Review Open Access
Copyright ©2009 Baishideng. All rights reserved.
World J Hepatol. Oct 31, 2009; 1(1): 67-71
Published online Oct 31, 2009. doi: 10.4254/wjh.v1.i1.67
Hepatitis C virus and peripheral blood mononuclear cell reservoirs Patricia Baré
Patricia Baré, Virology Laboratory, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires 1425, Argentina
Author contributions: Baré P solely contributed to this paper.
Correspondence to: Patricia Baré, PhD, Virology Laboratory, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Pacheco de Melo, Buenos Aires 3081, Argentina. pbare@hematologia.anm.edu.ar
Telephone: +54-11-48050712 Fax: +54-11-48039475
Received: March 12, 2009
Revised: September 2, 2009
Accepted: September 9, 2009
Published online: October 31, 2009

Abstract

The existence of hepatitis C virus (HCV) infection in extrahepatic sites has been widely demonstrated. Since peripheral blood mononuclear cells have been the most investigated, compelling evidence of an association with HCV has been shown. Different studies have revealed that HCV RNA can persist and replicate in immune cells but the relevance of its presence and persistence over time is still unknown. As the contribution of this extrahepatic reservoir could have several clinical implications in viral transmission, treatment response and disease pathogenesis, future studies are required to improve our knowledge of the extrahepatic manifestations of HCV and its possible consequences.

Key Words: Hepatitis C virus, Peripheral blood mononuclear cell, Reservoirs, Lymphotropism, Replication



INTRODUCTION

Hepatitis C virus (HCV) is a small positive-strand RNA virus responsible for an important burden of chronic hepatitis and hepatic related diseases around the world[1]. Although HCV is mainly hepatotropic, its presence in extrahepatic sites has been widely demonstrated[2] and it was calculated that the contribution of this second compartment is responsible for about 3.1% of virus in circulation[3]. Lymphoid cells are the most investigated extrahepatic site.

HCV infection of lymphoid cells was suggested for the first time by Hellings in 1985[4]. Mononuclear leucocytes (mainly lymphocytes), isolated by Ficoll-Paque gradient centrifugation of blood freshly drawn from a hemophilia A patient with chronic non-A, non-B hepatitis (NANB), caused NANB when infused in a susceptible chimpanzee.

Immediately after discovery of the virus in 1989[5], different groups attempted to demonstrate HCV replication in lymphoid cells by infecting macrophages, B and T lymphocytes[6-9]. Moreover, several reports describing the presence of the replicative intermediate or negative strand in peripheral blood mononuclear cells (PBMC) were published[9,10]. The HCV RNA negative strand is a viral replicative intermediate and its presence can be considered direct evidence of ongoing viral replication. Nevertheless, discordant results were obtained by different groups, and the association of HCV with PBMC and viral replication in this extrahepatic site remained controversial for many years[11-16]. Strong evidence for in vivo HCV infection of, and replication in PBMC was provided by Bronowicki and collaborators[17]. They demonstrated the persistence of the viral RNA sequences in mononuclear blood cells inoculated into immunosuppressed mice and they were able to perform a second in vivo passage by successful transmission of HCV-RNA-positive cells to other mice. However, later on, the SCID mice did not offer a suitable in vivo model to study HCV pathogenesis. Different studies have reported evidence for HCV replication in granulocytes, monocytes/macrophages, dendritic cells, B and T lymphocytes[18-23]. In addition, successful infections of lymphoid cells or establishment of stable HCV+ cell lines have been achieved[22,24].

HCV REPLICATION IN PBMC

As discussed above, the detection of replicative forms of HCV RNA in PBMC has been extensively reported but remains controversial. Earlier PCR methods have been suspected to lack specificity and/or sensitivity, possibly due to the very low concentration of negative HCV RNA strand in cells. Currently, methodological modifications have been used to overcome these difficulties and many reports demonstrated that HCV can certainly replicate in PBMC[25-29].

Some studies showed that replication in PBMC occurs at a very low level and the amount of intracellular HCV RNA is patient-specific and is a result of a dynamic process related to virologic and immunologic factors[26,28]. The role of HCV lymphotropism in the natural history of HCV infection is not yet resolved and reports remain arguable. Nowadays, although it is accepted that HCV can replicate in PBMC, the contribution of this extrahepatic site as a significant viral reservoir and the importance of viral persistence in aviremic subjects after spontaneous or therapeutical clearance is still under debate.

PBMC AS HCV RESERVOIRS

Lymphoid cells may represent privileged reservoirs that could favor HCV persistence leading to chronic HCV infection. The infection of immune cells may interfere with the efficiency of viral clearance by the host[30,31]. Different reports demonstrate the persistence of HCV RNA at very low levels in serum and peripheral lymphoid cells after apparently complete spontaneous or antiviral therapy-induced resolution of chronic hepatitis C[32,33]. The occult HCV persistence in lymphoid cells may have important epidemiological and pathogenic implications. Radkowski and collaborators[33] suggested that in patients with sustained virological response (SVR), small quantities of HCV RNA may persist in liver or PBMC for up to 9 years. These findings could explain the phenomenon of frequent persistence of humoral and cellular immunity for many years after supposed viral clearance but also, could present a potential risk for transmission and reactivation. It was also demonstrated that HCV may also persist and replicate in the liver and PBMC of healthy, anti-HCV antibody-positive, serum HCV RNA-negative patients who have persistently normal ALT levels[34]. It is possible that viral persistence and, specifically, the presence of HCV RNA in PBMC may lead to HCV reactivation under special circumstances. In patients with immunosuppresion, under immunomodulatory therapy or with coinfection, persistent replicating HCV could represent a potential source for viral recurrence. These findings suggest that sterilizing immunity with complete elimination of virus is unlikely.

EVIDENCE AGAINST PBMC AS LONG-LIVED HCV RESERVOIRS

In contrast with the above mentioned reports, some studies refute the role of PBMC as a long-lived HCV reservoir. Kaiser P and collaborators[26] evaluated 30 HIV/HCV coinfected patients for up to 40 mo. Total PBMC-associated HCV RNA and virion-enclosed PBMC-associated HCV RNA, that could represent viral particles nonspecifically attached to blood cells, were distinguished and they observed widespread presence of viral RNA in PBMC from HCV-viremic patients. Evidence for persistence of HCV in PBMC in the absence of HCV viremia in plasma could not be found. Their experiments supported a concept of low level replication in PBMC and suggested that the infection and expression of HCV in PBMC is of minor quantitative importance for systemic replication and persistence of HCV[26].

Another report in which HCV persistence was underestimated was published by Bernardin[35]. In their experiments, they could not find any HCV RNA detectable PBMC sample in 69 aviremic donors indicating that PBMC are unlikely to serve as a long-lived reservoir of HCV in aviremic subjects.

The slow decrease in anti-HCV antibody titers in subjects with spontaneously cleared viremia as well as the complete seroreversion detected in 7% of transfusion transmitted infections may also reflect an absence of ongoing antigenic stimulation, indirectly supporting clearance of infection in persons who test HCV RNA- negative in plasma[35].

HCV LYMPHOTROPISM

The presence of variants of the highly conserved 5′ untranslated region (UTR) have been observed between HCV from plasma and PBMC[36,37]. The identification of sequence polymorphisms in cells of the lymphatic system suggested possible adaptation of HCV to replicate in nonhepatic cells[29]. In addition, a compartmental distribution of HCV quasispecies and HCV genotypes has been demonstrated[37,38]. Concordant with our results in a hemophilic population, the HCV genotypes detected in PBMC were not detected in plasma in some individuals supporting independent replication in these cells[37,39].

These findings further support the lymphotropic nature of HCV and reinforce the concept that independent replication of HCV in lymphoid cells may constitute a potential risk for persistence, reactivation, recurrence or treatment resistance.

CLINICAL IMPLICATIONS OF PBMC AS HCV RESERVOIRS
HCV transmission

In some contexts, as in vertical transmission, the presence of HCV in PBMC played relevant roles. The risk of mother-to-child transmission is associated with the presence of HCV in maternal PBMC[40]. Likewise, the persistence of small quantities of HCV RNA in aviremic patients who are supposed to have solved the infection could have important implications for viral transmission.

Reactivation or recurrence

Several studies demonstrate that relapse after sustained virological response is extremely rare[41]. However, low-level intrahepatic viraemia despite negative serum HCV RNA testing has been shown to predict a higher likelihood of late relapse, particularly in the setting of immunosuppression[41].

The existence of PBMC reservoirs may be implicated in the recurrence of chronic hepatitis after apparently successful antiviral treatment. Previous findings suggest that in patients with spontaneous eradication or sustained virological response after therapy, small quantities of HCV RNA may persist in lymphoid cells for years[32,33]. The presence of positive/negative strand HCV RNA at the end of treatment was associated with relapse among HCV-HIV coinfected patients[42]. It has been suggested that low level replication of HCV in PBMCs may lead to reactivation of HCV after termination of therapy[2,32,33].

Reemergence of HCV RNA was demonstrated in apparent sustained viral responders receiving immune suppressive therapy[43,44]. This proved that the HCV reservoir requires continued innate or T cell immune surveillance to prevent disease activity even years after the infection in at least some sustained viral responders[43].

On the other hand, recurrent infection in transplant recipients was also described[45-47] and the utilization of antiviral therapy in HCV-infected patients awaiting liver transplantation as one of the strategies to prevent hepatitis C recurrence after transplantation was recommended[48]. It has been proposed that viral variants from extrahepatic compartments may be involved in infection recurrence after liver transplantation. One report has specifically investigated the origin of HCV recurrence and suggested that liver-derived virus remaining in circulation was the major responsible for the graft reinfection. However, virus variants of likely extrahepatic origin could be detected in serum early after transplantation[45].

Immune dysfunction and lymphoproliferative disorders

The interaction between HCV and the human immune system is likely to have important clinical consequences. First, HCV has a remarkable ability to evade the immune system, achieving almost 85% chronicity rates. On the other hand, HCV infection may induce extra-hepatic immune related manifestations in a high percentage of infected patients, including mixed cryoglobulinemia and non-Hodgkin lymphoma. At present, the possible mechanisms by which HCV modulates immune function are being examined.

Monocytes, B cells, and CD4+ and CD8+ lymphocytes can support HCV replication and can serve as reservoirs in symptomatic and occult HCV infections[29]. Endogenous presentation of HCV antigens by infected B cells and monocytes may contribute to immune tolerance of HCV, favoring its persistence[29]. Particularly, in perihepatic lymph nodes, HCV replication has been demonstrated and the results suggest that replication of HCV in T cells might contribute to disturbance of Th1 commitment or Th1 hyporesponsiveness in individuals with persistent HCV infection[31].

According to some reports, a predominant infection of B lymphocytes suggests a preferential tropism of HCV for B cells[49,24]. In consequence, chronic antigen stimulation by the virus may trigger B cell proliferation resulting in a wide spectrum of lymphoproliferative disorders, cryoglobulinemia and non-Hodgkin lymphoma, frequently observed in infected patients[2,50].

Treatment resistance

Different factors have been associated with lower treatment response, such as higher serum HCV viral load at baseline, HCV genotype 1 infection, co-treatment with antiretroviral therapy, the presence of IFN-neutralizing antibodies and a higher degree of immune deterioration[51-56]. The detection of HCV RNA in PBMC reservoirs might have important implications for effective treatment. One possible mechanism of relapse is that PBMC could serve as a viral reservoir resistant to IFN[30]. As demonstrated, clearance of HCV RNA in PBMC at the end of IFN treatment was a predictor of durable response to antiviral therapy in patients with chronic hepatitis[57]. Moreover, in the context of HCV-HIV coinfected patients, the presence of strand specific HCV RNA at the end of 48 wk of therapy was associated with viral relapse[41].

On the other hand, HCV lymphotropic variants corresponding to genotypes more resistant to treatment could persist in lymphoid cells[58,39]. Interestingly, Di Liberto and collaborators found that the presence of compartmentalization in PBMC was strongly predictive of sustained virologic response[59].

CONCLUSION

To date, different reports suggest that HCV replication in PBMC does definitely occur which proved to have important effects on different aspects of HCV infection. Nonetheless, as conflicting findings in vivo still exist, the contribution of this extrahepatic site in disease pathogenesis and treatment should be further explored and the mechanisms involved should be elucidated.

Footnotes

Peer reviewer: Ajay Duseja, MD, DM, FACG, Consultant, Department of Hepatology, Post Graduate Institute of Medical Education & Research,Chandigarh 160012, India

S- Editor Zhang HN L- Editor Lalor PF E- Editor Ma WH

References
1.  Alter MJ. Epidemiology of hepatitis C virus infection. World J Gastroenterol. 2007;13:2436-2441.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Blackard JT, Kemmer N, Sherman KE. Extrahepatic replication of HCV: insights into clinical manifestations and biological consequences. Hepatology. 2006;44:15-22.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Dahari H, Feliu A, Garcia-Retortillo M, Forns X, Neumann AU. Second hepatitis C replication compartment indicated by viral dynamics during liver transplantation. J Hepatol. 2005;42:491-498.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Hellings JA, van der Veen-du Prie J, Snelting-van Densen R, Stute R. Preliminary results of transmission experiments of non-A, non-B hepatitis by mononuclear leucocytes from a chronic patient. J Virol Methods. 1985;10:321-326.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science. 1989;244:359-362.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Shimizu YK, Iwamoto A, Hijikata M, Purcell RH, Yoshikura H. Evidence for in vitro replication of hepatitis C virus genome in a human T-cell line. Proc Natl Acad Sci USA. 1992;89:5477-5481.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Cribier B, Schmitt C, Bingen A, Kirn A, Keller F. In vitro infection of peripheral blood mononuclear cells by hepatitis C virus. J Gen Virol. 1995;76:2485-2491.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Kato N, Nakazawa T, Mizutani T, Shimotohno K. Susceptibility of human T-lymphotropic virus type I infected cell line MT-2 to hepatitis C virus infection. Biochem Biophys Res Commun. 1995;206:863-869.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Caussin-Schwemling C, Schmitt C, Stoll-Keller F. Study of the infection of human blood derived monocyte/macrophages with hepatitis C virus in vitro. J Med Virol. 2001;65:14-22.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Zignego AL, Macchia D, Monti M, Thiers V, Mazzetti M, Foschi M, Maggi E, Romagnani S, Gentilini P, Brechot C. Infection of peripheral mononuclear blood cells by hepatitis C virus. J Hepatol. 1992;15:382-386.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Muller HM, Pfaff E, Goeser T, Kallinowski B, Solbach C, Theilmann L. Peripheral blood leukocytes serve as a possible extrahepatic site for hepatitis C virus replication. J Gen Virol. 1993;74:669-676.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Muratori L, Giostra F, Cataleta M, Francesconi R, Ballardini G, Cassani F, Lenzi M, Bianchi FB. Testing for hepatitis C virus sequences in peripheral blood mononuclear cells of patients with chronic hepatitis C in the absence of serum hepatitis C virus RNA. Liver. 1994;14:124-128.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Lanford RE, Chavez D, Chisari FV, Sureau C. Lack of detection of negative-strand hepatitis C virus RNA in peripheral blood mononuclear cells and other extrahepatic tissues by the highly strand-specific rTth reverse transcriptase PCR. J Virol. 1995;69:8079-8083.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Laskus T, Radkowski M, Wang LF, Cianciara J, Vargas H, Rakela J. Hepatitis C virus negative strand RNA is not detected in peripheral blood mononuclear cells and viral sequences are identical to those in serum: a case against extrahepatic replication. J Gen Virol. 1997;78:2747-2750.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Mellor J, Haydon G, Blair C, Livingstone W, Simmonds P. Low level or absent in vivo replication of hepatitis C virus and hepatitis G virus/GB virus C in peripheral blood mononuclear cells. J Gen Virol. 1998;79:705-714.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Sansonno D, Lotesoriere C, Cornacchiulo V, Fanelli M, Gatti P, Iodice G, Racanelli V, Dammacco F. Hepatitis C virus infection involves CD34(+) hematopoietic progenitor cells in hepatitis C virus chronic carriers. Blood. 1998;92:3328-3337.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Bronowicki JP, Loriot MA, Thiers V, Grignon Y, Zignego AL, Brechot C. Hepatitis C virus persistence in human hematopoietic cells injected into SCID mice. Hepatology. 1998;28:211-218.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Lerat H, Rumin S, Habersetzer F, Berby F, Trabaud MA, Trepo C, Inchauspe G. In vivo tropism of hepatitis C virus genomic sequences in hematopoietic cells: influence of viral load, viral genotype, and cell phenotype. Blood. 1998;91:3841-3849.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Laskus T, Radkowski M, Piasek A, Nowicki M, Horban A, Cianciara J, Rakela J. Hepatitis C virus in lymphoid cells of patients coinfected with human immunodeficiency virus type 1: evidence of active replication in monocytes/macrophages and lymphocytes. J Infect Dis. 2000;181:442-448.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Goutagny N, Fatmi A, De Ledinghen V, Penin F, Couzigou P, Inchauspe G, Bain C. Evidence of viral replication in circulating dendritic cells during hepatitis C virus infection. J Infect Dis. 2003;187:1951-1958.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Radkowski M, Bednarska A, Horban A, Stanczak J, Wilkinson J, Adair DM, Nowicki M, Rakela J, Laskus T. Infection of primary human macrophages with hepatitis C virus in vitro: induction of tumour necrosis factor-alpha and interleukin 8. J Gen Virol. 2004;85:47-59.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Bare P, Massud I, Parodi C, Belmonte L, Garcia G, Nebel MC, Corti M, Pinto MT, Bianco RP, Bracco MM. Continuous release of hepatitis C virus (HCV) by peripheral blood mononuclear cells and B-lymphoblastoid cell-line cultures derived from HCV-infected patients. J Gen Virol. 2005;86:1717-1727.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  MacParland SA, Pham TN, Gujar SA, Michalak TI. De novo infection and propagation of wild-type Hepatitis C virus in human T lymphocytes in vitro. J Gen Virol. 2006;87:3577-3586.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Sung VM, Shimodaira S, Doughty AL, Picchio GR, Can H, Yen TS, Lindsay KL, Levine AM, Lai MM. Establishment of B-cell lymphoma cell lines persistently infected with hepatitis C virus in vivo and in vitro: the apoptotic effects of virus infection. J Virol. 2003;77:2134-2146.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Blackard JT, Smeaton L, Hiasa Y, Horiike N, Onji M, Jamieson DJ, Rodriguez I, Mayer KH, Chung RT. Detection of hepatitis C virus (HCV) in serum and peripheral-blood mononuclear cells from HCV-monoinfected and HIV/HCV-coinfected persons. J Infect Dis. 2005;192:258-265.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Kaiser P, Niederost B, Joos B, von Wyl V, Opravil M, Weber R, Gunthard HF, Fischer M. Equal amounts of intracellular and virion-enclosed hepatitis C virus RNA are associated with peripheral-blood mononuclear cells in vivo. J Infect Dis. 2006;194:1713-1723.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Pal S, Sullivan DG, Kim S, Lai KK, Kae J, Cotler SJ, Carithers RL Jr, Wood BL, Perkins JD, Gretch DR. Productive replication of hepatitis C virus in perihepatic lymph nodes in vivo: implications of HCV lymphotropism. Gastroenterology. 2006;130:1107-1116.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Laskus T, Operskalski EA, Radkowski M, Wilkinson J, Mack WJ, deGiacomo M, Al-Harthi L, Chen Z, Xu J, Kovacs A. Negative-strand hepatitis C virus (HCV) RNA in peripheral blood mononuclear cells from anti-HCV-positive/HIV-infected women. J Infect Dis. 2007;195:124-133.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Pham TN, King D, Macparland SA, McGrath JS, Reddy SB, Bursey FR, Michalak TI. Hepatitis C virus replicates in the same immune cell subsets in chronic hepatitis C and occult infection. Gastroenterology. 2008;134:812-822.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Zignego AL, Giannini C, Monti M, Gragnani L. Hepatitis C virus lymphotropism: lessons from a decade of studies. Dig Liver Dis. 2007;39 Suppl 1:S38-S45.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Kondo Y, Sung VM, Machida K, Liu M, Lai MM. Hepatitis C virus infects T cells and affects interferon-gamma signaling in T cell lines. Virology. 2007;361:161-173.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Pham TN, MacParland SA, Mulrooney PM, Cooksley H, Naoumov NV, Michalak TI. Hepatitis C virus persistence after spontaneous or treatment-induced resolution of hepatitis C. J Virol. 2004;78:5867-5874.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Radkowski M, Gallegos-Orozco JF, Jablonska J, Colby TV, Walewska-Zielecka B, Kubicka J, Wilkinson J, Adair D, Rakela J, Laskus T. Persistence of hepatitis C virus in patients successfully treated for chronic hepatitis C. Hepatology. 2005;41:106-114.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Carreno V, Pardo M, Lopez-Alcorocho JM, Rodriguez-Inigo E, Bartolome J, Castillo I. Detection of hepatitis C virus (HCV) RNA in the liver of healthy, anti-HCV antibody-positive, serum HCV RNA-negative patients with normal alanine aminotransferase levels. J Infect Dis. 2006;194:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Bernardin F, Tobler L, Walsh I, Williams JD, Busch M, Delwart E. Clearance of hepatitis C virus RNA from the peripheral blood mononuclear cells of blood donors who spontaneously or therapeutically control their plasma viremia. Hepatology. 2008;47:1446-1452.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Laskus T, Radkowski M, Wang LF, Nowicki M, Rakela J. Uneven distribution of hepatitis C virus quasispecies in tissues from subjects with end-stage liver disease: confounding effect of viral adsorption and mounting evidence for the presence of low-level extrahepatic replication. J Virol. 2000;74:1014-1017.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Roque-Afonso AM, Ducoulombier D, Di Liberto G, Kara R, Gigou M, Dussaix E, Samuel D, Feray C. Compartmentalization of hepatitis C virus genotypes between plasma and peripheral blood mononuclear cells. J Virol. 2005;79:6349-6357.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Roque Afonso AM, Jiang J, Penin F, Tareau C, Samuel D, Petit MA, Bismuth H, Dussaix E, Feray C. Nonrandom distribution of hepatitis C virus quasispecies in plasma and peripheral blood mononuclear cell subsets. J Virol. 1999;73:9213-9221.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Parodi C, Culasso A, Aloisi N, Garcia G, Baston M, Corti M, Bianco RP, Campos R, Ares BR, Bare P. Evidence of occult HCV genotypes in haemophilic individuals with unapparent HCV mixed infections. Haemophilia. 2008;14:816-822.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Azzari C, Resti M, Moriondo M, Ferrari R, Lionetti P, Vierucci A. Vertical transmission of HCV is related to maternal peripheral blood mononuclear cell infection. Blood. 2000;96:2045-2048.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Desmond CP, Roberts SK, Dudley F, Mitchell J, Day C, Nguyen S, Pianko S. Sustained virological response rates and durability of the response to interferon-based therapies in hepatitis C patients treated in the clinical setting. J Viral Hepat. 2006;13:311-315.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  de Felipe B, Leal M, Soriano-Sarabia N, Gutierrez A, Lopez-Cortes L, Molina-Pinelo S, Vallejo A. HCV RNA in peripheral blood cell subsets in HCV-HIV coinfected patients at the end of PegIFN/RBV treatment is associated with virologic relapse. J Viral Hepat. 2009;16:21-27.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Lee WM, Polson JE, Carney DS, Sahin B, Gale M Jr. Reemergence of hepatitis C virus after 8.5 years in a patient with hypogammaglobulinemia: evidence for an occult viral reservoir. J Infect Dis. 2005;192:1088-1092.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Lin A, Thadareddy A, Goldstein MJ, Lake-Bakaar G. Immune suppression leading to hepatitis C virus re-emergence after sustained virological response. J Med Virol. 2008;80:1720-1722.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Laskus T, Radkowski M, Wilkinson J, Vargas H, Rakela J. The origin of hepatitis C virus reinfecting transplanted livers: serum-derived versus peripheral blood mononuclear cell-derived virus. J Infect Dis. 2002;185:417-421.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Zekri AR, Mohamed WS, Samra MA, Sherif GM, El-Shehaby AM, El-Sayed MH. Risk factors for cytomegalovirus, hepatitis B and C virus reactivation after bone marrow transplantation. Transpl Immunol. 2004;13:305-311.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Melon S, Galarraga MC, Villar M, Laures A, Boga JA, de Ona M, Gomez E. Hepatitis C virus reactivation in anti-hepatitic C virus-positive renal transplant recipients. Transplant Proc. 2005;37:2083-2085.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Forns X, Garcia-Retortillo M, Serrano T, Feliu A, Suarez F, de la Mata M, Garcia-Valdecasas JC, Navasa M, Rimola A, Rodes J. Antiviral therapy of patients with decompensated cirrhosis to prevent recurrence of hepatitis C after liver transplantation. J Hepatol. 2003;39:389-396.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Ducoulombier D, Roque-Afonso AM, Di Liberto G, Penin F, Kara R, Richard Y, Dussaix E, Feray C. Frequent compartmentalization of hepatitis C virus variants in circulating B cells and monocytes. Hepatology. 2004;39:817-825.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Landau DA, Saadoun D, Calabrese LH, Cacoub P. The pathophysiology of HCV induced B-cell clonal disorders. Autoimmun Rev. 2007;6:581-587.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Strader DB, Wright T, Thomas DL, Seeff LB. Diagnosis, management, and treatment of hepatitis C. Hepatology. 2004;39:1147-1171.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Zeuzem S, Hultcrantz R, Bourliere M, Goeser T, Marcellin P, Sanchez-Tapias J, Sarrazin C, Harvey J, Brass C, Albrecht J. Peginterferon alfa-2b plus ribavirin for treatment of chronic hepatitis C in previously untreated patients infected with HCV genotypes 2 or 3. J Hepatol. 2004;40:993-999.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Jorns C, Holzinger D, Thimme R, Spangenberg HC, Weidmann M, Rasenack J, Blum HE, Haller O, Kochs G. Rapid and simple detection of IFN-neutralizing antibodies in chronic hepatitis C non-responsive to IFN-alpha. J Med Virol. 2006;78:74-82.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Nunez M, Marino A, Miralles C, Berdun MA, Sola J, Hernandez-Burruezo JJ, Galindo MJ, Barreiro P, Martin-Carbonero L, Soriano V. Baseline serum hepatitis C virus (HCV) RNA level and response at week 4 are the best predictors of relapse after treatment with pegylated interferon plus ribavirin in HIV/HCV-coinfected patients. J Acquir Immune Defic Syndr. 2007;45:439-444.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Farnik H, Mihm U, Zeuzem S. Optimal therapy in genotype 1 patients. Liver Int. 2009;29 Suppl 1:23-30.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Sulkowski MS, Thomas DL, Chaisson RE, Moore RD. Hepatotoxicity associated with antiretroviral therapy in adults infected with human immunodeficiency virus and the role of hepatitis C or B virus infection. JAMA. 2000;283:74-80.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Xu DZ, Xie Y, Li ZQ. Clearance of HCV RNA in peripheral blood mononuclear cell as a predictor of response to antiviral therapy in patients with chronic hepatitis C. Hepatobiliary Pancreat Dis Int. 2005;4:550-553.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Schroter M, Feucht HH, Zollner B, Schafer P, Laufs R. Multiple infections with different HCV genotypes: prevalence and clinical impact. J Clin Virol. 2003;27:200-204.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Di Liberto G, Roque-Afonso AM, Kara R, Ducoulombier D, Fallot G, Samuel D, Feray C. Clinical and therapeutic implications of hepatitis C virus compartmentalization. Gastroenterology. 2006;131:76-84.  [PubMed]  [DOI]  [Cited in This Article: ]