Editorial Open Access
Copyright ©2009 The WJG Press and Baishideng. All rights reserved.
World J Gastroenterol. Nov 7, 2009; 15(41): 5129-5140
Published online Nov 7, 2009. doi: 10.3748/wjg.15.5129
Costimulatory molecule programmed death-1 in the cytotoxic response during chronic hepatitis C
Juan Ramón Larrubia, Selma Benito-Martínez, Joaquín Miquel, Miryam Calvino, Eduardo Sanz-de-Villalobos, Trinidad Parra-Cid, Translational Hepatology Unit, Guadalajara University Hospital, University of Alcalá, 19002 Guadalajara, Spain
Author contributions: Larrubia JR and Benito-Martínez S contributed equally towards the conception and design of the review; Larrubia JR and Benito-Martínez S co-wrote the manuscript; Larrubia JR revised the manuscript; Miquel J, Parra-Cid T, Calvino M and Sanz-de-Villalobos E contributed equally to the supportive work and supervision.
Supported by Grants from “Fiscam” J.C.C.M (Ayuda para proyectos de investigación en salud; PI-2007/32) and “Fundación de Investigación Médica Mutua Madrileña” (Beca Ayudas a la Investigación FMMM; 2548/2008) from Spain. Benito-Martínez S was supported by a research grant from “Fiscam” J.C.C.M (“Perfeccionamiento y movilidad de investigadores”; MOV-2007_JI/18), Spain. Calvino M was supported by a research grant from “Instituto de Salud Carlos III” (Contrato de apoyo a la investigación en el SNS’’; CA07/00157), Spain
Correspondence to: Juan Ramón Larrubia, MD, MSc, PhD, Translational Hepatology Unit, Guadalajara University Hospital, University of Alcalá, Donante de Sangre st, 19002 Guadalajara, Spain. jlarrubiam@meditex.es
Telephone: +34-949-209200 Fax: +34-949-209259
Received: August 20, 2009
Revised: September 28, 2009
Accepted: October 9, 2009
Published online: November 7, 2009

Abstract

Hepatitis C virus (HCV)-specific CD8+ T cells play an important role in the resolution of HCV infection. Nevertheless, during chronic hepatitis C these cells lack their effector functions and fail to control the virus. HCV has developed several mechanisms to escape immune control. One of these strategies is the up-regulation of negative co-stimulatory molecules such us programmed death-1 (PD-1). This molecule is up-regulated on intrahepatic and peripheral HCV-specific cytotoxic T cells during acute and chronic phases of the disease, whereas PD-1 expression is low in resolved infection. PD-1 expressing HCV-specific CD8+ T cells are exhausted with impairment of several effector mechanisms, such as: type-1 cytokine production, expansion ability after antigen encounter and cytotoxic ability. However, PD-1 associated exhaustion can be restored by blocking the interaction between PD-1 and its ligand (PD-L1). After this blockade, HCV-specific CD8+ T cells reacquire their functionality. Nevertheless, functional restoration depends on PD-1 expression level. High PD-1-expressing intrahepatic HCV-specific CD8+ T cells do not restore their effector abilities after PD-1/PD-L1 blockade. The mechanisms by which HCV is able to induce PD-1 up-regulation to escape immune control are unknown. Persistent TCR stimulation by a high level of HCV antigens could favour early PD-1 induction, but the interaction between HCV core protein and gC1q receptor could also participate in this process. The PD-1/PD-L1 pathway modulation could be a therapeutic strategy, in conjunction with the regulation of others co-stimulatory pathways, in order to restore immune response against HCV to succeed in clearing the infection.

Key Words: Chronic hepatitis, Exhaustion, Hepatitis C virus core, Hepatitis C virus, Programmed death-1, Programmed death-1 ligand



INTRODUCTION
Table 1 Summary of CD28 family co-stimulatory and co-inhibitory pathways.
ReceptorExpressionLigandLigand expressionT cell response regulation
CD28T cells (naive and some memory)CD80B, T and DC, macrophagesPositive
PD-1Activate T cells, B cells, macrophagesPD-L1 PD-L2B, T and DC, macrophages, non-lymphoid cells (pancreas, placenta, heart)Negative
CTLA-4Activate T cells and regulatory T cellsCD80 CD86B, T and DC, macrophagesNegative
BTLAB and T cellsPD-L2Macrophages, DCNegative
B7-H3T and B cells, NK
B7-H4
ICOST cells (memory and effector)ICOS-LB, T cells, macrophages and DCPositive
Figure 1
Figure 1 Programmed death-1 (PD-1) structure and interactions. Interaction between the PD-1 molecule expressed on T cells and its ligand PD-L1 expressed on antigen-presenting cells leads to immunoreceptor tyrosine-based switch motif (ITSM) motif phosphorylation in its cytoplasmic tyrosines which are recognized by src homology 2 domain-containing tyrosine phosphatase 2 (SHP-2). All of these interactions cause T cell anergy due to T cell receptor (TCR)-dependent MAP Kinase-pathway signalling inhibition which avoids interleukin (IL)-2 gene transduction. PD-1 expression is induced by TCR activation but could also be favoured by HCV-core protein through interaction with gC1qR. PD-L1 is up-regulated on antigen presenting cells by the effect of γ-interferon produced during HCV infection by activated lymphocytes.
Figure 2
Figure 2 Liver microenvironment. Circulating HCV-specific CD8+ T cells migrating through the hepatic sinusoid interact with resident liver cells [Kupffer cells, dendritic cells (DC), hepatocytes, liver sinusoidal endothelial cells (LSECs), stellate cells] that could act as antigen presenting cells. These cells up-regulate PD-L1 expression during persistent HCV infection and interact with the PD-1 molecule expressed on HCV-specific CD8+ T cells. This interaction leads to T cell anergy. PD-1 up-regulation is produced by TCR stimulation in addition to the interaction between HCV-core protein and the complement receptor (gC1qR). In this micro-environment the regulatory T cells (Treg) also participate, whose activity is also regulated by the PD-1/PD-L1 pathway.
Figure 3
Figure 3 PD-L1 expression on Kupffer cells and PD-1 expression on HCV-specific CD8+ T cells. A: PD-L1-FITC FACS® histograms gated on CD11b+ CD54+ cells from liver (L) and peripheral blood (PB), showing a higher PD-L1 expression on intrahepatic Kuppfer cells; B: FACS® dot-plots and histograms of peripheral blood and intrahepatic T cells stained with CD8-Cy mAb, HCV-tetramers-PE and PD-1-FITC mAb. Gated Tetramer+/CD8+ cells are presented in the histograms for PD-1-FITC expression. PD-1 is up-regulated on intrahepatic HCV-specific CD8+ cells.
Figure 4
Figure 4 Direct ex-vivo PD-1, CD127 expression and IFN-γ production. FACS® dot-plots and histograms of peripheral blood T cells from two representative patients, one with persistent HCV infection and the other with resolved HCV infection. T cells are stained with PD-1-FITC, CD127-FITC, γ-IFN-FITC and CD8-Cy mAbs and HCV-tetramers. In persistent infection, HCV-specific CD8+ cells maintain a PD-1+/CD127-/IFN-γ- phenotype while the resolved infection case displays an opposite phenotype PD-1-/CD127+/IFN-γ+.
Figure 5
Figure 5 Proliferation restoration after PD-1 blockade. FACS® dot-plots of peripheral blood T cells from two representative patients, one with persistent HCV infection and the other with resolved HCV infection, and a control case, after specific stimulation in the presence or absence of anti-PD-L1 mAb. T cells were stimulated for 10 d with the HCV–specific peptide plus IL-2. After stimulation, T cells were stained with CD8-Cy mAbs and HCV-tetramers-PE. PD-1/PD-L1 pathway blockade by anti-PD-L1 antibodies increases the HCV-specific cell proliferation in the chronic patient that had a high level of PD-1 expression.
Figure 6
Figure 6 PD-1 up-regulation induced by HCV-core protein. A: FACS® dot-plots and histograms of peripheral blood CD8+ cells stained with PD-1-FITC and CD8-Cy mAbs from a healthy subject. CD8+ cells were stimulated with B-galactosidase, HCV-core protein, CD3-CD28 mAb, and HCV-core protein plus CD3-CD28 mAbs. PD-1 expression was highly up-regulated on CD8+ cells after non-specific stimulation by anti CD3/CD28 mAbs in the presence of HCV core protein; B: FACS® dot-plots and histograms of peripheral blood CD8+ cells stained with Ki-67 FTIC and CD8-Cy mAbs from the same healthy subject to test proliferation ability of CD8+ cells after incubation with B-galactosidase, CD3-CD28 mAb, HCV-core protein plus CD3-CD28 mAb and HCV-core protein plus CD3-CD28 and anti-PD-L1 mAbs. HCV-core protein decreased the proliferation induced by CD3-CD28 mAb stimulation. This proliferation impairment induced by HCV-core protein was resolved by anti-PD-L1 mAb treatment.

Hepatitis C virus (HCV) is a hepatotropic non-cytopathic positive-strand RNA virus which belongs to the Flaviviridae family. HCV infection is a major public problem, affecting more than 200 million people worldwide[1]. Only around a quarter of acute HCV infections resolve within a few months, while in the majority of cases the virus establishes a persistent infection, and a significant proportion of cases progress to fibrosis, cirrhosis, liver failure or even hepatocellular carcinoma[2-4]. Nowadays standard anti-HCV therapy resolves about 50% of chronic infections[5-7], therefore new therapeutic strategies should be designed to control this disease. HCV-specific cytotoxic T lymphocytes (CTLs) play a major role in viral control during acute infection[8]. Nevertheless, during persistent infection HCV-specific CTL effector functions are significantly impaired, and this situation is a major cause of host inability to eliminate the persistent virus[9,10]. Appropriate activation of primed virus-specific CTLs in the infected site depends on the engagement between T cell receptor (TCR) and HLA-I/epitope complex plus interaction among positive co-stimulatory molecules and their ligands[11,12]. Virus-specific CTLs, after developing their effector function, express negative co-stimulatory molecules to switch-off their activity. The appropriate virus-specific CTL response development correlates with the adequate balance between positive and negative co-stimulatory signals (Table 1)[13,14]. Programmed death-1 (PD-1) is one of the negative co-stimulatory molecules. Engagement of PD-1 and its ligand (PD-L1) delivers a negative signal to the TCR activation pathway, avoiding proliferation, and interleukin (IL)-2 production, which leads to T cell anergy[15,16]. Evidence that PD-1 suppresses activation of the immune response comes from studies in which mice deficient in PD-1 developed autoimmune diseases, such as systemic lupus erythematosus, dilated cardiomyopathy, rheumatoid arthritis and type I diabetes mellitus, due to the uncontrolled persistent T cell activation against different epitopes[17,18]. The PD-1 induced exhaustion on virus-specific T cells was first described by Barber et al[19], in a murine model of lymphocytic choriomeningitis virus (LCMV) infection. The authors demonstrated that the majority of LCMV-specific CD8+ T cells were anergic during the chronic phase of infection in association with PD-1 up-regulation. Mice treated with anti-PD-L1 monoclonal antibodies restored the LCMV-specific cyototoxic response and facilitated viral control. These experimental data suggested that the PD-1/PD-L1 pathway could play a major role in the development of persistent infections by non-cytopathic viruses, and different groups started to research the role of this pathway in different chronic viral infections in humans, such as hepatitis B virus (HBV), HCV and human immunodeficiency virus (HIV) infections. Bearing in mind these data, the PD-1/PD-L1 pathway could be an effective escape mechanism and its blockade could be a therapeutic target to reverse T-cell dysfunction. In this editorial, the current state of knowledge about the role of PD-1 expression on specific cytotoxic responses during HCV infection is reviewed.

STRUCTURE AND EXPRESSION OF PD-1 AND PD-L1

PD-1 is a 55 kDa glycoprotein which belongs to the CD28 immunoglobulin superfamily of transmembrane proteins[20]. PD-1 shares a 23% homology with CTLA-4, which is another member of this family, although PD-1 has lost the MYPPPY motif for binding to B7 molecules[20], and the cysteine residue necessary for homodimerization[21]. PD-1 is expressed on activated T cells, B cells and myeloid cells (Table 1)[20]. The PD-1 structure consists of two regions; the extracellular region is formed by a single IgV-like domain and its cytoplasmic region contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) and an immunoreceptor tyrosine-based switch motif (ITSM)[22,23]. Following antigen stimulation, PD-1 recruits the protein tyrosine phosphatase src homology 2 domain-containing tyrosine phosphatase 2 (SHP-2) to ITSM but not to the ITIM motif, and subsequently, SHP-2 dephosphorylates effector molecules downstream of the TCR-induced nuclear regulatory pathway[24,25]. The direct result of PD-1 mediated inhibition of T cell proliferation is cell cycle arrest in G0/G1 and the inhibition of IL-2 production[15,16] (Figure 1). The ligands for PD-1 (PD-L1 and PD-L2) are type I transmembrane proteins with IgV and IgC-like domains in the extracellular region[26,27]. PD-L1 is expressed on resting and activated B and T cells, and on non-lymphoid cells such as pancreas, placenta and heart, while PD-L2 is induced on dendritic cells (DC) and macrophages[26-30] (Table 1). Interestingly, PD-L1 can be up-regulated on hepatocytes by α-interferon (IFN) and γ-IFN, and also by activated lymphocytes, and by direct viral infection (perhaps also through IFN pathways)[31-33]. PD-1 plays an important physiological role in regulating the cellular immune response, tuning-down the cellular effector functions after T cells have developed their tasks. This physiological function of PD-1 can be damaged by persistent viruses inducing a tolerogenic-like status on specific T cells to avoid immune viral control.

PD-1 EXPRESSION IN THE LIVER

The liver is characterised by being an immunotolerant organ prepared to deal with intense contact with antigens from the gut, and PD-1/PD-L1 is expressed in resident and infiltrating liver cells to carry out this task[34]. The liver is also the primary site for HCV replication and disease pathogenesis[35], and HCV can take advantage of the PD-1/PD-L1 pathway to impair the HCV-specific response reaching the infected liver in order to escape immune control. The liver is exposed to antigens and microbiologically-derived molecules which cause a unique microenvironment that requires liver immunological properties to induce tolerance rather than immunity[36-38]. Hepatic tolerance contributes to the common ineffectiveness of immune response against HCV which often results in chronic viral persistence[39]. When naive T cells reach the liver from the bloodstream they are activated by resident antigen presenting cells and are prone to become anergic, and this process could take part in the interaction between PD-1 and PD-L1 (Figure 2)[34,40]. On the other hand, primed effector HCV-specific T cells, reaching the infected liver, are also conducted through anergy by several mechanisms. One of them is PD-1 up-regulation on T cells in the liver[41,42], and the expression of its ligand on resident liver cells, such as hepatocytes, Kuppfer cells and sinusoidal endothelial cells (Figure 2)[31]. Usually, PD-L1 is constitutively expressed in non-lymphoid tissues such as heart, lung, placenta, kidney, and liver[43-45], but during chronic HCV infection, this molecule is up-regulated on parenchymal liver cells, as previously commented (Figure 3A). The regulation of HCV-specific effector CTLs is also controlled by intrahepatic CD4+CD25+FoxP3+ cells (regulatory T cells, Treg). These cells have an important role in maintaining the balance between tolerance and immunity in HCV infection[46-48]. The PD-1/PD-L1 pathway is also important in modulating the regulatory activity of these Treg cells. The PD-1/PD-L1 interaction on intrahepatic Treg suppresses their regulatory activity, favouring CTL response[49-51]. Nevertheless, when it is necessary to down-modulate HCV-specific CTL response in order to avoid liver damage, PD-1/PD-L1 engagement is not produced between Tregs and intrahepatic PD-L1 expressing cells, due to PD-L1 down-regulation on resident liver cells, allowing Tregs to down-modulate HCV-specific CTLs effector functions (Figure 2)[52]. The important role of PD-1 in liver pathogenesis during HCV chronic infection is evident, as it is shown by the high PD-1 expression on total intrahepatic T cells[53-56], indicating that some non-specific HCV-dependent stimulus is acting in liver infiltrating T cells to favour PD-1 up-regulation. Previous reports suggest that this factor could be HCV-core protein and this will be discussed later. In addition to this non-specific stimulation on T cells, PD-1 expression is also induced by persistent specific TCR stimulation. PD-1 expression is higher on intrahepatic than in peripheral HCV-specific CD8+ cells[41] (Figure 3B). These data suggest that the intense TCR activation produced in the liver in conjunction with the high level of HCV-core protein leads to the highest PD-1 expression on HCV-specific CD8+ cells. This high PD-1 up-regulation on intrahepatic specific CD8+ cells is exquisitely HCV specific, so that PD-1 expression on other virus-specific CD8+ T cells is not up-regulated during chronic HCV infection[41]. Therefore, liver environment conditions produce a huge PD-1 up-regulation on HCV-specific CTLs during persistent infection, and this could impair viral control by the cellular immune response through anergy induction[41].

DIFFERENTIAL PD-1 EXPRESSION IN ACUTE, CHRONIC AND RESOLVED HEPATITIS C VIRUS INFECTION

During the initial phase of acute infection, HCV-specific CD8+ T cells are dysfunctional irrespective of the final outcome of the disease, and this impairment persists when infection becomes chronic[10]. In contrast, effector and memory CD8+ T cells generated after acute onset are highly functional in cases of resolving infection[57,58]. One of the possible mechanisms responsible for impairment of virus-specific CTL response could be the exhaustion of these cells caused by PD-1 up-regulation. The exhaustion of virus-specific CD8+ T cells has been observed in different human infections such as HIV, HBV and HCV infections[56,59-65]. In HCV infection, during the early period of primo-infection irrespective of the final outcome, PD-1 is up-regulated on all HCV-specific CD8+ T cells[53,66]. However, after the acute stage of the disease PD-1 expression is modulated depending on the progression. Therefore, during self-limited infection HCV-specific CD8+ cells down-regulate PD-1 expression, and acquire a CD127+ phenotype which correlates with appropriate effector functions (Figure 4)[67]. CD127 is the IL-7 receptor (IL-7R) which plays an essential role in mature lymphocyte survival through a pathway activated by the interaction with IL-7[67]. However, in persistent infection HCV-specific CD8+ cells remain CD127 negative, and maintain high levels of PD-1 expression[66,68] (Figure 4). Therefore, PD-1+ CD127- expressing HCV-specific CD8+ cells during persistent infection are not only anergic, but also prone to apoptosis after antigen encounter due to the absence of CD127 expression. Furthermore, PD-1 up-regulation on peripheral and intrahepatic HCV-specific CD8+ cells during the acute and chronic phases of infection is correlated with the apoptosis susceptibility of these cells[55]. As a result, the majority of high PD-1 expressing HCV-specific CD8+ cells could follow an apoptotic process[69], indicating that PD-1 is involved in anergy induction but could also be implicated in specific T cell deletion. Probably, both mechanisms are damaged by HCV infection to escape cellular immune response.

CORRELATION BETWEEN PD-1 EXPRESSION AND EFFECTOR FUNCTION IMPAIRMENT ON HCV-SPECIFIC CTLs

Once differential PD-1 expression on HCV-specific CD8+ T cells between chronic and resolved patients has been described, the next point to address is to analyse whether this difference translates into different quality of HCV-specific CTLs effector functions. Cytotoxic T-cell exhaustion represents a spectrum of effector defects that are correlated with the level of PD-1 expression. Recent reports show that patients with HCV chronic infection, whose CTLs display high PD-1 expression, have impaired CTL capacity to synthesise type-1 cytokines, such us γ-IFN, α-tumor necrosis factor (TNF) and IL-2, in addition to cytolytic molecules, such as perforin and granzyme B, after direct ex-vivo specific in-vitro challenge[41,54]. One of the variables determining viral control has been suggested to be the ability of virus-specific CD8+ cells to clonally expand after antigen encounter[45]. HCV-specific CD8+ T cells during persistent infection also displayed impaired proliferation ability after specific stimulation, which correlated with PD-1 expression level[70,71]. Because of the role of the PD-1/PD-L1 pathway in proliferation impairment, subsequent works were aimed at trying to enhance HCV-specific CD8+ T cell proliferation by modulating this pathway. Blocking the interaction between PD-1 and its ligand increased the proliferation ability of peripheral HCV-specific CD8+ cells from some chronic HCV patients, characterised by high PD-1 expression, but did not occur in others, suggesting the presence of another anti-proliferative mechanism not yet described[54,63]. During HCV-specific CTL exhaustion, not all effector functions are altered at the same time; proliferative potential and IL-2 production are lost at an early phase, whereas cytokine production and cytolytic function are lost later[72]. This progressive impairment could be related to the level of PD-1 up-regulation. Interestingly, the exhaustion of CTLs during chronic HCV infection is highly antigen-specific and related to the level of antigenemia, not being present in either CTLs against other specifities or HCV-specific CTLs from patients with resolved infection[41,66]. In these two situations PD-1 is not up-regulated on specific CTLs. As commented before, intrahepatic HCV-specific CD8+ T cells are highly PD-1 positive and they do not expand after antigen encounter and do not produce either γ-IFN or perforin, whereas intrahepatic specific CTLs against other viruses, such as influenza virus-specific CD8+ T cells, expand efficiently and present a high level of perforin expression, but interestingly they are PD-1 negative[41]. High PD-1 expressing intrahepatic HCV-specific CTLs do not respond to anti-PD-L1 treatment[41]. Therefore, when PD-1 expression is extremely up-regulated, treatment with anti-PD-L1 antibodies can not counteract the HCV-specific CTLs exhaustion, induced by the PD-1/PD-L1 pathway.

HCV-SPECIFIC CTL FUNCTIONAL RESTORATION AFTER PD-1/PD-L1 INTERACTION BLOCKADE

Previous studies developed an LCMV infection animal model, and specific CTL function restoration during persistent infection after treatment with anti-PD-L1 monoclonal antibodies was shown[19,73]. This finding could have clinical implications in the treatment of persistent viral infections, as will be discussed later. In HCV infection, the in-vitro blockade of the PD-1/PD-L1 pathway with anti-PDL-1 antibodies increases proliferation capacity after antigen encounter in peripheral PD-1 expressing HCV-specific CTLs from chronic patients (Figure 5). In-vitro treatment with anti-PD-L1 antibodies also restored γ-IFN, perforin, CD107a, IL-2 and IL-13 production after antigen specific stimulation[41,54,74]. However, this PD-1/PD-L1 pathway blockade is not efficient on intrahepatic HCV-specific CD8+ T cells, which are characterised by a higher PD-1 expression, as previously discussed. These cells failed to proliferate and produce perforin, γ-IFN and CD107a after specific stimulation in the presence of anti-PD-L1 antibodies[41]. All these findings suggest that PD-1 expression level correlates inversely with HCV-specific CD8+ T cells functional restoration by PD-1/PD-L1 blockade. PD-1/PD-L1 blockade may increase the functionality of peripheral HCV-specific CD8+ T cells with intermediate PD-1 expression, whereas this blockade did not enhance the effector functions of intrahepatic PD-1high expressing HCV-specific CD8+ T cells. High antigenic stimulation in the liver induces other negative co-stimulatory molecules, such as CTLA-4[41], which could maintain the anergic status, despite blockage of the PD-1/PD-L1 pathway. Therefore, the functional restoration of intrahepatic HCV-specific CTLs could be obtained by the combined blocking of different negative co-stimulatory molecules. In fact, a previous report has shown that combined PD-1 and CTLA-4 blockade induces a restoration of intrahepatic HCV-specific CD8+ T cell function in chronically HCV infected patients[75]. Obviously, the modulation of different co-stimulatory molecules on HCV-specific T cells, such as CD137[76], OX40[77] and ICOS[78] should be tested in combination with PD-1/PD-L1 blockade in order to restore HCV-specific CTL effector functions[79,80]. Nevertheless, blocking the engagement between PD-1 and PD-L1 is not enough in many cases to restore peripheral HCV-specific CTL functionality in chronic patients, even in combination with the blockade of other negative co-stimulatory molecules. It is reasonable to assume that these cells, exposed to high persistent antigenic stimulus, are prone to apoptosis. Previous data on HBV chronic infection showed an up-regulation of the pro-apoptotic molecule Bim on HBV-specific CTLs[81]. In this chronic infection, only CD127+ (IL-7R) cells maintained the ability to expand after antigen encounter. These CD127+ cells could be protected from apoptosis due to the antiapoptotic molecule Mcl-1, induced by IL-7. Otherwise, CD127- HBV-specific CTLs would die due to apoptosis after antigen encounter, mediated by the Bim pathway. Bearing in mind these data on HBV infection, it is possible that the benefit observed by blocking the PD-1/PD-L1 interaction may occur only in specific T cells protected against apoptosis by CD127 expression. This phenotype is quite rare in patients with long-standing HCV infection, and this could explain why not all PD-1 expressing HCV-specific CTLs respond to anti-PD-L1 treatment. This theoretical scenario should be tested in the near future.

HCV CORE PROTEIN INDUCES PD-1 UP-REGULATION

The PD-1 up-regulation on intrahepatic total T cells[53,56] suggests that something other than TCR stimulation is involved in the PD-1 expression regulation during HCV infection. HCV-core protein binding to the complement receptor gC1q (gC1qR) is responsible for impairing T cell proliferation ability[82] through down-regulation of the high affinity IL-2 receptor[83]. A recent report suggests that this process could be mediated by PD-1 expression induction[84]. In fact, in an intrahepatic HCV-core protein expressing mouse model, liver infiltration by PD-1 expressing cytotoxic T cells unable to clear the virus has been shown. However, the liver from HCV-core non-expressing mice was infiltrated by non-PD-1 expressing specific-CTLs which could control the viral infection[85]. These data suggest that HCV-core protein could play a role in early PD-1 induction on T cells, mainly in the liver environment where this protein is richly expressed[86,87]. At least in-vitro, PD-1 and PD-L1 expression are up-regulated on activated T cells in the presence of HCV-core protein[84]. PD-1 up-regulation induced by HCV-core protein translated into impairment of T cell proliferation ability[84]. However, this dysfunction could be partially restored by blocking the PD-1/PD-L1 pathway with anti-PD-L1 antibodies (Figures 1 and 6) and by blocking the interaction between HCV-core protein and gC1qR[84]. Probably the interaction between HCV-core protein and gC1qR co-operate with the continuous TCR stimulation to produce an early PD-1 up-regulation in order to induce a premature anergy on HCV-specific CTLs as an efficient HCV escape mechanism.

PD-1/PD-L1 BLOCKADE AS A THERAPUTIC TOOL

As previously commented, a defective virus-specific cytotoxic T cell response is one of the most important causes of host inability to eliminate a persistent viral infection. Several studies have highlighted the role of the PD-1/PD-L1 pathway in the development of anergy on virus-specific CD8+ T cells, and how PD-1/PD-L1 blockade could enhance virus-specific CD8+ T cell functionality in-vitro[41,74,88-92]. Recently, several works have been carried out to analyse whether modulation of the PD-1/PD-L1 pathway could improve T cell response against persistent viral infections either directly, using anti PD-L1 antibodies alone, or in combination with a therapeutic vaccine. Therapeutic vaccine usually fails to induce a vigorous T cell response due to the tolerogenic-like status of HCV-specific T cells[93,94]. This scenario could be positive if negative co-stimulatory molecules, such as PD-1, were blocked when the therapeutic vaccine is administered in order to enhance the specific immune response against the supplied epitopes. In the chronic LCMV infection animal model, the administration of a therapeutic vaccine in combination with PD-1/PD-L1 interaction blockade enhances expansion and improves the function of LCMV-specific CD8+ T cells. In addition, this combinatorial therapeutic vaccination accelerates viral control compared with either therapeutic vaccine or PD-1 blockade alone[95]. Moreover, the effect of anti-PD-L1 antibodies alone could also be effective in controlling persistent viral infection by restoring specific CTL response. The administration of anti-PD-L1 monoclonal antibodies during simian immunodeficiency virus (SIV) chronic infection in macaques resulted in a rapid expansion and restoration of SIV-specific CD8+ T cells[96,97]. Although these results seem to be quite promising, the blockade of negative co-stimulatory pathways could lead to the development of autoimmune diseases[17,18], which could prevent the use of this strategy as a therapeutic tool in humans. Therefore, more research is necessary in this field before blockade of the PD-1/PD-L1 pathway is suitable for the treatment of chronic HCV infection.

CONCLUSION

In summary, the PD-1/PD-L1 pathway displays an important role in the induction of anergy on HCV-specific cytotoxic T cells, and could be important in the development of HCV persistent infection. Blocking the PD-1/PD-L1 interaction, probably in association with the modulation of other co-stimulatory molecules, could be an interesting strategy to restore HCV-specific CTL response in patients unresponsive to standard anti-HCV treatment.

Footnotes

Peer reviewers: Rudi Beyaert, Professor, Department of Molecular Biomedical Research, Flanders Interuniversity Institute for Biotechnology and Ghent University, Technologiepark 927, B-9052 Gent, Belgium; Sabine Mihm, Professor, Department of Gastroenterology, Georg-August-Universität, Robert-Koch-Str.40, Göttingen D-37099, Germany

S- Editor Tian L L- Editor Webster JR E- Editor Lin YP

References
1.  Lauer GM, Walker BD. Hepatitis C virus infection. N Engl J Med. 2001;345:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Bowen DG, Walker CM. Adaptive immune responses in acute and chronic hepatitis C virus infection. Nature. 2005;436:946-952.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Liang TJ, Heller T. Pathogenesis of hepatitis C-associated hepatocellular carcinoma. Gastroenterology. 2004;127:S62-S71.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Afdhal NH. The natural history of hepatitis C. Semin Liver Dis. 2004;24 Suppl 2:3-8.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Fried MW, Shiffman ML, Reddy KR, Smith C, Marinos G, Gonçales FL Jr, Häussinger D, Diago M, Carosi G, Dhumeaux D. Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N Engl J Med. 2002;347:975-982.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Manns MP, McHutchison JG, Gordon SC, Rustgi VK, Shiffman M, Reindollar R, Goodman ZD, Koury K, Ling M, Albrecht JK. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet. 2001;358:958-965.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Hayashi N, Takehara T. Antiviral therapy for chronic hepatitis C: past, present, and future. J Gastroenterol. 2006;41:17-27.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Grüner NH, Gerlach TJ, Jung MC, Diepolder HM, Schirren CA, Schraut WW, Hoffmann R, Zachoval R, Santantonio T, Cucchiarini M. Association of hepatitis C virus-specific CD8+ T cells with viral clearance in acute hepatitis C. J Infect Dis. 2000;181:1528-1536.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Spangenberg HC, Viazov S, Kersting N, Neumann-Haefelin C, McKinney D, Roggendorf M, von Weizsäcker F, Blum HE, Thimme R. Intrahepatic CD8+ T-cell failure during chronic hepatitis C virus infection. Hepatology. 2005;42:828-837.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Gruener NH, Lechner F, Jung MC, Diepolder H, Gerlach T, Lauer G, Walker B, Sullivan J, Phillips R, Pape GR. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J Virol. 2001;75:5550-5558.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Mueller DL, Jenkins MK, Schwartz RH. Clonal expansion versus functional clonal inactivation: a costimulatory signalling pathway determines the outcome of T cell antigen receptor occupancy. Annu Rev Immunol. 1989;7:445-480.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Bretscher P. The two-signal model of lymphocyte activation twenty-one years later. Immunol Today. 1992;13:74-76.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Nurieva R, Thomas S, Nguyen T, Martin-Orozco N, Wang Y, Kaja MK, Yu XZ, Dong C. T-cell tolerance or function is determined by combinatorial costimulatory signals. EMBO J. 2006;25:2623-2633.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Frauwirth KA, Thompson CB. Activation and inhibition of lymphocytes by costimulation. J Clin Invest. 2002;109:295-299.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Carter L, Fouser LA, Jussif J, Fitz L, Deng B, Wood CR, Collins M, Honjo T, Freeman GJ, Carreno BM. PD-1:PD-L inhibitory pathway affects both CD4(+) and CD8(+) T cells and is overcome by IL-2. Eur J Immunol. 2002;32:634-643.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. 2001;2:261-268.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999;11:141-151.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, Sasayama S, Mizoguchi A, Hiai H, Minato N. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291:319-322.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682-687.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765-772.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Zhang X, Schwartz JC, Guo X, Bhatia S, Cao E, Lorenz M, Cammer M, Chen L, Zhang ZY, Edidin MA. Structural and functional analysis of the costimulatory receptor programmed death-1. Immunity. 2004;20:337-347.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Grakoui A, John Wherry E, Hanson HL, Walker C, Ahmed R. Turning on the off switch: regulation of anti-viral T cell responses in the liver by the PD-1/PD-L1 pathway. J Hepatol. 2006;45:468-472.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Okazaki T, Honjo T. The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 2006;27:195-201.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci USA. 2001;98:13866-13871.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173:945-954.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Ishida M, Iwai Y, Tanaka Y, Okazaki T, Freeman GJ, Minato N, Honjo T. Differential expression of PD-L1 and PD-L2, ligands for an inhibitory receptor PD-1, in the cells of lymphohematopoietic tissues. Immunol Lett. 2002;84:57-62.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Yamazaki T, Akiba H, Iwai H, Matsuda H, Aoki M, Tanno Y, Shin T, Tsuchiya H, Pardoll DM, Okumura K. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol. 2002;169:5538-5545.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Liang SC, Latchman YE, Buhlmann JE, Tomczak MF, Horwitz BH, Freeman GJ, Sharpe AH. Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses. Eur J Immunol. 2003;33:2706-2716.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Wiendl H, Mitsdoerffer M, Schneider D, Chen L, Lochmüller H, Melms A, Weller M. Human muscle cells express a B7-related molecule, B7-H1, with strong negative immune regulatory potential: a novel mechanism of counterbalancing the immune attack in idiopathic inflammatory myopathies. FASEB J. 2003;17:1892-1894.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Salama AD, Chitnis T, Imitola J, Ansari MJ, Akiba H, Tushima F, Azuma M, Yagita H, Sayegh MH, Khoury SJ. Critical role of the programmed death-1 (PD-1) pathway in regulation of experimental autoimmune encephalomyelitis. J Exp Med. 2003;198:71-78.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Mühlbauer M, Fleck M, Schütz C, Weiss T, Froh M, Blank C, Schölmerich J, Hellerbrand C. PD-L1 is induced in hepatocytes by viral infection and by interferon-alpha and -gamma and mediates T cell apoptosis. J Hepatol. 2006;45:520-528.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Maier H, Isogawa M, Freeman GJ, Chisari FV. PD-1:PD-L1 interactions contribute to the functional suppression of virus-specific CD8+ T lymphocytes in the liver. J Immunol. 2007;178:2714-2720.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Meier A, Bagchi A, Sidhu HK, Alter G, Suscovich TJ, Kavanagh DG, Streeck H, Brockman MA, LeGall S, Hellman J. Upregulation of PD-L1 on monocytes and dendritic cells by HIV-1 derived TLR ligands. AIDS. 2008;22:655-658.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Willberg C, Barnes E, Klenerman P. HCV immunology--death and the maiden T cell. Cell Death Differ. 2003;10 Suppl 1:S39-S47.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Bartenschlager R, Lohmann V. Replication of hepatitis C virus. J Gen Virol. 2000;81:1631-1648.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Knolle PA, Gerken G. Local control of the immune response in the liver. Immunol Rev. 2000;174:21-34.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Knolle PA, Limmer A. Neighborhood politics: the immunoregulatory function of organ-resident liver endothelial cells. Trends Immunol. 2001;22:432-437.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Crispe IN. Hepatic T cells and liver tolerance. Nat Rev Immunol. 2003;3:51-62.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Griffith TS, Brunner T, Fletcher SM, Green DR, Ferguson TA. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science. 1995;270:1189-1192.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Isogawa M, Furuichi Y, Chisari FV. Oscillating CD8(+) T cell effector functions after antigen recognition in the liver. Immunity. 2005;23:53-63.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Nakamoto N, Kaplan DE, Coleclough J, Li Y, Valiga ME, Kaminski M, Shaked A, Olthoff K, Gostick E, Price DA. Functional restoration of HCV-specific CD8 T cells by PD-1 blockade is defined by PD-1 expression and compartmentalization. Gastroenterology. 2008;134:1927-1937, 1937.e1-e2.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Iwai Y, Terawaki S, Ikegawa M, Okazaki T, Honjo T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J Exp Med. 2003;198:39-50.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027-1034.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med. 1999;5:1365-1369.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Nowak MA, Bangham CR. Population dynamics of immune responses to persistent viruses. Science. 1996;272:74-79.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Boettler T, Spangenberg HC, Neumann-Haefelin C, Panther E, Urbani S, Ferrari C, Blum HE, von Weizsäcker F, Thimme R. T cells with a CD4+CD25+ regulatory phenotype suppress in vitro proliferation of virus-specific CD8+ T cells during chronic hepatitis C virus infection. J Virol. 2005;79:7860-7867.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Cabrera R, Tu Z, Xu Y, Firpi RJ, Rosen HR, Liu C, Nelson DR. An immunomodulatory role for CD4(+)CD25(+) regulatory T lymphocytes in hepatitis C virus infection. Hepatology. 2004;40:1062-1071.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Rushbrook SM, Ward SM, Unitt E, Vowler SL, Lucas M, Klenerman P, Alexander GJ. Regulatory T cells suppress in vitro proliferation of virus-specific CD8+ T cells during persistent hepatitis C virus infection. J Virol. 2005;79:7852-7859.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Gavin MA, Clarke SR, Negrou E, Gallegos A, Rudensky A. Homeostasis and anergy of CD4(+)CD25(+) suppressor T cells in vivo. Nat Immunol. 2002;3:33-41.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Raimondi G, Shufesky WJ, Tokita D, Morelli AE, Thomson AW. Regulated compartmentalization of programmed cell death-1 discriminates CD4+CD25+ resting regulatory T cells from activated T cells. J Immunol. 2006;176:2808-2816.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Polanczyk MJ, Hopke C, Vandenbark AA, Offner H. Treg suppressive activity involves estrogen-dependent expression of programmed death-1 (PD-1). Int Immunol. 2007;19:337-343.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Franceschini D, Paroli M, Francavilla V, Videtta M, Morrone S, Labbadia G, Cerino A, Mondelli MU, Barnaba V. PD-L1 negatively regulates CD4+CD25+Foxp3+ Tregs by limiting STAT-5 phosphorylation in patients chronically infected with HCV. J Clin Invest. 2009;119:551-564.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Kasprowicz V, Schulze Zur Wiesch J, Kuntzen T, Nolan BE, Longworth S, Berical A, Blum J, McMahon C, Reyor LL, Elias N. High level of PD-1 expression on hepatitis C virus (HCV)-specific CD8+ and CD4+ T cells during acute HCV infection, irrespective of clinical outcome. J Virol. 2008;82:3154-3160.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Golden-Mason L, Palmer B, Klarquist J, Mengshol JA, Castelblanco N, Rosen HR. Upregulation of PD-1 expression on circulating and intrahepatic hepatitis C virus-specific CD8+ T cells associated with reversible immune dysfunction. J Virol. 2007;81:9249-9258.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Radziewicz H, Ibegbu CC, Fernandez ML, Workowski KA, Obideen K, Wehbi M, Hanson HL, Steinberg JP, Masopust D, Wherry EJ. Liver-infiltrating lymphocytes in chronic human hepatitis C virus infection display an exhausted phenotype with high levels of PD-1 and low levels of CD127 expression. J Virol. 2007;81:2545-2553.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Golden-Mason L, Klarquist J, Wahed AS, Rosen HR. Cutting edge: programmed death-1 expression is increased on immunocytes in chronic hepatitis C virus and predicts failure of response to antiviral therapy: race-dependent differences. J Immunol. 2008;180:3637-3641.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Zhang Z, Jin B, Zhang JY, Xu B, Wang H, Shi M, Wherry EJ, Lau GK, Wang FS. Dynamic decrease in PD-1 expression correlates with HBV-specific memory CD8 T-cell development in acute self-limited hepatitis B patients. J Hepatol. 2009;50:1163-1173.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Ye P, Weng ZH, Zhang SL, Zhang JA, Zhao L, Dong JH, Jie SH, Pang R, Wei RH. Programmed death-1 expression is associated with the disease status in hepatitis B virus infection. World J Gastroenterol. 2008;14:4551-4557.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Peng G, Li S, Wu W, Tan X, Chen Y, Chen Z. PD-1 upregulation is associated with HBV-specific T cell dysfunction in chronic hepatitis B patients. Mol Immunol. 2008;45:963-970.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198-1202.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350-354.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Li YG, Chen LE, Chen GF, Wang FS. [Expressions and significance of B7-H1 and programmed death-1 in lymphocytes from patients with chronic hepatitis B virus infection]. Zhonghua Ganzangbing Zazhi. 2007;15:738-741.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Penna A, Pilli M, Zerbini A, Orlandini A, Mezzadri S, Sacchelli L, Missale G, Ferrari C. Dysfunction and functional restoration of HCV-specific CD8 responses in chronic hepatitis C virus infection. Hepatology. 2007;45:588-601.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Boni C, Fisicaro P, Valdatta C, Amadei B, Di Vincenzo P, Giuberti T, Laccabue D, Zerbini A, Cavalli A, Missale G. Characterization of hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection. J Virol. 2007;81:4215-4225.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Rutebemberwa A, Ray SC, Astemborski J, Levine J, Liu L, Dowd KA, Clute S, Wang C, Korman A, Sette A. High-programmed death-1 levels on hepatitis C virus-specific T cells during acute infection are associated with viral persistence and require preservation of cognate antigen during chronic infection. J Immunol. 2008;181:8215-8225.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Urbani S, Amadei B, Tola D, Massari M, Schivazappa S, Missale G, Ferrari C. PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. J Virol. 2006;80:11398-11403.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Boettler T, Panther E, Bengsch B, Nazarova N, Spangenberg HC, Blum HE, Thimme R. Expression of the interleukin-7 receptor alpha chain (CD127) on virus-specific CD8+ T cells identifies functionally and phenotypically defined memory T cells during acute resolving hepatitis B virus infection. J Virol. 2006;80:3532-3540.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Bengsch B, Spangenberg HC, Kersting N, Neumann-Haefelin C, Panther E, von Weizsäcker F, Blum HE, Pircher H, Thimme R. Analysis of CD127 and KLRG1 expression on hepatitis C virus-specific CD8+ T cells reveals the existence of different memory T-cell subsets in the peripheral blood and liver. J Virol. 2007;81:945-953.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Radziewicz H, Ibegbu CC, Hon H, Osborn MK, Obideen K, Wehbi M, Freeman GJ, Lennox JL, Workowski KA, Hanson HL. Impaired hepatitis C virus (HCV)-specific effector CD8+ T cells undergo massive apoptosis in the peripheral blood during acute HCV infection and in the liver during the chronic phase of infection. J Virol. 2008;82:9808-9822.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Wedemeyer H, He XS, Nascimbeni M, Davis AR, Greenberg HB, Hoofnagle JH, Liang TJ, Alter H, Rehermann B. Impaired effector function of hepatitis C virus-specific CD8+ T cells in chronic hepatitis C virus infection. J Immunol. 2002;169:3447-3458.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Folgori A, Spada E, Pezzanera M, Ruggeri L, Mele A, Garbuglia AR, Perrone MP, Del Porto P, Piccolella E, Cortese R. Early impairment of hepatitis C virus specific T cell proliferation during acute infection leads to failure of viral clearance. Gut. 2006;55:1012-1019.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Keir ME, Francisco LM, Sharpe AH. PD-1 and its ligands in T-cell immunity. Curr Opin Immunol. 2007;19:309-314.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Yao S, Chen L. Reviving exhausted T lymphocytes during chronic virus infection by B7-H1 blockade. Trends Mol Med. 2006;12:244-246.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Jeong HY, Lee YJ, Seo SK, Lee SW, Park SJ, Lee JN, Sohn HS, Yao S, Chen L, Choi I. Blocking of monocyte-associated B7-H1 (CD274) enhances HCV-specific T cell immunity in chronic hepatitis C infection. J Leukoc Biol. 2008;83:755-764.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Nakamoto N, Cho H, Shaked A, Olthoff K, Valiga ME, Kaminski M, Gostick E, Price DA, Freeman GJ, Wherry EJ. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathog. 2009;5:e1000313.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Arribillaga L, Sarobe P, Arina A, Gorraiz M, Borrás-Cuesta F, Ruiz J, Prieto J, Chen L, Melero I, Lasarte JJ. Enhancement of CD4 and CD8 immunity by anti-CD137 (4-1BB) monoclonal antibodies during hepatitis C vaccination with recombinant adenovirus. Vaccine. 2005;23:3493-3499.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Zabaleta A, Arribillaga L, Llopiz D, Dotor J, Lasarte JJ, Prieto J, Borrás-Cuesta F, Esteban JI, Quer J, Vayreda F. Induction of potent and long-lasting CD4 and CD8 T-cell responses against hepatitis C virus by immunization with viral antigens plus poly(I:C) and anti-CD40. Antiviral Res. 2007;74:25-35.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Nanji SA, Hancock WW, Anderson CC, Adams AB, Luo B, Schur CD, Pawlick RL, Wang L, Coyle AJ, Larsen CP. Multiple combination therapies involving blockade of ICOS/B7RP-1 costimulation facilitate long-term islet allograft survival. Am J Transplant. 2004;4:526-536.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DA, Wherry EJ. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol. 2009;10:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, Castelblanco N, Kuchroo V, Gretch DR, Rosen HR. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol. 2009;83:9122-9130.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Lopes AR, Kellam P, Das A, Dunn C, Kwan A, Turner J, Peppa D, Gilson RJ, Gehring A, Bertoletti A. Bim-mediated deletion of antigen-specific CD8 T cells in patients unable to control HBV infection. J Clin Invest. 2008;118:1835-1845.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Kittlesen DJ, Chianese-Bullock KA, Yao ZQ, Braciale TJ, Hahn YS. Interaction between complement receptor gC1qR and hepatitis C virus core protein inhibits T-lymphocyte proliferation. J Clin Invest. 2000;106:1239-1249.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Sundström S, Ota S, Dimberg LY, Masucci MG, Bergqvist A. Hepatitis C virus core protein induces an anergic state characterized by decreased interleukin-2 production and perturbation of mitogen-activated protein kinase responses. J Virol. 2005;79:2230-2239.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Yao ZQ, King E, Prayther D, Yin D, Moorman J. T cell dysfunction by hepatitis C virus core protein involves PD-1/PDL-1 signaling. Viral Immunol. 2007;20:276-287.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Lukens JR, Cruise MW, Lassen MG, Hahn YS. Blockade of PD-1/B7-H1 interaction restores effector CD8+ T cell responses in a hepatitis C virus core murine model. J Immunol. 2008;180:4875-4884.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Maillard P, Krawczynski K, Nitkiewicz J, Bronnert C, Sidorkiewicz M, Gounon P, Dubuisson J, Faure G, Crainic R, Budkowska A. Nonenveloped nucleocapsids of hepatitis C virus in the serum of infected patients. J Virol. 2001;75:8240-8250.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Masalova OV, Atanadze SN, Samokhvalov EI, Petrakova NV, Kalinina TI, Smirnov VD, Khudyakov YE, Fields HA, Kushch AA. Detection of hepatitis C virus core protein circulating within different virus particle populations. J Med Virol. 1998;55:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Xu B, Zhang Z, Shi Y, Chen XY, Wang FS. [PD-1 up-regulation influenced apoptosis of HBV-specific CD8 T cells in patients with acute resolved hepatitis B]. Zhonghua Yixue Zazhi. 2009;89:1158-1161.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Boasso A, Hardy AW, Landay AL, Martinson JL, Anderson SA, Dolan MJ, Clerici M, Shearer GM. PDL-1 upregulation on monocytes and T cells by HIV via type I interferon: restricted expression of type I interferon receptor by CCR5-expressing leukocytes. Clin Immunol. 2008;129:132-144.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Iglesias-Chiesa MC, Crabtree-Ramírez B, Reyes-Terán G. [A new therapeutic strategy to control HIV? The PD1 molecule and its role in inhibiting cellular immune responses]. Gac Med Mex. 2008;144:75-78.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Trautmann L, Chomont N, Sékaly RP. [Inhibition of the PD-1 pathway restores the effector function of HIV-specific T cells]. Med Sci (Paris). 2007;23:24-25.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Urbani S, Amadei B, Tola D, Pedrazzi G, Sacchelli L, Cavallo MC, Orlandini A, Missale G, Ferrari C. Restoration of HCV-specific T cell functions by PD-1/PD-L1 blockade in HCV infection: effect of viremia levels and antiviral treatment. J Hepatol. 2008;48:548-558.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  von Herrath MG, Berger DP, Homann D, Tishon T, Sette A, Oldstone MB. Vaccination to treat persistent viral infection. Virology. 2000;268:411-419.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Dikici B, Kalayci AG, Ozgenc F, Bosnak M, Davutoglu M, Ece A, Ozkan T, Ozeke T, Yagci RV, Haspolat K. Therapeutic vaccination in the immunotolerant phase of children with chronic hepatitis B infection. Pediatr Infect Dis J. 2003;22:345-349.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Ha SJ, Mueller SN, Wherry EJ, Barber DL, Aubert RD, Sharpe AH, Freeman GJ, Ahmed R. Enhancing therapeutic vaccination by blocking PD-1-mediated inhibitory signals during chronic infection. J Exp Med. 2008;205:543-555.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Velu V, Titanji K, Zhu B, Husain S, Pladevega A, Lai L, Vanderford TH, Chennareddi L, Silvestri G, Freeman GJ. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature. 2009;458:206-210.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Finnefrock AC, Tang A, Li F, Freed DC, Feng M, Cox KS, Sykes KJ, Guare JP, Miller MD, Olsen DB. PD-1 blockade in rhesus macaques: impact on chronic infection and prophylactic vaccination. J Immunol. 2009;182:980-987.  [PubMed]  [DOI]  [Cited in This Article: ]