, M.; Meinke, G.; Bohm, A.; Moore, P. S.; Chang, Y. The Minimum Replication Origin of Merkel Cell Polyomavirus Has a Unique Large T-Antigen Loading Architecture and Requires Small T-Antigen Expression for Optimal Replication. J. Virol. 2009, 83 (23), 12118– 12128, DOI: 10.1128/JVI.01336-09Google Scholar103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVyrtLvM&md5=237b6a1865f86cbe20ac51aba0b9c36cThe minimum replication origin of Merkel cell polyomavirus has a unique large T-antigen loading architecture and requires small T-antigen expression for optimal replicationKwun, Hyun Jin; Guastafierro, Anna; Shuda, Masahiro; Meinke, Gretchen; Bohm, Andrew; Moore, Patrick S.; Chang, YuanJournal of Virology (2009), 83 (23), 12118-12128CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Merkel cell polyomavirus (MCV) is a recently discovered human polyomavirus causing the majority of human Merkel cell carcinomas. We mapped a 71-bp minimal MCV replication core origin sufficient for initiating eukaryotic DNA replication in the presence of wild-type MCV large T protein (LT). The origin includes a poly(T)-rich tract and eight variably oriented, GAGGC-like pentanucleotide sequences (PS) that serve as LT recognition sites. Mutation anal. shows that only four of the eight PS are required for origin replication. A single point mutation in one origin PS from a naturally occurring, tumor-derived virus reduces LT assembly on the origin and eliminates viral DNA replication. Tumor-derived LT having mutations truncating either the origin-binding domain or the helicase domain also prevent LT-origin assembly. Optimal MCV replication requires coexpression of MCV small T protein (sT), together with LT. An intact DnaJ domain on the LT is required for replication but is dispensable on the sT. In contrast, PP2A targeting by sT is required for enhanced replication. The MCV origin provides a novel model for eukaryotic replication from a defined DNA element and illustrates the selective pressure within tumors to abrogate independent MCV replication.
- 104Diaz, J.; Wang, X.; Tsang, S. H.; Jiao, J.; You, J. Phosphorylation of Large T Antigen Regulates Merkel Cell Polyomavirus Replication. Cancers (Basel). 2014, 6 (3), 1464, DOI: 10.3390/cancers6031464Google Scholar104https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cbgvVagtg%253D%253D&md5=026f085242b3afde971c748532cd28baPhosphorylation of large T antigen regulates merkel cell polyomavirus replicationDiaz Jason; Wang Xin; Tsang Sabrina H; You Jianxin; Jiao JingCancers (2014), 6 (3), 1464-86 ISSN:2072-6694.Merkel Cell Polyomavirus (MCPyV) was recently discovered as a novel human polyomavirus that is associated with ~80% of Merkel Cell Carcinomas. The Large Tumor antigen (LT) is an early viral protein which has a variety of functions, including manipulation of the cell cycle and initiating viral DNA replication. Phosphorylation plays a critical regulatory role for polyomavirus LT proteins, but no investigation of MCPyV LT phosphorylation has been performed to date. In this report mass spectrometry analysis reveals three unique phosphorylation sites: T271, T297 and T299. In vivo replication assays confirm that phosphorylation of T271 does not play a role in viral replication, while modification at T297 and T299 have dramatic and opposing effects on LT's ability to initiate replication from the viral origin. We test these mutants for their ability to bind, unwind, and act as a functional helicase at the viral origin. These studies provide a framework for understanding how phosphorylation of LT may dynamically regulate viral replication. Although the natural host cell of MCPyV has not yet been established, this work provides a foundation for understanding how LT activity is regulated and provides tools for better exploring this regulation in both natural host cells and Merkel cells.
- 105Houben, R.; Shuda, M.; Weinkam, R.; Schrama, D.; Feng, H.; Chang, Y.; Moore, P. S.; Becker, J. C. Merkel Cell Polyomavirus-Infected Merkel Cell Carcinoma Cells Require Expression of Viral T Antigens. J. Virol. 2010, 84 (14), 7064– 7072, DOI: 10.1128/JVI.02400-09Google Scholar105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVWls7jM&md5=605804b73156f383a947b79b6833db4fMerkel cell polyomavirus-infected Merkel cell carcinoma cells require expression of viral T antigensHouben, Roland; Shuda, Masahiro; Weinkam, Rita; Schrama, David; Feng, Huichen; Chang, Yuan; Moore, Patrick S.; Becker, Juergen C.Journal of Virology (2010), 84 (14), 7064-7072CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Merkel cell carcinoma (MCC) is the most aggressive skin cancer. Recently, it was demonstrated that human Merkel cell polyomavirus (MCV) is clonally integrated in ∼80% of MCC tumors. However, direct evidence for whether oncogenic viral proteins are needed for the maintenance of MCC cells is still missing. To address this question, we knocked down MCV T-antigen (TA) expression in MCV-pos. MCC cell lines using three different short hairpin RNA (shRNA)-expressing vectors targeting exon 1 of the TAs. The MCC cell lines used include three newly generated MCV-infected cell lines and one MCV-neg. cell line from MCC tumors. Notably, all MCV-pos. MCC cell lines underwent growth arrest and/or cell death upon TA knockdown, whereas the proliferation of MCV-neg. cell lines remained unaffected. Despite an increase in the no. of annexin V-pos., 7-amino-actinomycin D (7-AAD)-neg. cells upon TA knockdown, activation of caspases or changes in the expression and phosphorylation of Bcl-2 family members were not consistently detected after TA suppression. Our study provides the first direct exptl. evidence that TA expression is necessary for the maintenance of MCV-pos. MCC and that MCV is the infectious cause of MCV-pos. MCC.
- 106Houben, R.; Adam, C.; Baeurle, A.; Hesbacher, S.; Grimm, J.; Angermeyer, S.; Henzel, K.; Hauser, S.; Elling, R.; Bröcker, E. B.; Gaubatz, S.; Becker, J. C.; Schrama, D. An Intact Retinoblastoma Protein-Binding Site in Merkel Cell Polyomavirus Large T Antigen Is Required for Promoting Growth of Merkel Cell Carcinoma Cells. Int. J. cancer 2012, 130 (4), 847– 856, DOI: 10.1002/ijc.26076Google Scholar106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1egsL%252FM&md5=ae6b16a7739f7c6e863285f6a1af0959An intact retinoblastoma protein-binding site in Merkel cell polyomavirus large T antigen is required for promoting growth of Merkel cell carcinoma cellsHouben, Roland; Adam, Christian; Baeurle, Anne; Hesbacher, Sonja; Grimm, Johannes; Angermeyer, Sabrina; Henzel, Katharina; Hauser, Stefanie; Elling, Roland; Broecker, Eva-B.; Gaubatz, Stefan; Becker, Juergen C.; Schrama, DavidInternational Journal of Cancer (2012), 130 (4), 847-856CODEN: IJCNAW; ISSN:0020-7136. (John Wiley & Sons, Inc.)Merkel cell carcinoma (MCC) is a highly aggressive skin cancer that frequently harbours Merkel cell polyomavirus (MCV) DNA integrated in the genome of the tumor cells. In our study, we elaborate our recent finding that MCV-pos. MCC cell lines require the expression of the viral T antigens (TA). Indeed, in a xeno-transplantation model, we prove that TA expression is essential also in an in vivo situation, as knock down of TA leads to tumor regression. Moreover, rescuing TA short hairpin RNA (shRNA)-treated MCV-pos. MCC cells by ectopic expression of shRNA-insensitive TAs clearly demonstrates that the obsd. effect is caused by TA knockdown. Notably, introduction of a mutation in the LTA protein interfering with LTA binding to the retinoblastoma protein (RB) ablated this rescue. The importance of this interaction was further confirmed as LTA-specific knockdown leads to explicit cell growth inhibition. In summary, the presented data demonstrate that established MCV-pos. MCC tumors critically depend on TA expression, in particular the LTA and RB interaction, for sustained tumor growth. Consequently, interference with LTA/RB interaction appears as promising strategy to treat MCC.
- 107Cimino, P. J.; Robirds, D. H.; Tripp, S. R.; Pfeifer, J. D.; Abel, H. J.; Duncavage, E. J. Retinoblastoma Gene Mutations Detected by Whole Exome Sequencing of Merkel Cell Carcinoma. Mod. Pathol. 2014, 27 (8), 1073– 1087, DOI: 10.1038/modpathol.2013.235Google Scholar107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotFagug%253D%253D&md5=d8c78b26a9839ba213e67a714b7c2883Retinoblastoma gene mutations detected by whole exome sequencing of Merkel cell carcinomaCimino, Patrick J.; Robirds, Diane H.; Tripp, Sheryl R.; Pfeifer, John D.; Abel, Haley J.; Duncavage, Eric J.Modern Pathology (2014), 27 (8), 1073-1087CODEN: MODPEO; ISSN:0893-3952. (Nature Publishing Group)Merkel cell carcinoma is a highly aggressive cutaneous neuroendocrine tumor that has been assocd. with Merkel cell polyomavirus in up to 80% of cases. Merkel cell polyomavirus is believed to influence pathogenesis, at least in part, through expression of the large T antigen, which includes a retinoblastoma protein-binding domain. However, there appears to be significant clin. and morphol. overlap between polyomavirus-pos. and polyomavirus-neg. Merkel cell carcinoma cases. Although much of the recent focus of Merkel cell carcinoma pathogenesis has been on polyomavirus, the pathogenesis of polyomavirus-neg. cases is still poorly understood. We hypothesized that there are underlying human somatic mutations that unify Merkel cell carcinoma pathogenesis across polyomavirus status, and to investigate we performed whole exome sequencing on five polyomavirus-pos. cases and three polyomavirus-neg. cases. We found that there were no significant differences in the overall no. of single-nucleotide variations, copy no. variations, insertion/deletions, and chromosomal rearrangements when comparing polyomavirus-pos. to polyomavirus-neg. cases. However, we did find that the retinoblastoma pathway genes harbored a high no. of mutations in Merkel cell carcinoma. Furthermore, the retinoblastoma gene (RB1) was found to have nonsense truncating protein mutations in all three polyomavirus-neg. cases; no such mutations were found in the polyomavirus-pos. cases. In all eight cases, the retinoblastoma pathway dysregulation was confirmed by immunohistochem. Although polyomavirus-pos. Merkel cell carcinoma is believed to undergo retinoblastoma dysregulation through viral large T antigen expression, our findings demonstrate that somatic mutations in polyomavirus-neg. Merkel cell carcinoma lead to retinoblastoma dysregulation through an alternative pathway. This novel finding suggests that the retinoblastoma pathway dysregulation leads to an overlapping Merkel cell carcinoma phenotype and that oncogenesis occurs through either a polyomavirus-dependent (viral large T antigen expression) or polyomavirus-independent (host somatic mutation) mechanism.
- 108Harms, P. W.; Vats, P.; Verhaegen, M. E.; Robinson, D. R.; Wu, Y. M.; Dhanasekaran, S. M.; Palanisamy, N.; Siddiqui, J.; Cao, X.; Su, F.; Wang, R.; Xiao, H.; Kunju, L. P.; Mehra, R.; Tomlins, S. A.; Fullen, D. R.; Bichakjian, C. K.; Johnson, T. M.; Dlugosz, A. A.; Chinnaiyan, A. M. The Distinctive Mutational Spectra of Polyomavirus-Negative Merkel Cell Carcinoma. Cancer Res. 2015, 75 (18), 3720– 3727, DOI: 10.1158/0008-5472.CAN-15-0702Google Scholar108https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsV2gt7fP&md5=07a60536465c72220f31a13bd4746618The Distinctive Mutational Spectra of Polyomavirus-Negative Merkel Cell CarcinomaHarms, Paul William; Vats, Pankaj; Verhaegen, Monique Elise; Robinson, Dan R.; Wu, Yi-Mi; Dhanasekaran, Saravana Mohan; Palanisamy, Nallasivam; Siddiqui, Javed; Cao, Xuhong; Su, Fengyun; Wang, Rui; Xiao, Hong; Kunju, Lakshmi P.; Mehra, Rohit; Tomlins, Scott A.; Fullen, Douglas Randall; Bichakjian, Christopher Keram; Johnson, Timothy M.; Dlugosz, Andrzej Antoni; Chinnaiyan, Arul M.Cancer Research (2015), 75 (18), 3720-3727CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Merkel cell carcinoma (MCC) is a rare but highly aggressive cutaneous neuroendocrine tumor. Merkel cell polyomavirus (MCPyV) may contribute to tumorigenesis in a subset of tumors via inhibition of tumor suppressors such as retinoblastoma (RB1) by mutated viral T antigens, but the mol. pathogenesis of MCPyV-neg. MCC is largely unexplored. Through our MI-ONCOSEQ precision oncol. study, we performed integrative sequencing on two cases of MCPyV-neg. MCC, as well as a validation cohort of 14 addnl. MCC cases (n = 16). In addn. to previously identified mutations in TP53, RB1, and PIK3CA, we discovered activating mutations of oncogenes, including HRAS and loss-of-function mutations in PRUNE2 and NOTCH family genes in MCPyV-neg. MCC. MCPyV-neg. tumors also displayed high overall mutation burden (10.09 ± 2.32 mutations/Mb) and were characterized by a prominent UV-signature pattern with C > T transitions comprising 85% of mutations. In contrast, mutation burden was low in MCPyV-pos. tumors (0.40 ± 0.09 mutations/Mb) and lacked a UV signature. These findings suggest a potential ontol. dichotomy in MCC, characterized by either viral-dependent or UV-dependent tumorigenic pathways. Cancer Res; 75(18); 3720-7. ©2015 AACR.
- 109Sahi, H.; Savola, S.; Sihto, H.; Koljonen, V.; Bohling, T.; Knuutila, S. RB1 Gene in Merkel Cell Carcinoma: Hypermethylation in All Tumors and Concurrent Heterozygous Deletions in the Polyomavirus-Negative Subgroup. APMIS 2014, 122 (12), 1157– 1166, DOI: 10.1111/apm.12274Google Scholar109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVektLvL&md5=be382c90f93f02dc801701ea6ce74b66RB1 gene in Merkel cell carcinoma: hypermethylation in all tumors and concurrent heterozygous deletions in the polyomavirus-negative subgroupSahi, Helka; Savola, Suvi; Sihto, Harri; Koljonen, Virve; Bohling, Tom; Knuutila, SakariAPMIS (2014), 122 (12), 1157-1166CODEN: APMSEL; ISSN:1600-0463. (Wiley-Blackwell)Sequestration of the tumor suppressor retinoblastoma protein (RB) by the Merkel cell polyomavirus (MCV) is a crucial step in the pathogenesis of Merkel cell carcinoma (MCC). RB expression is frequently lost, particularly in MCV-neg. MCC tumors, through yet unknown mechanisms. We compared the genomic copy no. changes of 13 MCV-pos. and 13 -neg. MCC tumors by array comparative genomic hybridization. The anal. revealed increased genomic instability, amplification of 1p34.3-1p34.2, and losses of 11p in the absence of MCV infection. Deletions of the RB1 locus were also detected at high rates in MCV-neg. tumors. None of the tumors with heterozygous RB1 losses expressed RB in immunohistochem. RB1 promoter hypermethylation was studied with a methylation-specific multiplex ligation-dependent probe amplification technique. The RB1 promoter was methylated in all tumor specimens at CpG islands located close to the ATG start codon, albeit at low levels. The pattern of hypermethylation was similar in all MCC samples, despite RB expression, survival or MCV status. In conclusion, the frequent heterozygous losses of the RB1 locus could partly explain the decreased RB expression in MCV-neg. MCC tumors, although the effects of RB1 mutations, coinciding promoter hypermethylation and, for example, miRNA regulation, cannot be excluded.
- 110Starrett, G. J.; Marcelus, C.; Cantalupo, P. G.; Katz, J. P.; Cheng, J.; Akagi, K.; Thakuria, M.; Rabinowits, G.; Wang, L. C.; Symer, D. E.; Pipas, J. M.; Harris, R. S.; DeCaprio, J. A. Merkel Cell Polyomavirus Exhibits Dominant Control of the Tumor Genome and Transcriptome in Virus-Associated Merkel Cell Carcinoma. MBio 2017, 8 (1), e02079, DOI: 10.1128/mBio.02079-16Google Scholar110https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXjsFeksr0%253D&md5=3fe9739322b5e3a30d43032768296e00Merkel cell polyomavirus exhibits dominant control of the tumor genome and transcriptome in virus-associated merkel cell carcinomaStarrett, Gabriel J.; Marcelus, Christina; Cantalupo, Paul G.; Katz, Joshua P.; Cheng, Jingwei; Akagi, Keiko; Thakuria, Manisha; Rabinowits, Guilherme; Wang, Linda C.; Symer, David E.; Pipas, James M.; Harris, Reuben S.; DeCaprio, James A.mBio (2017), 8 (1), e02079-16/1-e02079-16/14CODEN: MBIOCL; ISSN:2150-7511. (American Society for Microbiology)Merkel cell polyomavirus is the primary etiol. agent of the aggressive skin cancer Merkel cell carcinoma (MCC). Recent studies have revealed that UV radiation is the primary mechanism for somatic mutagenesis in nonviral forms of MCC. Here, we analyze the whole transcriptomes and genomes of primary MCC tumors. Our study reveals that virus-assocd. tumors have minimally altered genomes compared to non-virus-assocd. tumors, which are dominated by UV-mediated mutations. Although virus-assocd. tumors contain relatively small mutation burdens, they exhibit a distinct mutation signature with observable transcriptionally biased kataegic events. In addn., viral integration sites overlap focal genome amplifications in virus-assocd. tumors, suggesting a potential mechanism for these events. Collectively, our studies indicate that Merkel cell polyomavirus is capable of hijacking cellular processes and driving tumorigenesis to the same severity as tens of thousands of somatic genome alterations.
- 111Borchert, S.; Czech-Sioli, M.; Neumann, F.; Schmidt, C.; Wimmer, P.; Dobner, T.; Grundhoff, A.; Fischer, N. High-Affinity Rb Binding, P53 Inhibition, Subcellular Localization, and Transformation by Wild-Type or Tumor-Derived Shortened Merkel Cell Polyomavirus Large T Antigens. J. Virol. 2014, 88 (6), 3144– 3160, DOI: 10.1128/JVI.02916-13Google Scholar111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmsVOqs7s%253D&md5=be1a7d0c0ded90ad295f5c97e8b1759aHigh-affinity Rb binding, p53 inhibition, subcellular localization and transformation by wild-type or tumor-derived shortened Merkel cell polyomavirus large T antigensBorchert, Sophie; Czech-Sioli, Manja; Neumann, Friederike; Schmidt, Claudia; Wimmer, Peter; Dobner, Thomas; Grundhoff, Adam; Fischer, NicoleJournal of Virology (2014), 88 (6), 3144-3160, 18 pp.CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Interference with tumor suppressor pathways by polyomavirus-encoded tumor antigens (T-Ags) can result in transformation. Consequently, it is thought that T-Ags encoded by Merkel cell polyomavirus (MCPyV), a virus integrated in ∼90% of all Merkel cell carcinoma (MCC) cases, are major contributors to tumorigenesis. The MCPyV large T-Ag (LT-Ag) has preserved the key functional domains present in all family members but has also acquired unique regions that flank the LxCxE motif. As these regions may mediate unique functions or may modulate those shared with T-Ags of other polyomaviruses, functional studies of MCPyV T-Ags are required. Here, we have performed a comparative study of full-length or MCC-derived truncated LT-Ags with regard to their biochem. characteristics, their ability to bind to retinoblastoma (Rb) and p53 proteins and their transforming potential. We provide evidence that full-length MCPyV LT-Ag may not directly bind to p53 but nevertheless can significantly reduce p53-dependent transcription in reporter assays. Although early region expression constructs harboring either full-length or MCC-derived truncated LT-Ag genes can transform primary baby rat kidney cells, truncated LT-Ags do not bind to p53 or reduce p53-dependent transcription. Interestingly, shortened LT-Ags exhibit a very high binding affinity for Rb, as shown by coimmunopptn. and in vitro binding studies. Addnl., we show that truncated MCPyV LT-Ag proteins are expressed at higher levels than those for the wild-type protein and are able to partially relocalize Rb to the cytoplasm, indicating that truncated LT proteins may have gained addnl. features that distinguish them from the full-length protein.
- 112Cheng, J.; Rozenblatt-Rosen, O.; Paulson, K. G.; Nghiem, P.; DeCaprio, J. A. Merkel Cell Polyomavirus Large T Antigen Has Growth-Promoting and Inhibitory Activities. J. Virol. 2013, 87 (11), 6118, DOI: 10.1128/JVI.00385-13Google Scholar112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFaktrnL&md5=57b360e79fe2aa525b16cd0036862a49Merkel cell polyomavirus large T antigen has growth-promoting and inhibitory activitiesCheng, Jingwei; Rozenblatt-Rosen, Orit; Paulson, Kelly G.; Nghiem, Paul; DeCaprio, James A.Journal of Virology (2013), 87 (11), 6118-6126CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)Merkel cell carcinoma (MCC) is a rare and aggressive form of skin cancer. In at least 80% of all MCC, Merkel cell polyomavirus (MCPyV) DNA has undergone clonal integration into the host cell genome, and most tumors express the MCPyV large and small T antigens. In all cases of MCC reported to date, the integrated MCPyV genome has undergone mutations in the large T antigen. These mutations result in expression of a truncated large T antigen that retains the Rb binding or LXCXE motif but deletes the DNA binding and helicase domains. However, the transforming functions of full-length and truncated MCPyV large T antigen are unknown. We compared the transforming activities of full-length, truncated, and alternatively spliced 57kT forms of MCPyV large T antigen. MCPyV large T antigen could bind to Rb but was unable to bind to p53. Furthermore, MCPyV-truncated large T antigen was more effective than full-length and 57kT large T antigen in promoting the growth of human and mouse fibroblasts. In contrast, expression of the MCPyV large T antigen C-terminal 100 residues could inhibit the growth of several different cell types. These data imply that the deletion of the C terminus of MCPyV large T antigen found in MCC serves not only to disrupt viral replication but also results in the loss of a distinct growth-inhibitory function intrinsic to this region.
- 113Li, J.; Wang, X.; Diaz, J.; Tsang, S. H.; Buck, C. B.; You, J. Merkel Cell Polyomavirus Large T Antigen Disrupts Host Genomic Integrity and Inhibits Cellular Proliferation. J. Virol. 2013, 87 (16), 9173, DOI: 10.1128/JVI.01216-13Google Scholar113https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1WgtLrF&md5=bff6460e974c4895719ab3158da6021aMerkel cell polyomavirus large T antigen disrupts host genomic integrity and inhibits cellular proliferationLi, Jing; Wang, Xin; Diaz, Jason; Tsang, Sabrina H.; Buck, Christopher B.; You, JianxinJournal of Virology (2013), 87 (16), 9173-9188CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Clonal integration of Merkel cell polyomavirus (MCV) DNA into the host genome has been obsd. in at least 80% of Merkel cell carcinoma (MCC). The integrated viral genome typically carries mutations that truncate the C-terminal DNA binding and helicase domains of the MCV large T antigen (LT), suggesting a selective pressure to remove this MCV LT region during tumor development. In this study, we show that MCV infection leads to the activation of host DNA damage responses (DDR). This activity was mapped to the C-terminal helicase-contg. region of the MCV LT. The MCV LT-activated DNA damage kinases, in turn, led to enhanced p53 phosphorylation, upregulation of p53 downstream target genes, and cell cycle arrest. Compared to the N-terminal MCV LT fragment that is usually preserved in mutants isolated from MCC tumors, full-length MCV LT shows a decreased potential to support cellular proliferation, focus formation, and anchorage-independent cell growth. These apparently antitumorigenic effects can be reversed by a dominant-neg. p53 inhibitor. Our results demonstrate that MCV LT-induced DDR activates p53 pathway, leading to the inhibition of cellular proliferation. This study reveals a key difference between MCV LT and simian vacuolating virus 40 LT, which activates a DDR but inhibits p53 function. This study also explains, in part, why truncation mutations that remove the MCV LT C-terminal region are necessary for the oncogenic progression of MCV-assocd. cancers.
- 114Shuda, M.; Kwun, H. J.; Feng, H.; Chang, Y.; Moore, P. S. Human Merkel Cell Polyomavirus Small T Antigen Is an Oncoprotein Targeting the 4E-BP1 Translation Regulator. J. Clin. Invest. 2011, 121 (9), 3623– 3634, DOI: 10.1172/JCI46323Google Scholar114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFOhsbfP&md5=6cf79ee7790b245d54ba7d95aea5bfbbHuman Merkel cell polyomavirus small T antigen is an oncoprotein targeting the 4E-BP1 translation regulatorShuda, Masahiro; Kwun, Hyun Jin; Feng, Huichen; Chang, Yuan; Moore, Patrick S.Journal of Clinical Investigation (2011), 121 (9), 3623-3634CODEN: JCINAO; ISSN:0021-9738. (American Society for Clinical Investigation)Merkel cell polyomavirus (MCV) is the recently discovered cause of most Merkel cell carcinomas (MCCs), an aggressive form of nonmelanoma skin cancer. Although MCV is known to integrate into the tumor cell genome and to undergo mutation, the mol. mechanisms used by this virus to cause cancer are unknown. Here, we show that MCV small T (sT) antigen is expressed in most MCC tumors, where it is required for tumor cell growth. Unlike the closely related SV40 sT, MCV sT transformed rodent fibroblasts to anchorage- and contact-independent growth and promoted serum-free proliferation of human cells. These effects did not involve protein phosphatase 2A (PP2A) inhibition. MCV sT was found to act downstream in the mammalian target of rapamycin (mTOR) signaling pathway to preserve eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) hyperphosphorylation, resulting in dysregulated cap-dependent translation. MCV sT-assocd. 4E-BP1 serine 65 hyperphosphorylation was resistant to mTOR complex (mTORC1) and mTORC2 inhibitors. Steady-state phosphorylation of other downstream Akt-mTOR targets, including S6K and 4E-BP2, was also increased by MCV sT. Expression of a constitutively active 4E-BP1 that could not be phosphorylated antagonized the cell transformation activity of MCV sT. Taken together, these expts. showed that 4E-BP1 inhibition is required for MCV transformation. Thus, MCV sT is an oncoprotein, and its effects on dysregulated cap-dependent translation have clin. implications for the prevention, diagnosis, and treatment of MCV-related cancers.
- 115Rodriguez-Viciana, P.; Collins, C.; Fried, M. Polyoma and SV40 Proteins Differentially Regulate PP2A to Activate Distinct Cellular Signaling Pathways Involved in Growth Control. Proc. Natl. Acad. Sci. U. S. A. 2006, 103 (51), 19290, DOI: 10.1073/pnas.0609343103Google Scholar115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhs1ymtQ%253D%253D&md5=0c75f0e6ef92c031b84b996442d0b30ePolyoma and SV40 proteins differentially regulate PP2A to activate distinct cellular signaling pathways involved in growth controlRodriguez-Viciana, Pablo; Collins, Crista; Fried, MikeProceedings of the National Academy of Sciences of the United States of America (2006), 103 (51), 19290-19295CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Binding of Src family kinases to membrane-assocd. polyoma virus middle T-antigen (PyMT) can result in the phosphorylation of PyMT tyrosine 250, which serves as a docking site for the binding of Shc and subsequent activation of the Raf-MEK-ERK (MAP) kinase cascade. In a screen for PyMT variants that could not activate the ARF tumor suppressor, we isolated a cytoplasmic nontransforming mutant (MTA) that encoded a C-terminal truncated form of the PyMT protein. Surprisingly, MTA was able to strongly activate the MAP kinase pathway in the absence of Src family kinase and Shc binding. Interestingly, the polyoma small T-antigen (PyST), which shares with MTA both partial amino acid sequence homol. and cellular location, also activates the MAP kinase cascade. Activation of the MAP kinase cascade by both MTA and PyST has been demonstrated to be PP2A-dependent. Neither MTA nor PyST activate the phosphorylation of AKT. The SV40 small T-antigen, which is similar to PyST in contg. a J domain and in binding to the PP2A AC dimer, does not activate the MAP kinase cascade, but does stimulate phosphorylation of AKT in a PP2A-dependent manner. These findings highlight a novel role of PP2A in stimulating the MAP kinase cascade and indicate that the similar polyoma and SV40 small T-antigens influence PP2A to activate discrete cellular signaling pathways involved in growth control.
- 116Kwun, H. J.; Shuda, M.; Camacho, C. J.; Gamper, A. M.; Thant, M.; Chang, Y.; Moore, P. S. Restricted Protein Phosphatase 2A Targeting by Merkel Cell Polyomavirus Small T Antigen. J. Virol. 2015, 89 (8), 4191– 4200, DOI: 10.1128/JVI.00157-15Google Scholar116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlvVWltLY%253D&md5=dd42397520a30eb370830a66b89bddf3Restricted protein phosphatase 2A targeting by Merkel cell polyomavirus small T antigenKwun, Hyun Jin; Shuda, Masahiro; Camacho, Carlos J.; Gamper, Armin M.; Thant, Mamie; Chang, Yuan; Moore, Patrick S.Journal of Virology (2015), 89 (8), 4191-4200CODEN: JOVIAM; ISSN:1098-5514. (American Society for Microbiology)Merkel cell polyomavirus (MCV) is a newly discovered human cancer virus encoding a small T (sT) oncoprotein. We performed MCV sT FLAG-affinity purifn. followed by mass spectroscopy (MS) anal., which identified several protein phosphatases (PP), including PP2A A and C subunits and PP4C, as potential cellular interacting proteins. PP2A targeting is crit. for the transforming properties of nonhuman polyomaviruses, such as simian virus 40 (SV40), but is not required for MCV sT-induced rodent cell transformation. We compared similarities and differences in PP2A binding between MCV and SV40 sT. While SV40 sT coimmunopurified with subunits PP2A Aα and PP2A C, MCV sT coimmunopurified with PP2A Aα, PP2A Aβ, and PP2A C. Scanning alanine mutagenesis at 29 sites across the MCV sT protein revealed that PP2A-binding domains lie on the opposite mol. surface from a previously described large T stabilization domain (LSD) loop that binds E3 ligases, such as Fbw7. MCV sT-PP2A interactions can be functionally distinguished by mutagenesis from MCV sT LSD-dependent 4E-BP1 hyperphosphorylation and viral DNA replication enhancement. MCV sT has a restricted range for PP2A B subunit substitution, inhibiting only the assembly of B56α into the phosphatase holoenzyme. In contrast, SV40 sT inhibits the assembly of B55α, B56α and B56ε into PP2A. We conclude that MCV sT is required for Merkel cell carcinoma growth, but its in vitro transforming activity depends on LSD interactions rather than PP2A targeting.
- 117Munde, P. B.; Khandekar, S. P.; Dive, A. M.; Sharma, A. Pathophysiology of Merkel Cell. J. Oral Maxillofac. Pathol. 2013, 17 (3), 408, DOI: 10.4103/0973-029X.125208Google Scholar117https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cfls1GmtA%253D%253D&md5=2be52b2b5ad6ad91234e363e3b084b04Pathophysiology of merkel cellMunde Prashant Balasaheb; Khandekar Shubhangi P; Dive Alka M; Sharma AparnaJournal of oral and maxillofacial pathology : JOMFP (2013), 17 (3), 408-12 ISSN:0973-029X.The objective of this review is to introduce Merkel cells (MCs), to provide a basic overview on the theoretical background of function, development and clinical importance of MCs. The origin of human MCs have been controversial. Some investigators believe that it is a neural crest derivate, whereas others have proposed that it is a differentiation product of the fetal epidermal keratinocytes. MCs are cells primarily localized in the epidermal basal layer of vertebrates and concentrated in touch-sensitive areas in glabrous, hairy skin and in some mucosa. In routine light microscopy, human MCs can hardly be identified. Cytokeratin 20 (CK20) is a reliable marker with highest degree of specificity. MCs can be also distinguished by electron microscopy. MC carcinoma (MCC) is an uncommon and often aggressive malignancy and found mainly in elderly patients. It occurs most frequently in the head and neck region. Diagnosis is based on typical histological presentation on hematoxylin and eosin (H and E) stained slides together with the results of immunohistochemistry. Histologically, MCC has been classified into three distinct subtypes: Trabecular, intermediate and small cell type.
- 118zur Hausen, A.; Rennspiess, D.; Winnepenninckx, V.; Speel, E.-J.; Kurz, A. K. Early B-Cell Differentiation in Merkel Cell Carcinomas: Clues to Cellular Ancestry. Cancer Res. 2013, 73 (16), 4982– 4987, DOI: 10.1158/0008-5472.CAN-13-0616Google Scholar118https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXht1OisrbI&md5=076bff27272b1f48c191ad37f95583efEarly B-Cell Differentiation in Merkel Cell Carcinomas: Clues to Cellular Ancestryzur Hausen, Axel; Rennspiess, Dorit; Winnepenninckx, Veronique; Speel, Ernst-Jan; Kurz, Anna KordeliaCancer Research (2013), 73 (16), 4982-4987CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Merkel cell carcinoma (MCC) is a highly malignant neuroendocrine nonmelanoma skin cancer, which is assocd. with the Merkel cell polyoma virus (MCPyV). Recently, expression of the terminal deoxynucleotidyl transferase (TdT) and the paired box gene 5 (PAX 5) has been consistently reported in the majority of MCCs. We tested 21 MCCs for the expression of MCPyV, TdT, PAX5, IgG, IgM, IgA, kappa, and lambda by immunohistochem. and assessed IgH and Igk rearrangement in all 21 MCCs. All of the MCCs revealed specific expression of PAX5 and 72.8% of the MCCs expressed TdT. In addn., most of the MCCs revealed specific expression of one or more Ig subclasses and kappa or lambda. One MCC did reveal monoclonal IgH and Igk rearrangement next to two other MCCs showing Igk rearrangement. As coexpression of TdT and PAX5 under physiol. circumstances is restricted to pro/pre- and pre-B cells we propose, on the basis of our results, that the cell of origin of MCCs is a pro/pre- or pre-B cell rather than the postmitotic Merkel cells. MCPyV infection and transformation of pro-/pre-B cells are likely to induce the expression of simple cytokeratins as has been shown for SV40 in other nonepithelial cells. This model of cellular ancestry of MCCs might impact therapy and possibly helps to understand why approx. 20% of MCCs are MCPyV-neg.
- 119Calder, K. B.; Smoller, B. R. New Insights into Merkel Cell Carcinoma. Adv. Anat. Pathol. 2010, 17 (3), 155– 161, DOI: 10.1097/PAP.0b013e3181d97836Google Scholar119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3c3oslygtA%253D%253D&md5=1db9780d2115c1ca562c2470441cac1aNew insights into merkel cell carcinomaCalder Kenneth B; Smoller Bruce RAdvances in anatomic pathology (2010), 17 (3), 155-61 ISSN:.Merkel cell carcinoma (MCC) is a rare aggressive cutaneous malignancy of the elderly and immunocompromised populations. The clinical presentation of MCC is nonspecific, with the majority of cases presenting as localized skin involvement. Histologically and immunophenotypically, MCC is defined by both neuroendocrine and epithelial differentiation. Recently, the Merkel cell polyomavirus has been implicated in the pathogenesis of MCC. In addition, there have been numerous studies evaluating the histologic and immunohistochemical characteristics of MCC as they relate to diagnosis and prognosis. The purpose of this paper is to review the most salient and clinically relevant updates in the pathogenesis and histologic features of MCC. Specific attention is given to the clinical and histologic predictors of prognosis, staging, and the controversies concerning sentinel lymph node biopsy and therapy.
- 120Fan, K.; Gravemeyer, J.; Ritter, C.; Rasheed, K.; Gambichler, T.; Moens, U.; Shuda, M.; Schrama, D.; Becker, J. C. MCPyV Large T Antigen-Induced Atonal Homolog 1 Is a Lineage-Dependency Oncogene in Merkel Cell Carcinoma. J. Invest. Dermatol. 2020, 140 (1), 56– 65, e3 DOI: 10.1016/j.jid.2019.06.135Google Scholar120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvFSqsb7P&md5=e3b1a675c05fe32a366d7966b808fc3dMCPyV Large T Antigen-Induced Atonal Homolog 1 Is a Lineage-Dependency Oncogene in Merkel Cell CarcinomaFan, Kaiji; Gravemeyer, Jan; Ritter, Cathrin; Rasheed, Kashif; Gambichler, Thilo; Moens, Ugo; Shuda, Masahiro; Schrama, David; Becker, Juergen C.Journal of Investigative Dermatology (2020), 140 (1), 56-65.e3CODEN: JIDEAE; ISSN:0022-202X. (Elsevier Inc.)Despite the fact that the transcription factor ATOH1 is a master regulator of Merkel cell development, its role in Merkel cell carcinoma (MCC) carcinogenesis remains controversial. Here, we provide several lines of evidence that ATOH1 is a lineage-dependent oncogene in MCC. Luciferase assays revealed binding of ATOH1 and subsequent activation to the promoter of miR-375, which is one of the most abundant microRNAs in MCCs. Overexpression of ATOH1 in variant MCC cell lines and fibroblasts induced miR-375 expression, whereas ATOH1 knockdown in classical MCC cell lines reduced miR-375 expression. Moreover, ATOH1 overexpression in these cells changed their growth characteristics from adherent to suspension and/orspheroidal growth, i.e., resembling the neuroendocrine growth pattern of classical MCC cell lines. Notably, ectopic expression of different Merkel cell polyomavirus (MCPyV)-derived truncated large T antigens induced ATOH1 expression in fibroblasts, which was paralleled by miR-375 expression and similar morphol. changes. In summary, MCPyV-assocd. carcinogenesis is likely to induce the characteristic neuroendocrine features of MCC via induction of ATOH1; thus, ATOH1 can be regarded as a lineage-dependent oncogene in MCC.
- 121Liu, W.; You, J. Molecular Mechanisms of Merkel Cell Polyomavirus Transformation and Replication. Annu. Rev. Virol. 2020, 7 (1), 289, DOI: 10.1146/annurev-virology-011720-121757Google Scholar121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlSrtrzI&md5=21328f9572ce5c9aaba65432670e7c73Molecular Mechanisms of Merkel Cell Polyomavirus Transformation and ReplicationLiu, Wei; You, JianxinAnnual Review of Virology (2020), 7 (), 289-307CODEN: ARVNBS; ISSN:2327-0578. (Annual Reviews)Viral infection underlies a significant share of the global cancer burden. Merkel cell polyomavirus (MCPyV) is the newest member of the human oncogenic virus family. Its discovery over a decade ago marked the beginning of an exciting era in human tumor virol. Since then, significant evidences have emerged to support the etiol. role of MCPyV in Merkel cell carcinoma (MCC), an extremely lethal form of skin cancer. MCPyV infection is widespread in the general population. MCC diagnoses have tripled over the past 20 years, but effective treatments are currently lacking. In this review, we highlight recent discoveries that have shaped our understanding of MCPyV oncogenic mechanism and host cellular tropism, as well as the mol. events occurring in the viral infectious life cycle. These insights will guide future efforts in developing novel virus-targeted therapeutic strategies for treating the devastating human cancers assocd. with this new tumorigenic virus.
- 122Midgard, H.; Weir, A.; Palmateer, N.; Lo Re, V.; Pineda, J. A.; Macías, J.; Dalgard, O. HCV Epidemiology in High-Risk Groups and the Risk of Reinfection. J. Hepatol. 2016, 65 (1), S33– S45, DOI: 10.1016/j.jhep.2016.07.012Google ScholarThere is no corresponding record for this reference.
- 123Shepard, C. W.; Finelli, L.; Alter, M. J. Global Epidemiology of Hepatitis C Virus Infection. Lancet Infect. Dis. 2005, 5 (9), 558– 567, DOI: 10.1016/S1473-3099(05)70216-4Google Scholar123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD2Mvls1Wlsg%253D%253D&md5=4d5f81fa0c2191b8c32b328e39933fadGlobal epidemiology of hepatitis C virus infectionShepard Colin W; Finelli Lyn; Alter Miriam JThe Lancet. Infectious diseases (2005), 5 (9), 558-67 ISSN:1473-3099.Hepatitis C virus (HCV) is a major cause of liver disease worldwide and a potential cause of substantial morbidity and mortality in the future. The complexity and uncertainty related to the geographic distribution of HCV infection and chronic hepatitis C, determination of its associated risk factors, and evaluation of cofactors that accelerate its progression, underscore the difficulties in global prevention and control of HCV. Because there is no vaccine and no post-exposure prophylaxis for HCV, the focus of primary prevention efforts should be safer blood supply in the developing world, safe injection practices in health care and other settings, and decreasing the number of people who initiate injection drug use.
- 124Goto, K.; Roca Suarez, A. A.; Wrensch, F.; Baumert, T. F.; Lupberger, J. Hepatitis C Virus and Hepatocellular Carcinoma: When the Host Loses Its Grip. Int. J. Mol. Sci. 2020, 21 (9), 3057, DOI: 10.3390/ijms21093057Google Scholar124https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvVOjsrnJ&md5=80a80f5160612d4e1874026907ef2ac2Hepatitis C virus and hepatocellular carcinoma: when the host loses its gripGoto, Kaku; Suarez, Armando Andres Roca; Wrensch, Florian; Baumert, Thomas F.; Lupberger, JoachimInternational Journal of Molecular Sciences (2020), 21 (9), 3057CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)A review. Chronic infection with hepatitis C virus (HCV) is a major cause of hepatocellular carcinoma (HCC). Novel treatments with direct-acting antivirals achieve high rates of sustained virol. response; however, the HCC risk remains elevated in cured patients, esp. those with advanced liver disease. Long-term HCV infection causes a persistent and accumulating damage of the liver due to a combination of direct and indirect pro-oncogenic mechanisms. This review describes the processes involved in virus-induced disease progression by viral proteins, derailed signaling, immunity, and persistent epigenetic deregulation, which may be instrumental to develop urgently needed prognostic biomarkers and as targets for novel chemopreventive therapies.
- 125Koike, K.; Tsutsumi, T.; Fujie, H.; Shintani, Y.; Moriya, K. Molecular Mechanism of Viral Hepatocarcinogenesis. Oncology 2002, 62 (SUPPL. 1), 29– 37, DOI: 10.1159/000048273Google Scholar125https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD387jsVSktA%253D%253D&md5=5972ec14d9f850bfbc01674d546a9e3bMolecular mechanism of viral hepatocarcinogenesisKoike Kazuhiko; Tsutsumi Takeya; Fujie Hajime; Shintani Yoshizumi; Kyoji MoriyaOncology (2002), 62 Suppl 1 (), 29-37 ISSN:0030-2414.Overwhelming lines of epidemiological evidence have indicated that chronic infection with hepatitis B virus (HBV) or hepatitis C virus (HCV) is a major risk for the development of hepatocellular carcinoma (HCC). In the pathogenesis of HCC associated with HBV or HCV, it remains controversial whether these hepatitis viruses play a direct role or merely an indirect role. By virtue of transgenic mice established by us, it has become evident that the product of the HBV X gene (HBx protein) and the core protein of HCV have an oncogenic potential, although the pathways through which these two viral proteins operate may differ. The findings in our studies indicate that HBV and HCV are directly involved in hepatocarcinogenesis, albeit other factors such as continued cell death and regeneration associated with chronic hepatitis may play a role as well. Combined, our results suggest that there might be a mechanism in the development of HCC in persistent infection with hepatitis viruses that is distinct from that in other cancers. Similarly to the pathogenesis of other malignancies represented by colorectal cancer, the accumulation of a set of genetic aberrations may also be necessary for a multistage development of HCC. However, HBx protein and HCV core protein, to which an oncogenic potential is attributed, may allow some of the multiple stages skipped in hepatocarcinogenesis. Unlike for the other cancers, therefore, infection with HBV or HCV may be capable of inducing HCC in the absence of a complete set of genetic aberrations. Such a scenario would explain an unusually high incidence and multicentric nature of HCC developing in chronic hepatitis B or C.
- 126Catanese, M. T.; Uryu, K.; Kopp, M.; Edwards, T. J.; Andrus, L.; Rice, W. J.; Silvestry, M.; Kuhn, R. J.; Rice, C. M. Ultrastructural Analysis of Hepatitis C Virus Particles. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (23), 9505– 9510, DOI: 10.1073/pnas.1307527110Google Scholar126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFGrtrnF&md5=c71cc8d12de58b40c4d7a0785fec65fbUltrastructural analysis of hepatitis C virus particlesCatanese, Maria Teresa; Uryu, Kunihiro; Kopp, Martina; Edwards, Thomas J.; Andrus, Linda; Rice, William J.; Silvestry, Mariena; Kuhn, Richard J.; Rice, Charles M.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (23), 9505-9510, S9505/1-S9505/6CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Hepatitis C virus (HCV) is a major cause of chronic liver disease, with an estd. 170 million people infected worldwide. Low yields, poor stability, and inefficient binding to conventional EM grids have posed significant challenges to the purifn. and structural anal. of HCV. In this report, we generated an infectious HCV genome with an affinity tag fused to the E2 envelope glycoprotein. Using affinity grids, previously described to isolate proteins and macromol. complexes for single-particle EM, we were able to purify enveloped particles directly from cell culture media. This approach allowed for rapid in situ purifn. of virions and increased particle d. that were instrumental for cryo-EM and cryoelectron tomog. (cryo-ET). Moreover, it enabled ultrastructural anal. of virions produced by primary human hepatocytes. HCV appears to be the most structurally irregular member of the Flaviviridae family. Particles are spherical, with spike-like projections, and heterogeneous in size ranging from 40 to 100 nm in diam. Exosomes, although isolated from unfractionated culture media, were absent in highly infectious, purified virus prepns. Cryo-ET studies provided low-resoln. 3D structural information of highly infectious virions. In addn. to apolipoprotein (apo)E, HCV particles also incorporate apoB and apoA-I. In general, host apolipoproteins were more readily accessible to antibody labeling than HCV glycoproteins, suggesting either lower abundance or masking by host proteins.
- 127Echeverría, N.; Moratorio, G.; Cristina, J.; Moreno, P. Hepatitis C Virus Genetic Variability and Evolution. World J. Hepatol. 2015, 7 (6), 831, DOI: 10.4254/wjh.v7.i6.831Google Scholar127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2Mfgt1Kkug%253D%253D&md5=4b53b6079d254b5e81110dc97fb48fd7Hepatitis C virus genetic variability and evolutionEcheverria Natalia; Moratorio Gonzalo; Cristina Juan; Moreno PilarWorld journal of hepatology (2015), 7 (6), 831-45 ISSN:1948-5182.Hepatitis C virus (HCV) has infected over 170 million people worldwide and creates a huge disease burden due to chronic, progressive liver disease. HCV is a single-stranded, positive sense, RNA virus, member of the Flaviviridae family. The high error rate of RNA-dependent RNA polymerase and the pressure exerted by the host immune system, has driven the evolution of HCV into 7 different genotypes and more than 67 subtypes. HCV evolves by means of different mechanisms of genetic variation. On the one hand, its high mutation rates generate the production of a large number of different but closely related viral variants during infection, usually referred to as a quasispecies. The great quasispecies variability of HCV has also therapeutic implications since the continuous generation and selection of resistant or fitter variants within the quasispecies spectrum might allow viruses to escape control by antiviral drugs. On the other hand HCV exploits recombination to ensure its survival. This enormous viral diversity together with some host factors has made it difficult to control viral dispersal. Current treatment options involve pegylated interferon-α and ribavirin as dual therapy or in combination with a direct-acting antiviral drug, depending on the country. Despite all the efforts put into antiviral therapy studies, eradication of the virus or the development of a preventive vaccine has been unsuccessful so far. This review focuses on current available data reported to date on the genetic mechanisms driving the molecular evolution of HCV populations and its relation with the antiviral therapies designed to control HCV infection.
- 128Ghany, M. G.; Strader, D. B.; Thomas, D. L.; Seeff, L. B. Diagnosis, Management, and Treatment of Hepatitis C: An Update. Hepatology 2009, 49 (4), 1335, DOI: 10.1002/hep.22759Google Scholar128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXkslWjs7o%253D&md5=14f112636daff9f246fded6f6b2e23eeDiagnosis, management, and treatment of hepatitis C: an updateGhany, Marc G.; Strader, Doris B.; Thomas, David L.; Seeff, Leonard B.Hepatology (Hoboken, NJ, United States) (2009), 49 (4), 1335-1374CODEN: HPTLD9; ISSN:0270-9139. (John Wiley & Sons, Inc.)A review. This document aims to provide clinicians with evidence-based approaches to the prevention, diagnosis, and management of hepatitis C virus (HCV) infection. Current data on the recommended treatments of patients with chronic HCV infection are included. The sequential steps recommended for managing and treating persons chronically infected with hepatitis C for whom treatment is considered clearly appropriate are summarized.
- 129Zhu, Y. Z.; Qian, X. J.; Zhao, P.; Qi, Z. T. How Hepatitis C Virus Invades Hepatocytes: The Mystery of Viral Entry. World J. Gastroenterol. 2014, 20 (13), 3457– 3467, DOI: 10.3748/wjg.v20.i13.3457Google Scholar129https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhsVSlt7bF&md5=397b8e7d4111d1cf5fdd0399e1ae9abdHow hepatitis C virus invades hepatocytes: the mystery of viral entryZhu, Yong-Zhe; Qian, Xi-Jing; Zhao, Ping; Qi, Zhong-TianWorld Journal of Gastroenterology (2014), 20 (13), 3457-3467, 12CODEN: WJGAF2; ISSN:1007-9327. (Baishideng Publishing Group Co., Ltd.)A review. Hepatitis C virus (HCV) infection is a global health problem, with an estd. 170 million people being chronically infected. HCV cell entry is a complex multi-step process, involving several cellular factors that trigger virus uptake into the hepatocytes. The high- d. lipoprotein receptor scavenger receptor class B type I, tetraspanin CD81, tight junction protein claudin-1, and occludin are the main receptors that mediate the initial step of HCV infection. In addn., the virus uses cell receptor tyrosine kinases as entry regulators, such as epidermal growth factor receptor and ephrin receptor A2. This review summarizes the current understanding about how cell surface mols. are involved in HCV attachment, internalization, and membrane fusion, and how host cell kinases regulate virus entry. The advances of the potential antiviral agents targeting this process are introduced.
- 130Gu, M.; Rice, C. M. Structures of Hepatitis C Virus Nonstructural Proteins Required for Replicase Assembly and Function. Curr. Opin. Virol. 2013, 3 (2), 129– 136, DOI: 10.1016/j.coviro.2013.03.013Google Scholar130https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmsFaks7g%253D&md5=8d6f71bc4e8e5044afdd9fd1856b024cStructures of hepatitis C virus nonstructural proteins required for replicase assembly and functionGu, Meigang; Rice, Charles M.Current Opinion in Virology (2013), 3 (2), 129-136CODEN: COVUAF; ISSN:1879-6257. (Elsevier B. V.)A review. Approx. 3% of the world population is infected with hepatitis C virus (HCV), causing a serious public health burden. Like other pos.-strand RNA viruses, HCV assembles replicase complexes in assocn. with cellular membranes and produces progeny RNA genomes through neg.-strand intermediates. The viral proteins required for RNA replication are nonstructural (NS) proteins NS3 to NS5B. Owing to many obstacles and limitations in structural characterization of proteins and complexes with multiple transmembrane segments, attempts to understand the assembly and action of the HCV replicase complex have been challenging. Nevertheless, great progress has been made in obtaining structural information for several replicase components, providing insights into some aspects of the viral genome replication machinery.
- 131Pawlotsky, J. M. Hepatitis C Virus: Standard-of-Care Treatment. Adv. Pharmacol. 2013, 67, 169– 215, DOI: 10.1016/B978-0-12-405880-4.00005-6Google Scholar131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1Kku7vK&md5=7a1e40c7b665c61a9156189c2c5c0ff8Hepatitis C virus: standard-of-care treatmentPawlotsky, Jean-MichelAdvances in Pharmacology (San Diego, CA, United States) (2013), 67 (Antiviral Agents), 169-215CODEN: ADPHEL; ISSN:1054-3589. (Elsevier Inc.)A review. Hepatitis C virus (HCV) infection is curable by therapy. The antiviral treatment of chronic hepatitis C has been based for decades on the use of interferon (IFN)-α, combined with ribavirin. More recently, new therapeutic approaches that target essential components of the HCV life cycle have been developed, including direct-acting antiviral (DAA) and host-targeted agents (HTA). A new std.-of-care treatment has been approved in 2011 for patients infected with HCV genotype 1, based on a triple combination of pegylated IFN-α, ribavirin, and either telaprevir or boceprevir, two inhibitors of the HCV protease. New triple and quadruple combination therapies including pegylated IFN-α, ribavirin, and one or two DAAs/HTAs, resp., are currently being evaluated in Phase II and III clin. trials. In addn., various options for all-oral, IFN-free regimens are currently being evaluated. This chapter describes the characteristics of the different drugs used in the treatment of chronic hepatitis C and those currently in development and provides an overview of the current and future std.-of-care treatments of chronic hepatitis C.
- 132Bartenschlager, R.; Lohmann, V.; Penin, F. The Molecular and Structural Basis of Advanced Antiviral Therapy for Hepatitis C Virus Infection. Nat. Rev. Microbiol. 2013, 11 (7), 482– 496, DOI: 10.1038/nrmicro3046Google Scholar132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptVamtbs%253D&md5=3be643822234ebc9e4773c78ec9f3ee5The molecular and structural basis of advanced antiviral therapy for hepatitis C virus infectionBartenschlager, Ralf; Lohmann, Volker; Penin, FrancoisNature Reviews Microbiology (2013), 11 (7), 482-496CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)A review. The availability of the first mol. clone of the hepatitis C virus (HCV) genome allowed the identification and biochem. characterization of two viral enzymes that are targets for antiviral therapy: the protease NS3-4A and the RNA-dependent RNA polymerase NS5B. With the advent of cell culture systems that can recapitulate either the intracellular steps of the viral replication cycle or the complete cycle, addnl. drug targets have been identified, most notably the phosphoprotein NS5A, but also host cell factors that promote viral replication, such as cyclophilin A. Here, we review insights into the structures of these proteins and the mechanisms by which they contribute to the HCV replication cycle, and discuss how these insights have facilitated the development of new, directly acting antiviral compds. that have started to enter the clinic.
- 133Pawlotsky, J. M. What Are the Pros and Cons of the Use of Host-Targeted Agents against Hepatitis C?. Antiviral Res. 2014, 105 (1), 22, DOI: 10.1016/j.antiviral.2014.02.008Google Scholar133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmsVKksro%253D&md5=fc79a4183eb968a8579514d8274d82daWhat are the pros and cons of the use of host-targeted agents against hepatitis C?Pawlotsky, Jean-MichelAntiviral Research (2014), 105 (), 22-25CODEN: ARSRDR; ISSN:0166-3542. (Elsevier B.V.)A review. Hepatitis C virus (HCV) therapy is living a revolution. Host-targeted agents (HTAs) block HCV prodn. by interacting with host cell components. Because they target conserved host proteins, not variable viral proteins, HTAs have the potential for pangenotypic antiviral activity and a high barrier to resistance. Only two HTAs have reached clin. development, including specific inhibitors of cyclophilin A peptidyl-prolyl cis/trans isomerase activity and antagonists of microRNA-122. Cyclophilin inhibitors have proven to be relatively well tolerated and can be confidently used as backbones of all-oral, interferon-free regimens. In addn., HTAs such as cyclophilin inhibitors offer opportunities for "panviral" approaches when they target mechanisms common to viruses of the same or different families. This article forms part of a symposium in Antiviral Research on "Hepatitis C: next steps toward global eradication.".
- 134Scheel, T. K. H.; Rice, C. M. Understanding the Hepatitis C Virus Life Cycle Paves the Way for Highly Effective Therapies. Nat. Med. 2013, 19 (7), 837– 849, DOI: 10.1038/nm.3248Google Scholar134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVKru7rI&md5=dabc9fbc7dc0bc56115d5ede0d8ae072Understanding the hepatitis C virus life cycle paves the way for highly effective therapiesScheel, Troels K. H.; Rice, Charles M.Nature Medicine (New York, NY, United States) (2013), 19 (7), 837-849CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)A review. More than two decades of intense research has provided a detailed understanding of hepatitis C virus (HCV), which chronically infects 2% of the world's population. This effort has paved the way for the development of antiviral compds. to spare patients from life-threatening liver disease. An exciting new era in HCV therapy dawned with the recent approval of two viral protease inhibitors, used in combination with pegylated interferon-α and ribavirin; however, this is just the beginning. Multiple classes of antivirals with distinct targets promise highly efficient combinations, and interferon-free regimens with short treatment duration and fewer side effects are the future of HCV therapy. Ongoing and future trials will det. the best antiviral combinations and whether the current seemingly rich pipeline is sufficient for successful treatment of all patients in the face of major challenges, such as HCV diversity, viral resistance, the influence of host genetics, advanced liver disease and other co-morbidities.
- 135Bartenschlager, R.; Penin, F.; Lohmann, V.; André, P. Assembly of Infectious Hepatitis C Virus Particles. Trends Microbiol. 2011, 19 (2), 95– 103, DOI: 10.1016/j.tim.2010.11.005Google Scholar135https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhsFyrt7w%253D&md5=335be1300e42e4bd6f3bd0637b95839eAssembly of infectious hepatitis C virus particlesBartenschlager, Ralf; Penin, Francois; Lohmann, Volker; Andre, PatriceTrends in Microbiology (2011), 19 (2), 95-103CODEN: TRMIEA; ISSN:0966-842X. (Elsevier Ltd.)A review. A hallmark of the hepatitis C virus (HCV) replication cycle is its tight link with host cell lipid synthesis. This is best illustrated by the peculiar pathway used for the assembly of infectious HCV particles. Research in the past few years has shown that formation of HC-virions is closely connected to lipid droplets that could serve as an assembly platform. Moreover, HCV particle prodn. appears to be strictly linked to very-low-d. lipoproteins. Here, the authors focus on new insights into the mol. aspects of the architecture and assembly of this unique type of virus particle.
- 136Pennycuick, A.; Villanueva, A. Signaling in Hepatocellular Carcinoma. Signal. Pathways Liver Dis. Third Ed. 2015, 435– 447, DOI: 10.1002/9781118663387.ch32Google ScholarThere is no corresponding record for this reference.
- 137Ferguson, K. M. Structure-Based View of Epidermal Growth Factor Receptor Regulation. Annu. Rev. Biophys. 2008, 37, 353– 373, DOI: 10.1146/annurev.biophys.37.032807.125829Google Scholar137https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXnsVGlurs%253D&md5=bea3654114d360a02bbc934933a5eb11Structure-based view of epidermal growth factor receptor regulationFerguson, Kathryn M.Annual Review of Biophysics (2008), 37 (), 353-373CODEN: ARBNCV ISSN:. (Annual Reviews Inc.)A review. High-resoln. X-ray crystal structures detd. in the past six years dramatically influence our view of ligand-induced activation of the epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases. Ligand binding to the extracellular region of EGFR promotes a major domain reorganization, plus local conformational changes, that are required to generate an entirely receptor-mediated dimer. In this activated complex the intracellular kinase domains assoc. to form an asym. dimer that supports the allosteric activation of one kinase. These models are discussed with emphasis on recent studies that add details or bolster the generality of this view of activation of this family of receptors. The EGFR family is implicated in several disease states, perhaps most notably in cancers. Activating tumor mutations have been identified in the intracellular and extracellular regions of EGFR. The impact of these tumor mutations on the understanding of EGFR activation and of its inhibition is discussed.
- 138Citri, A.; Yarden, Y. EGF-ERBB Signalling: Towards the Systems Level. Nat. Rev. Mol. Cell Biol. 2006, 7 (7), 505– 516, DOI: 10.1038/nrm1962Google Scholar138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xms1OhsLc%253D&md5=0d41fd52d0498765a16b422d66398037EGF-ERBB signaling: towards the systems levelCitri, Ami; Yarden, YosefNature Reviews Molecular Cell Biology (2006), 7 (7), 505-516CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Signaling through the ErbB/HER receptors is intricately involved in human cancer and already serves as a target for several cancer drugs. Because of its inherent complexity, it is useful to envision ErbB signaling as a bow-tie-configured, evolvable network, which shares modularity, redundancy, and control circuits with robust biol. and engineered systems. Because network fragility is an inevitable trade-off of robustness, systems-level understanding is expected to generate therapeutic opportunities to intercept aberrant network activation.
- 139Yarden, Y.; Sliwkowski, M. X. Untangling the ErbB Signalling Network. Nat. Rev. Mol. Cell Biol. 2001, 2 (2), 127– 137, DOI: 10.1038/35052073Google Scholar139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXivVWnt7k%253D&md5=1561a9666f77fce888be821399870cd6Untangling the ErbB signalling networkYarden, Yosef; Sliwkowski, Mark X.Nature Reviews Molecular Cell Biology (2001), 2 (2), 127-137CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review, with 112 refs. When epidermal growth factor and its relatives bind the ErbB family of receptors, they trigger a rich network of signaling pathways, culminating in responses ranging from cell division to death, motility to adhesion. The network is often dysregulated in cancer and lends credence to the mantra that mol. understanding yields clin. benefit: over 25,000 women with breast cancer have now been treated with Herceptin, a recombinant antibody designed to block the receptor ErbB2. Likewise, small-mol. enzyme inhibitors and monoclonal antibodies to ErbB1 are in advanced phases of clin. testing. What can this pathway teach us about translating basic science into clin. use.
- 140Mendelsohn, J.; Baselga, J. Epidermal Growth Factor Receptor Targeting in Cancer. Semin. Oncol. 2006, 33 (4), 369– 385, DOI: 10.1053/j.seminoncol.2006.04.003Google Scholar140https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsVersLk%253D&md5=68ddafac141c8351630137c8b28ca74eEpidermal growth factor receptor targeting in cancerMendelsohn, John; Baselga, JoseSeminars in Oncology (2006), 33 (4), 369-385CODEN: SOLGAV; ISSN:0093-7754. (Elsevier Inc.)A review. The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase of the ErbB family that is abnormally activated in many epithelial tumors. Several mechanisms lead to the receptor's aberrant activation that is obsd. in cancer, including receptor overexpression, mutation, ligand-dependent receptor dimerization, and ligand-independent activation. Two classes of anti-EGFR agents are currently approved for the treatment of patients with cancer: cetuximab, a monoclonal antibody directed at the extracellular domain of the receptor, and gefitinib and erlotinib, oral, low-mol.-wt. (MW), ATP-competitive inhibitors of the receptor's tyrosine kinase. Anti-EGFR monoclonal antibodies have demonstrated activity in the therapy of advanced colorectal carcinoma and in a variety of epithelial tumor types, including head and neck cancer and non-small cell lung cancer (NSCLC). The development of low MW, anti-EGFR tyrosine kinase inhibitors (TKIs) has been focused until recently on NSCLC, although responses have been reported for other types of cancer. Erlotinib was the only agent approved based on demonstrating improved survival, which was obsd. in patients with advanced NSCLC who previously had been treated with chemotherapy. Recent major advances in the EGFR field include the discovery of EGFR somatic mutations in NSCLC that have important implications for biol., treatment, clin. trial design, and methods for mutation detection. Clin. and survival benefits with anti-EGFR agents have been demonstrated in addnl. tumor types such as head and neck and pancreatic carcinomas. New agents with clin. activity are entering the clinic and new combinatorial approaches with anti-EGFR agents are being explored. Major efforts are, belatedly, attempting to identify mol. markers that can predict patients more likely to respond to anti-EGFR therapy.
- 141Bublil, E. M.; Yarden, Y. The EGF Receptor Family: Spearheading a Merger of Signaling and Therapeutics. Curr. Opin. Cell Biol. 2007, 19 (2), 124– 134, DOI: 10.1016/j.ceb.2007.02.008Google Scholar141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXjt1Slu7w%253D&md5=66aa3dff3faaf8589186c20a71773785The EGF receptor family: spearheading a merger of signaling and therapeuticsBublil, Erez M.; Yarden, YosefCurrent Opinion in Cell Biology (2007), 19 (2), 124-134CODEN: COCBE3; ISSN:0955-0674. (Elsevier Ltd.)A review. The ErbB receptor tyrosine kinases evolved as key regulatory entities enabling the extracellular milieu to communicate with the intracellular machinery to bring forth the appropriate biol. response in an ever-changing environment. Since its discovery, many aspects of the ErbB family have been deciphered, with emphasis on aberration of signaling in human diseases. However, only now, with the availability of the at. coordinates of these receptors, can we construct a comprehensive model of the mechanisms underlying ligand-induced receptor dimerization and subsequent tyrosine kinase activation. Furthermore, the recent introduction of new high-throughput screening methodologies, combined with the materialization of a systems biol. perspective, reveals an overwhelming network complexity, enabling robust signaling and evolvability. This knowledge is likely to impact our view of diseases as system perturbations and resistance to ErbB-targeted therapeutics as manifestations of robustness.
- 142Wee, P.; Wang, Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers (Basel). 2017, 9 (5), 52, DOI: 10.3390/cancers9050052Google ScholarThere is no corresponding record for this reference.
- 143Fuchs, B. C.; Hoshida, Y.; Fujii, T.; Wei, L.; Yamada, S.; Lauwers, G. Y.; Mcginn, C. M.; Deperalta, D. K.; Chen, X.; Kuroda, T.; Lanuti, M.; Schmitt, A. D.; Gupta, S.; Crenshaw, A.; Onofrio, R.; Taylor, B.; Winckler, W.; Bardeesy, N.; Caravan, P.; Golub, T. R.; Tanabe, K. K. Epidermal Growth Factor Receptor Inhibition Attenuates Liver Fibrosis and Development of Hepatocellular Carcinoma. Hepatology 2014, 59 (4), 1577– 1590, DOI: 10.1002/hep.26898Google Scholar143https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXkvVGls78%253D&md5=c62c85e8e0835c5b257105716f371618Epidermal growth factor receptor inhibition attenuates liver fibrosis and development of hepatocellular carcinomaFuchs, Bryan C.; Hoshida, Yujin; Fujii, Tsutomu; Wei, Lan; Yamada, Suguru; Lauwers, Gregory Y.; McGinn, Christopher M.; DePeralta, Danielle K.; Chen, Xintong; Kuroda, Toshihiko; Lanuti, Michael; Schmitt, Anthony D.; Gupta, Supriya; Crenshaw, Andrew; Onofrio, Robert; Taylor, Bradley; Winckler, Wendy; Bardeesy, Nabeel; Caravan, Peter; Golub, Todd R.; Tanabe, Kenneth K.Hepatology (Hoboken, NJ, United States) (2014), 59 (4), 1577-1590CODEN: HPTLD9; ISSN:0270-9139. (John Wiley & Sons, Inc.)Hepatocellular carcinoma (HCC) is the most rapidly increasing cause of cancer-related mortality in the United States. Because of the lack of viable treatment options for HCC, prevention in high-risk patients has been proposed as an alternative strategy. The main risk factor for HCC is cirrhosis and several lines of evidence implicate epidermal growth factor (EGF) in the progression of cirrhosis and development of HCC. We therefore examd. the effects of the EGF receptor (EGFR) inhibitor erlotinib on liver fibrogenesis and hepatocellular transformation in three different animal models of progressive cirrhosis: a rat model induced by repeated, low-dose injections of diethylnitrosamine (DEN), a mouse model induced by carbon tetrachloride (CCl4), and a rat model induced by bile duct ligation (BDL). Erlotinib reduced EGFR phosphorylation in hepatic stellate cells (HSC) and reduced the total no. of activated HSC. Erlotinib also decreased hepatocyte proliferation and liver injury. Consistent with all these findings, pharmacol. inhibition of EGFR signaling effectively prevented the progression of cirrhosis and regressed fibrosis in some animals. Moreover, by alleviating the underlying liver disease, erlotinib blocked the development of HCC and its therapeutic efficacy could be monitored with a previously reported gene expression signature predictive of HCC risk in human cirrhosis patients. Conclusion: These data suggest that EGFR inhibition using Food and Drug Administration-approved inhibitors provides a promising therapeutic approach for redn. of fibrogenesis and prevention of HCC in high-risk cirrhosis patients who can be identified and monitored by gene expression signatures. (Hepatol. 2014;59:1577-1590).
- 144Huang, W. C.; Chan, S. H.; Jang, T. H.; Chang, J. W.; Ko, Y. C.; Yen, T. C.; Chiang, S. L.; Chiang, W. F.; Shieh, T. Y.; Liao, C. T.; Juang, J. L.; Wang, H. C.; Cheng, A. J.; Lu, Y. C.; Wang, L. H. MiRNA-491–5p and GIT1 Serve as Modulators and Biomarkers for Oral Squamous Cell Carcinoma Invasion and Metastasis. Cancer Res. 2014, 74 (3), 751– 764, DOI: 10.1158/0008-5472.CAN-13-1297Google Scholar144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvVSms7c%253D&md5=bca58051e3f96eceee5ff99018d72fa6miRNA-491-5p and GIT1 Serve as Modulators and Biomarkers for Oral Squamous Cell Carcinoma Invasion and MetastasisHuang, Wei-Chieh; Chan, Shih-Hsuan; Jang, Te-Hsuan; Chang, Jer-Wei; Ko, Ying-Chin; Yen, Tzu-Chen; Chiang, Shang-Lun; Chiang, Wei-Fan; Shieh, Tien-Yu; Liao, Chun-Ta; Juang, Jyh-Lyh; Wang, Hsueh-Chun; Cheng, Ann-Joy; Lu, Ya-Ching; Wang, Lu-HaiCancer Research (2014), 74 (3), 751-764CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)MicroRNAs offer tools to identify and treat invasive cancers. Using highly invasive isogenic oral squamous cell carcinoma (OSCC) cells, established using in vitro and in vivo selection protocols from poorly invasive parental cell populations, we used microarray expression anal. to identify a relative and specific decrease in miR-491-5p in invasive cells. Lower expression of miR-491-5p correlated with poor overall survival of patients with OSCCs. MiR- 491-5p overexpression in invasive OSCC cells suppressed their migratory behavior in vitro and lung metastatic behavior in vivo. We defined the G-proteincoupled receptor kinase-interacting protein I (GITl)as a direct target gene for miR-491-5p control. GITl overexpression was sufficient to rescue miR-491-5p-mediated inhibition of migration/invasion and lung metastasis. Conversely, GITl silencing phenocopied the ability of miR-491-5p to inhibit migration/invasion and metastasis of OSCC cells. Mechanistic investigations indicated that miR-491-5p overexpression or GITl attenuation reduced focal adhesions, with a concurrent decrease in steady-state levels of paxillin, phospho-paxillin, phospho-FAK, EGF/EGFR-mediated extracellular signal-regulated kinase (ERK1/2) activation, and MMP2/9 levels and activities. In clin. specimens of OSCCs, GITl levels were elevated relative to paired normal tissues and were correlated with lymph node metastasis, with expression levels of miR-491-5p and GITI correlated inversely in OSCCs, where they informed tumor grade. Together, our findings identify a functional axis for OSCC invasion that suggests miR-491-5p and GITl as biomarkers for prognosis in this cancer.
- 145Zheng, K.; Kitazato, K.; Wang, Y. Viruses Exploit the Function of Epidermal Growth Factor Receptor. Rev. Med. Virol. 2014, 24 (4), 274– 286, DOI: 10.1002/rmv.1796Google Scholar145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtV2mu7vO&md5=a58081a6fa69c3d26937ef6acb1d0222Viruses exploit the function of epidermal growth factor receptorZheng, Kai; Kitazato, Kaio; Wang, YifeiReviews in Medical Virology (2014), 24 (4), 274-286CODEN: RMVIEW; ISSN:1052-9276. (John Wiley & Sons Ltd.)A review. SUMMARY : Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that regulates cellular homeostatic processes. Following ligand binding, EGFR activates different downstream signalling cascades that promote cell survival, proliferation, motility, and angiogenesis and induces F-actin-dependent EGFR endocytosis, which relocalizes the activated receptors for degrdn. or recycling. The responses that are induced by ligand binding to EGFR, including cell signalling activation, protein kinase phosphorylation and cytoskeletal network rearrangement, resemble those induced by virus infection. Increasing evidence demonstrates that many viruses usurp EGFR endocytosis or EGFR-mediated signalling for entry, replication, inflammation, and viral antagonism to the host antiviral system. In addn., viruses have acquired sophisticated mechanisms to regulate EGFR functions by interrupting the EGFR-recycling process and modulating EGFR expression. In this review, we provide an overview of the mechanisms by which viruses alter EGFR signalling in favor of their continued survival.
- 146Lupberger, J.; Zeisel, M. B.; Xiao, F.; Thumann, C.; Fofana, I.; Zona, L.; Davis, C.; Mee, C. J.; Turek, M.; Gorke, S.; Royer, C.; Fischer, B.; Zahid, M. N.; Lavillette, D.; Fresquet, J.; Cosset, F. L.; Rothenberg, S. M.; Pietschmann, T.; Patel, A. H.; Pessaux, P.; Dofföl, M.; Raffelsberger, W.; Poch, O.; McKeating, J. A.; Brino, L.; Baumert, T. F. EGFR and EphA2 Are Host Factors for Hepatitis C Virus Entry and Possible Targets for Antiviral Therapy. Nat. Med. 2011, 17 (5), 589– 595, DOI: 10.1038/nm.2341Google Scholar146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXltFyqtL4%253D&md5=9a772f9cb03077335c7e7f5918a6e1efEGFR and EphA2 are host factors for hepatitis C virus entry and possible targets for antiviral therapyLupberger, Joachim; Zeisel, Mirjam B.; Xiao, Fei; Thumann, Christine; Fofana, Isabel; Zona, Laetitia; Davis, Christopher; Mee, Christopher J.; Turek, Marine; Gorke, Sebastian; Royer, Cathy; Fischer, Benoit; Zahid, Muhammad N.; Lavillette, Dimitri; Fresquet, Judith; Cosset, Francois-Loic; Rothenberg, S. Michael; Pietschmann, Thomas; Patel, Arvind H.; Pessaux, Patrick; Doffoel, Michel; Raffelsberger, Wolfgang; Poch, Olivier; McKeating, Jane A.; Brino, Laurent; Baumert, Thomas F.Nature Medicine (New York, NY, United States) (2011), 17 (5), 589-595CODEN: NAMEFI; ISSN:1078-8956. (Nature Publishing Group)Hepatitis C virus (HCV) is a major cause of liver disease, but therapeutic options are limited and there are no prevention strategies. Viral entry is the first step of infection and requires the cooperative interaction of several host cell factors. Using a functional RNAi kinase screen, we identified epidermal growth factor receptor and ephrin receptor A2 as host cofactors for HCV entry. Blocking receptor kinase activity by approved inhibitors broadly impaired infection by all major HCV genotypes and viral escape variants in cell culture and in a human liver chimeric mouse model in vivo. The identified receptor tyrosine kinases (RTKs) mediate HCV entry by regulating CD81-claudin-1 co-receptor assocns. and viral glycoprotein-dependent membrane fusion. These results identify RTKs as previously unknown HCV entry cofactors and show that tyrosine kinase inhibitors have substantial antiviral activity. Inhibition of RTK function may constitute a new approach for prevention and treatment of HCV infection.
- 147Zona, L.; Lupberger, J.; Sidahmed-Adrar, N.; Thumann, C.; Harris, H. J.; Barnes, A.; Florentin, J.; Tawar, R. G.; Xiao, F.; Turek, M.; Durand, S. C.; Duong, F. H. T.; Heim, M. H.; Cosset, F. L.; Hirsch, I.; Samuel, D.; Brino, L.; Zeisel, M. B.; Le Naour, F.; McKeating, J. A.; Baumert, T. F. HRas Signal Transduction Promotes Hepatitis C Virus Cell Entry by Triggering Assembly of the Host Tetraspanin Receptor Complex. Cell Host Microbe 2013, 13 (3), 302– 313, DOI: 10.1016/j.chom.2013.02.006Google Scholar147https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXktFWgtL4%253D&md5=07cc5c59baae9a28ae84993fdf1a9785HRas Signal Transduction Promotes Hepatitis C Virus Cell Entry by Triggering Assembly of the Host Tetraspanin Receptor ComplexZona, Laetitia; Lupberger, Joachim; Sidahmed-Adrar, Nazha; Thumann, Christine; Harris, Helen J.; Barnes, Amy; Florentin, Jonathan; Tawar, Rajiv G.; Xiao, Fei; Turek, Marine; Durand, Sarah C.; Duong, Francois H. T.; Heim, Markus H.; Cosset, Francois-Loic; Hirsch, Ivan; Samuel, Didier; Brino, Laurent; Zeisel, Mirjam B.; Le Naour, Francois; McKeating, Jane A.; Baumert, Thomas F.Cell Host & Microbe (2013), 13 (3), 302-313CODEN: CHMECB; ISSN:1931-3128. (Elsevier Inc.)Hepatitis C virus (HCV) entry is dependent on coreceptor complex formation between the tetraspanin superfamily member CD81 and the tight junction protein claudin-1 (CLDN1) on the host cell membrane. The receptor tyrosine kinase EGFR acts as a cofactor for HCV entry by promoting CD81-CLDN1 complex formation via unknown mechanisms. We identify the GTPase HRas, activated downstream of EGFR signaling, as a key host signal transducer for EGFR-mediated HCV entry. Proteomic anal. revealed that HRas assocs. with tetraspanin CD81, CLDN1, and the previously unrecognized HCV entry cofactors integrin β1 and Ras-related protein Rap2B in hepatocyte membranes. HRas signaling is required for lateral membrane diffusion of CD81, which enables tetraspanin receptor complex assembly. HRas was also found to be relevant for entry of other viruses, including influenza. Our data demonstrate that viruses exploit HRas signaling for cellular entry by compartmentalization of entry factors and receptor trafficking.
- 148Baktash, Y.; Madhav, A.; Coller, K. E.; Randall, G. Single Particle Imaging of Polarized Hepatoma Organoids upon Hepatitis C Virus Infection Reveals an Ordered and Sequential Entry Process. Cell Host Microbe 2018, 23 (3), 382– 394, e5 DOI: 10.1016/j.chom.2018.02.005Google Scholar148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslSmsLk%253D&md5=0678419260c3f14266ab3e8fd40610ceSingle Particle Imaging of Polarized Hepatoma Organoids upon Hepatitis C Virus Infection Reveals an Ordered and Sequential Entry ProcessBaktash, Yasmine; Madhav, Anisha; Coller, Kelly E.; Randall, GlennCell Host & Microbe (2018), 23 (3), 382-394.e5CODEN: CHMECB; ISSN:1931-3128. (Elsevier Inc.)Hepatitis C virus (HCV) enters hepatocytes via various entry factors, including scavenger receptor BI (SR-B1), cluster of differentiation 81 (CD81), epidermal growth factor receptor (EGFR), claudin-1 (CLDN1), and occludin (OCLN). As CLDN1 and OCLN are not readily accessible due to their tight junctional localization, HCV likely accesses them by either disrupting cellular polarity or migrating to the tight junction. In this study, we image HCV entry into a three-dimensional polarized hepatoma system and reveal that the virus sequentially engages these entry factors through actin-dependent mechanisms. HCV initially localizes with the early entry factors SR-B1, CD81, and EGFR at the basolateral membrane and then accumulates at the tight junction in an actin-dependent manner. HCV assocs. with CLDN1 and then OCLN at the tight junction and is internalized via clathrin-mediated endocytosis by an active process requiring EGFR. Thus, HCV uses a dynamic and multi-step process to engage and enter host cells.
- 149Diao, J.; Pantua, H.; Ngu, H.; Komuves, L.; Diehl, L.; Schaefer, G.; Kapadia, S. B. Hepatitis C Virus Induces Epidermal Growth Factor Receptor Activation via CD81 Binding for Viral Internalization and Entry. J. Virol. 2012, 86 (20), 10935, DOI: 10.1128/JVI.00750-12Google Scholar149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVyru7bK&md5=514c49e7f64fca75b2fe4d3643615bd5Hepatitis C virus induces epidermal growth factor receptor activation via CD81 binding for viral internalization and entryDiao, Jingyu; Pantua, Homer; Ngu, Hai; Komuves, Laszlo; Diehl, Lauri; Schaefer, Gabriele; Kapadia, Sharookh B.Journal of Virology (2012), 86 (20), 10935-10949CODEN: JOVIAM; ISSN:0022-538X. (American Society for Microbiology)While epidermal growth factor receptor (EGFR) has been shown to be important in the entry process for multiple viruses, including hepatitis C virus (HCV), the mol. mechanisms by which EGFR facilitates HCV entry are not well understood. Using the infectious cell culture HCV model (HCVcc), we demonstrate that the binding of HCVcc particles to human hepatocyte cells induces EGFR activation that is dependent on interactions between HCV and CD81 but not claudin 1. EGFR activation can also be induced by antibody mediated crosslinking of CD81. In addn., EGFR ligands that enhance the kinetics of HCV entry induce EGFR internalization and colocalization with CD81. While EGFR kinase inhibitors inhibit HCV infection primarily by preventing EGFR endocytosis, antibodies that block EGFR ligand binding or inhibitors of EGFR downstream signaling have no effect on HCV entry. These data demonstrate that EGFR internalization is crit. for HCV entry and identify a hitherto-unknown assocn. between CD81 and EGFR.
- 150Mailly, L.; Xiao, F.; Lupberger, J.; Wilson, G. K.; Aubert, P.; Duong, F. H. T.; Calabrese, D.; Leboeuf, C.; Fofana, I.; Thumann, C.; Bandiera, S.; Lütgehetmann, M.; Volz, T.; Davis, C.; Harris, H. J.; Mee, C. J.; Girardi, E.; Chane-Woon-Ming, B.; Ericsson, M.; Fletcher, N.; Bartenschlager, R.; Pessaux, P.; Vercauteren, K.; Meuleman, P.; Villa, P.; Kaderali, L.; Pfeffer, S.; Heim, M. H.; Neunlist, M.; Zeisel, M. B.; Dandri, M.; McKeating, J. A.; Robinet, E.; Baumert, T. F. Clearance of Persistent Hepatitis C Virus Infection in Humanized Mice Using a Claudin-1-Targeting Monoclonal Antibody. Nat. Biotechnol. 2015, 33 (5), 549– 554, DOI: 10.1038/nbt.3179Google Scholar150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXkvFejuro%253D&md5=98a4d73f2b7bca389e822633d467c6fdClearance of persistent hepatitis C virus infection in humanized mice using a claudin-1-targeting monoclonal antibodyMailly, Laurent; Xiao, Fei; Lupberger, Joachim; Wilson, Garrick K.; Aubert, Philippe; Duong, Francois H. T.; Calabrese, Diego; Leboeuf, Celine; Fofana, Isabel; Thumann, Christine; Bandiera, Simonetta; Lutgehetmann, Marc; Volz, Tassilo; Davis, Christopher; Harris, Helen J.; Mee, Christopher J.; Girardi, Erika; Chane-Woon-Ming, Beatrice; Ericsson, Maria; Fletcher, Nicola; Bartenschlager, Ralf; Pessaux, Patrick; Vercauteren, Koen; Meuleman, Philip; Villa, Pascal; Kaderali, Lars; Pfeffer, Sebastien; Heim, Markus H.; Neunlist, Michel; Zeisel, Mirjam B.; Dandri, Maura; McKeating, Jane A.; Robinet, Eric; Baumert, Thomas F.Nature Biotechnology (2015), 33 (5), 549-554CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Hepatitis C virus (HCV) infection is a leading cause of liver cirrhosis and cancer. Cell entry of HCV and other pathogens is mediated by tight junction (TJ) proteins, but successful therapeutic targeting of TJ proteins has not been reported yet. Using a human liver-chimeric mouse model, we show that a monoclonal antibody specific for the TJ protein claudin-1 (ref. 7) eliminates chronic HCV infection without detectable toxicity. This antibody inhibits HCV entry, cell-cell transmission and virus-induced signaling events. Antibody treatment reduces the no. of HCV-infected hepatocytes in vivo, highlighting the need for de novo infection by means of host entry factors to maintain chronic infection. In summary, we demonstrate that an antibody targeting a virus receptor can cure chronic viral infection and uncover TJ proteins as targets for antiviral therapy.
- 151Igloi, Z.; Kazlauskas, A.; Saksela, K.; MacDonald, A.; Mankouri, J.; Harris, M. Hepatitis C Virus NS5A Protein Blocks Epidermal Growth Factor Receptor Degradation via a Proline Motif- Dependent Interaction. J. Gen. Virol. 2015, 96 (8), 2133– 2144, DOI: 10.1099/vir.0.000145Google Scholar151https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XjsFOlu70%253D&md5=7d04ae73adc61e3cd2bfa4cd98159980Hepatitis C virus NS5A protein blocks epidermal growth factor receptor degradation via a proline motif- dependent interactionIgloi, Zsofia; Kazlauskas, Arunas; Saksela, Kalle; MacDonald, Andrew; Mankouri, Jamel; Harris, MarkJournal of General Virology (2015), 96 (8), 2133-2144CODEN: JGVIAY; ISSN:0022-1317. (Society for General Microbiology)Hepatitis C virus (HCV) establishes a persistent infection that in many cases leads to cirrhosis and hepatocellular carcinoma. The non-structural 5A protein (NS5A) has been implicated in this process as it contains a C-terminal polyproline motif (termed P2) that binds to Src homol. 3 (SH3) domains to regulate cellular signalling and trafficking pathways. We have shown previously that NS5A impaired epidermal growth factor (EGF) receptor (EGFR) endocytosis, thereby inhibiting EGF-stimulated EGFR degrdn. by a mechanism that remained unclear. As EGFR has been implicated in HCV cell entry and trafficking of the receptor involves several SH3-domain contg. proteins, we investigated in more detail the mechanisms by which NS5A perturbs EGFR trafficking. We demonstrated that the P2 motif was required for the NS5A-mediated disruption to EGFR trafficking. We further demonstrated that the P2 motif was required for an interaction between NS5A and CMS, a homolog of CIN85 that has previously been implicated in EGFR endocytosis. We provided evidence that CMS was involved in the NS5A-mediated perturbation of EGFR trafficking. We also showed that NS5A effected a loss of EGFR ubiquitination in a P2-motif-dependent fashion. These data provide clues to the mechanism by which NS5A regulates the trafficking of a key cellular receptor and demonstrate for the first time the ability of NS5A to regulate host cell ubiquitination pathways.
- 152Xu, X.; Zheng, L.; Yuan, Q.; Zhen, G.; Crane, J. L.; Zhou, X.; Cao, X. Transforming Growth Factor-β in Stem Cells and Tissue Homeostasis. Bone Res. 2018, 6 (1), 2, DOI: 10.1038/s41413-017-0005-4Google Scholar152https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mvps1Wksg%253D%253D&md5=4f0e14ef7a37bd3373b89fc70bdc1312Transforming growth factor-β in stem cells and tissue homeostasisXu Xin; Zhou Xuedong; Zheng Liwei; Yuan Quan; Zhen Gehua; Crane Janet L; Cao Xu; Crane Janet LBone research (2018), 6 (), 2 ISSN:2095-4700.TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
- 153Chaudhury, A.; Howe, P. H. The Tale of Transforming Growth Factor-β (TGFβ) Signaling: A Soigné Enigma. IUBMB Life 2009, 61 (10), 929, DOI: 10.1002/iub.239Google Scholar153https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1WntbbL&md5=a89c0c3fdc9fe520452d861232e0f90dThe tale of transforming growth factor-beta (TGFβ) signaling: A soigne enigmaChaudhury, Arindam; Howe, Philip H.IUBMB Life (2009), 61 (10), 929-939CODEN: IULIF8; ISSN:1521-6543. (John Wiley & Sons Inc.)A review. Transforming growth factor-beta (TGFβ) is a secreted cytokine, which intricately controls a plethora of physiol. and pathol. processes during development and carcinogenesis. TGFβ exerts antiproliferative effects and functions as a tumor suppressor during early stages of tumorigenesis, whereas at later stages it functions as a tumor promoter aiding in metastatic progression through an autocrine TGFβ loop. Intricate knowledge of TGFβ signaling and its regulation are still evolving. In this review, we make an attempt to showcase the assocd. enigma of TGFβ signaling in its dual functional role as tumor suppressor and metastatic promoter during early and late stages of carcinogenesis, resp. © 2009 IUBMB IUBMB Life, 61(10): 929-939, 2009.
- 154Miyazawa, K.; Shinozaki, M.; Hara, T.; Furuya, T.; Miyazono, K. Two Major Smad Pathways in TGF-Beta Superfamily Signalling. Genes Cells 2002, 7 (12), 1191– 1204, DOI: 10.1046/j.1365-2443.2002.00599.xGoogle Scholar154Two major Smad pathways in TGF-β superfamily signallingMiyazawa, Keiji; Shinozaki, Masahiko; Hara, Takane; Furuya, Toshio; Miyazono, KoheiGenes to Cells (2002), 7 (12), 1191-1204CODEN: GECEFL; ISSN:1356-9597. (Blackwell Science Ltd.)A review. Members of the transforming growth factor-β (TGF-β) superfamily bind to two different serine/threonine kinase receptors, i.e., type I and type II receptors. Upon ligand binding, type I receptors specifically activate intracellular Smad proteins. R-Smads are direct substrates of type I receptors; Smads 2 and 3 are specifically activated by activin/nodal and TGF-β type I receptors, whereas Smads 1, 5 and 8 are activated by BMP type I receptors. Nearly 30 proteins have been identified as members of the TGF-β superfamily in mammals, and can be classified based on whether they activate activin/TGF-β-specific R-Smads (AR-Smads) or BMP-specific R-Smads (BR-Smads). R-Smads form complexes with Co-Smads and translocate into the nucleus, where they regulate the transcription of target genes. AR-Smads bind to various proteins, including transcription factors and transcriptional co-activators or co-repressors, whereas BR-Smads interact with other proteins less efficiently than AR-Smads. Id proteins are induced by BR-Smads, and play important roles in exhibiting some biol. effects of BMPs. Understanding the mechanisms of TGF-β superfamily signaling is thus important for the devel