J. M. Taylor, Virology of hepatitis D virus, Semin Liver Dis, vol.32, pp.195-200, 2012.

. Fattovich-g and . Christensen-e, influence of hepatitis delta virus infection on morbidity and mortality in compensated cirrhosis type B. the european concerted action on viral hepatitis (eurohep), Gut, vol.46, pp.420-426, 2000.

H. Sa, H. Wedemeyer, and P. M. Harrison, Hepatitis delta virus, The Lancet, vol.378, pp.73-85, 2011.

, rizzetto M. investigational drugs in development for Hepatitis D, vol.26, pp.999-1005, 2017.

, Sureau c, negro F. the hepatitis delta virus: replication and pathogenesis, J Hepatol, vol.64, pp.102-118, 2016.

. Webb, Kinetic parameters of trans scission by extended hdv-like ribozymes and the prospect for the discovery of genomic trans-cleaving rnas, Biochemistry, vol.57, pp.1440-50, 2018.

. Verrier and . Bach-c, a targeted functional rna interference screen uncovers glypican 5 as an entry factor for hepatitis B and D viruses, Hepatology, vol.63, pp.35-48, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01796197

. Verrier-er, . Colpitts-cc, and . Sureau-c, Hepatitis B virus receptors and molecular drug targets, Hepatol Int, vol.10, pp.567-73, 2016.

. Baumert-tf and M. Verrier-er, Host-targeting agents for treatment of hepatitis B virus infection, Curr Opin Virol, vol.14, pp.41-47, 2015.

. Colpitts-cc and E. Verrier, Baumert tF. targeting viral entry for treatment of Hepatitis B and c virus infections, ACS Infect Dis, vol.1, pp.420-427, 2015.

. Harak-c, M. Meyrath, and . Romero-brey-i, tuning a cellular lipid kinase activity adapts hepatitis c virus to replication in cell culture, Nat Microbiol, vol.2, p.16247, 2016.

. Mailly-l, F. Xiao, and J. Lupberger, clearance of persistent hepatitis c virus infection in humanized mice using a claudin-1-targeting monoclonal antibody, Nat Biotechnol, vol.33, pp.549-54, 2015.

. Koh-c and H. Dahari, Oral prenylation inhibition with lonafarnib in chronic hepatitis D infection: a proof-of-concept randomised, double-blind, placebo-controlled phase 2a trial, Lancet Infect Dis, vol.15, pp.1167-74, 2015.

P. Bogomolov and . Voronkova-n, treatment of chronic hepatitis D with the entry inhibitor myrcludex B: first results of a phase ib/iia study, J Hepatol, vol.65, pp.490-498, 2016.

Q. Li, ng a, et al. a genome-wide genetic screen for host factors required for hepatitis c virus propagation, Proc Natl Acad Sci U S A, vol.106, pp.16410-16415, 2009.

D. M. Dykxhoorn and Y. Benita, identification of host proteins required for HiV infection through a functional genomic screen, Science, vol.319, pp.921-927, 2008.

M. Busse-wicher and K. B. Wicher, Kusche-gullberg M. the exostosin family: proteins with many functions, Matrix Biol, vol.35, pp.25-33, 2014.

K. Sharma-n, rpb4 and rpb7: multifunctional subunits of rna polymerase ii, Crit Rev Microbiol, vol.39, pp.362-72, 2013.

M. W. Hull, K. Mckune, and W. Na, rna polymerase ii subunit rPB9 is required for accurate start site selection, Genes Dev, vol.9, pp.481-90, 1995.

. Kraus-rj, X. Yu, and . Cordes-ba, Hypoxia-inducible factor-1? plays roles in epstein-Barr virus's natural life cycle and tumorigenesis by inducing lytic infection through direct binding to the immediate-early BZlF1 gene promoter, PLoS Pathog, vol.13, p.1006404, 2017.

F. Morinet, M. Parent, and S. Pillet, Hypoxia inducible factor one alpha and human viral pathogens, Curr Res Transl Med, vol.65, pp.7-9, 2017.

D. K. Chiu and . Xu-im, Hypoxia inducible factor HiF-1 promotes myeloidderived suppressor cells accumulation through entPD2/cD39l1 in hepatocellular carcinoma, Nat Commun, vol.8, p.517, 2017.

D. Luo, Z. Wang, and J. Wu, the role of Hypoxia inducible Factor-1 in Hepatocellular carcinoma, Biomed Res Int, vol.2014, pp.1-11, 2014.

, on October 15, 2019 at INSERM Consortia. Protected by copyright, 2019.

. Verrier-er, Gut, vol.0, pp.1-10, 2019.

, Hepatology

D. Lin and J. Wu, Hypoxia inducible factor in hepatocellular carcinoma: a therapeutic target, World J Gastroenterol, vol.21, pp.12171-12179, 2015.

. Evans-dr and H. Guy, Mammalian pyrimidine biosynthesis: fresh insights into an ancient pathway, J Biol Chem, vol.279, pp.33035-33043, 2004.

M. Löffler, . Fairbanks-ld, and . Zameitat-e, Pyrimidine pathways in health and disease, Trends Mol Med, vol.11, pp.430-437, 2005.

Y. Wang, Z. Wang, and . Liu-t, effects of flanking regions on HDV cotranscriptional folding kinetics, RNA, vol.24, pp.1229-1269, 2018.

. Swyryd-ea and S. S. Seaver, Stark gr. n-(phosphonacetyl)-l-aspartate, a potent transition state analog inhibitor of aspartate transcarbamylase, blocks proliferation of mammalian cells in culture, J Biol Chem, vol.249, pp.6945-50, 1974.

M. Lucas-hourani, D. Dauzonne, and P. Jorda, inhibition of pyrimidine biosynthesis pathway suppresses viral growth through innate immunity, PLoS Pathog, vol.9, p.1003678, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01113535

. Reyes-gr, ribavirin: recent insights into antiviral mechanisms of action, Curr Opin Drug Discov Devel, vol.4, pp.651-657, 2001.

M. S. Sulkowski, . Cooper-c, and B. Hunyady, Management of adverse effects of Peg-iFn and ribavirin therapy for hepatitis c, Nat Rev Gastroenterol Hepatol, vol.8, pp.212-235, 2011.

F. Perry-cm, M. Je, and . Pl, nelfinavir: a review of its use in the management of HiV infection, Drugs, vol.65, pp.2209-2253, 2005.

. Jeselsohn-r and . Buchwalter, De angelis c, et al. eSr1 mutations-a mechanism for acquired endocrine resistance in breast cancer, Nat Rev Clin Oncol, vol.12, pp.573-83, 2015.

J. Ellmén, P. Hakulinen, and . Partanen-a, estrogenic effects of toremifene and tamoxifen in postmenopausal breast cancer patients, Breast Cancer Res Treat, vol.82, pp.103-114, 2003.

D. Connolly-gp and . Ja, Uridine and its nucleotides: biological actions, therapeutic potentials, Trends Pharmacol Sci, vol.20, pp.218-243, 1999.

. Kelly-re and . Pastra-landis-sc, Oligomeric structure of the multifunctional protein caD that initiates pyrimidine biosynthesis in mammalian cells, Proc Natl Acad Sci U S A, vol.82, pp.6802-6808, 1985.

. Deans-rm, D. W. Morgens, and . Ökesli-a, Parallel shrna and criSPr-cas9 screens enable antiviral drug target identification, Nat Chem Biol, vol.12, pp.361-367, 2016.

D. M. Franks and H. Kitagawa, elegans pharyngeal morphogenesis requires both de novo synthesis of pyrimidines and synthesis of heparan sulfate proteoglycans, Dev Biol, vol.296, pp.409-429, 2006.

M. Droog, M. Mensink, and W. Zwart, the estrogen receptor ?-cistrome beyond breast cancer, Mol Endocrinol, vol.30, pp.1046-58, 2016.

V. Loustaud-ratti, M. Debette-gratien, and J. Jacques, ribavirin: past, present and future, World J Hepatol, vol.8, pp.123-153, 2016.

Y. Debing, E. Su, and Y. Wang, ribavirin inhibits in vitro hepatitis e virus replication through depletion of cellular gtP pools and is moderately synergistic with alpha interferon, Antimicrob Agents Chemother, vol.58, pp.267-73, 2014.

M. Levrero, B. Zoulim, and F. , HBV cure: why, how, when?, Curr Opin Virol, vol.18, pp.135-178, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01797257

M. B. Zeisel and . Baumert-tf, clinical development of hepatitis c virus host-targeting agents, Lancet, vol.389, pp.674-679, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01910174

R. Van-der, . Mh, J. M. De-vree, and F. Stelma, Safety, tolerability, and antiviral effect of rg-101 in patients with chronic hepatitis c: a phase 1B, double-blind, randomised controlled trial, Lancet, vol.389, pp.709-726, 2017.

M. Zhang-c,-chu, leflunomide: a promising drug with good antitumor potential, Biochem Biophys Res Commun, vol.496, pp.726-756, 2018.

. Bar-or-a, . Pachner-a, and F. Menguy-vacheron, teriflunomide and its mechanism of action in multiple sclerosis, Drugs, vol.74, pp.659-74, 2014.

P. Baumann, S. Mandl-weber, and . Volkl-a, Dihydroorotate dehydrogenase inhibitor a771726 (leflunomide) induces apoptosis and diminishes proliferation of multiple myeloma cells, Mol Cancer Ther, vol.8, pp.366-75, 2009.

J. Lupberger, M. B. Zeisel, and F. Xiao, egFr and epha2 are host factors for hepatitis c virus entry and possible targets for antiviral therapy, Nat Med, vol.17, pp.589-95, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00705829

Y. Verrier-er, . Sa, and . Heydmann-l, Hepatitis B virus evasion from cyclic guanosine monophosphate-adenosine monophosphate synthase sensing in human Hepatocytes, Hepatology, vol.68, pp.1695-709, 2018.

. Verrier and . Bach-c, Solute carrier ntcP regulates innate antiviral immune responses targeting Hepatitis c virus infection of hepatocytes, Cell Rep, vol.17, pp.1357-68, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01911211

, on October 15, 2019 at INSERM Consortia. Protected by copyright, 2019.