icon star paper   Hepatitis B Articles (HBV)  
Back grey_arrow_rt.gif
 
 
ART May Reconstitute HBV Immunity
 
 
  "Effect of HIV Infection and Antiretroviral Therapy on Hepatitis B Virus (HBV) -Specific T Cell Responses in Patients Who Have Resolved HBV Infection"
 
The Journal of Infectious Diseases 2005;191:1169-1179
 
R. Monica Lascar,1,a A. Ross Lopes,2,3,a Richard J. Gilson,1 Claire Dunn,3 Ruth Johnstone,1 Andrew Copas,1 Stephanie Reignat,2 George Webster,2 Antonio Bertoletti,2 and Mala K. Maini1,2,3
 
1Centre for Sexual Health and HIV Research, 2Institute of Hepatology, and 3Division of Infection and Immunity, Royal Free and University College Medical School, London, United Kingdom
 
note from Jules Levin: Its also important to bear in mind that after starting HAART reconstitution of the immune system can cause ALT flares which may be a problem. So HBV+ patients should be monitored after starting HAART for this. In addition, resistance to 3TC can occur on HAART & this may result in ALT flares & harm to the liver (liver decompoensation). High HBV DNA (viral load) and low nadir CD4 counts may contribute to ALT flares occurring. When a patient is on HAART and if 3TC ror FTC esistance develops, it has been suggested to continue 3TC and add another HBV drug to reduce HBV DNA.
 
Worldwide 350-400 million people have chronic hepatitis B infection. 15-40% develop cirrhosis, liver failure or liver cancer. About 1 million people per year die from HBV associated liver disease. In the USA about 1.25 million people have chronic hepatitis B. 10-20% of people develop chronic hepatitis B after infection. 5-8% of people with HIV also have chronic hepatitis B.
 
"....we explored the effect of HIV immunodepletion and HAART-related immune restoration on reshaping immune responses to HBV infection. We observed a decrease in functionally active HBV-specific CD8+ T cell responses in association with HIV infection in HBV-immune patients..
 
....The findings of the present study could be extrapolated to suggest that, in patients treated for HBV/HIV coinfection, reconstitution of HBV-specific T cell responses may involve 2 distinct components: the reconstitution of responses associated with reduction in HBV load [20], as seen in treatment of HBV monoinfection [41, 42], and the more gradual, sustained reconstitution associated with the prolonged suppression of HIV viremia and reconstitution of HBV-specific CD4+ T cell responses demonstrated here. Thus, antiretroviral therapy can lead to an increase in functional CD4+ and CD8+ T cell responses to HBV infection, supporting the potential of HAART to reconstitute immune responses to clinically important pathogens...
 
ABSTRACT
Coinfection with hepatitis B virus (HBV) is a common occurrence in human immunodeficiency virus (HIV) -positive patients and an increasing cause of morbidity and mortality. The CD8+ T cell response is critical for long-term control of HBV in patients resolving acute infection. Here, we examine the effect of HIV on HBV-specific CD8+ T cell responses in patients who have resolved HBV infection. A cross-sectional study showed a reduction in HBV-specific CD8+ T cell responses in HIV-positive, HBV-immune patients, compared with those in HIV-negative, HBV-immune patients. A longitudinal study of a subgroup of patients examined whether this attrition could be reversed by effective antiretroviral therapy. The introduction of highly active antiretroviral therapy (HAART) resulted in reconstitution of some HBV-specific CD4+ and CD8+ T cell responses, in association with restoration of CD4+ T cell counts. These data provide a mechanism to account for the observed impairment of control of HBV infection in the setting of HIV infection and support the ability of HAART to reconstitute functionally active T cell responses.
 
"....In a group of 12 HAART-naive, HIV-positive patients who also had serological evidence of past HBV infection without a history of symptoms, HBV-specific CD8+ T cell responses were markedly diminished...
 
....... HBV-specific CD8+ T cell responses were detectable in the treated HIV-positive patients but not in the untreated HIV-positive, HBV-immune patients tested directly ex vivo...."
 
TEXT OF ARTICLE
Coinfection with hepatitis B virus (HBV) is a frequent occurrence in HIV-positive patients and an increasing cause of morbidity and mortality in the context of the prolonged survival associated with highly active antiretroviral therapy (HAART) [1 -4]. The proportion of HIV-positive patients with serological evidence of previous exposure to HBV ranges from 64% to 84% in published cohorts; 10% -15% of such patients are chronically infected with HBV [5, 6]. HIV-related immunodepletion influences the natural history of HBV infection. Epidemiological studies have revealed that HIV-positive patients are more likely to have a prolonged duration of acute illness after HBV infection and to have lower rates of clearance of hepatitis B e antigen [7]. They have higher circulating levels of HBV DNA and higher rates of reactivation of HBV infection [8]. These findings suggest impaired immune control of HBV infection during HIV infection.
 
In the present study, we explore, for the first time, the effect of HIV-related immunodepletion on HBV-specific CD8+ T cell responses in patients who have resolved HBV infection. Recent data have directly demonstrated that CD8+ T cells are critical effectors in the control of acute HBV infection [9]. HBV-specific CD8+ T cell responses are difficult to detect in patients chronically infected with HBV [10], making them an unsuitable group in which to study the impact of HIV infection. We therefore studied patients who resolved HBV infection, since, in the absence of HIV, such patients maintain strong CD4+ and CD8+ T cell responses for many years after complete clinical and serological recovery [11, 12]. Patients without a history of jaundice found to have natural immunity to HBV (hepatitis B core antibody [HBcAb] plus/minus hepatitis B surface antibody [HBsAb]) as part of routine screening before HBV vaccination have never previously been studied immunologically, with regard to their HBV-specific immune responses, but are common among HIV cohorts [5, 13]. HBV-specific responses were compared in HIV-positive and HIV-negative patients, with a focus on those who had resolved HBV infection without symptoms. We used the sensitive technique of intracellular cytokine staining for interferon (IFN) -E, to explore the breadth of functionally active HBV-specific CD8+ T cell responses across a range of previously defined HLA-A2 -restricted HBV epitopes [14 -16].
 
To investigate to what extent any loss of HBV-specific immune responses could be reversed by antiretroviral therapy, a group of HBV-immune patients starting HAART were studied prospectively. There is a growing body of evidence suggesting that HAART may lead to successful restoration of specific immune responses to previously encountered pathogens [17 -19]. A previous small study of HIV patients chronically infected with HBV suggested that HAART may be associated with restoration of HBV-specific responses once HBV load is reduced [20]. Natural immunity to HBV provided a good model system in which to further explore the potential for functional restoration of HBV-specific CD4+ and CD8+ T cell responses in the setting of controlled HBV replication.
 
PATIENTS, MATERIALS, AND METHODS
 
Patients and controls.
Patients were recruited from the Mortimer Market Centre and provided written, informed consent, and the local ethics committee approved the study. All patients had HBV serologic test results confirming natural immunity to HBV (hepatitis B surface antigen [HBsAg] negative, HBcAb positive, and HBsAb positive) and were further categorized according to whether they had a history consistent with symptomatic acute hepatitis B.
 
Thirty-two HIV-positive gay men with natural immunity to HBV were screened, to identify 16 HLA-A2 -positive patients. Two had a history of symptomatic acute hepatitis B, and 14 had no history of jaundice or other relevant symptoms. Patients had a median CD4+ T cell count of 320 cells/E L and a median HIV load of 36,000 copies/mL (table 1). Fourteen patients were HAART naive, and 2 patients were already receiving antiretroviral therapy; 4 HLA-A2 -positive, HAART-naive patients were followed up longitudinally after starting combination therapy. Twenty-five HIV-negative patients (tested within the previous year) with natural immunity to HBV were screened, to identify 14 HLA-A2 -positive patients, of whom 3 had a history of symptomatic acute hepatitis B.
 
The HIV-positive and HIV-negative groups had similar demographic characteristics, with all but 1 patient who resolved asymptomatic HBV infection being gay men, and with all being white, apart from 1 Asian patient in the HIV-positive group. The HIV-negative group had a mean age of 38 years (median, 39 years), and the HIV-positive group had a mean age of 39 years (median, 39 years).
 
Tissue typing. Screening for the HLA-A2 haplotype was performed by staining peripheral blood mononuclear cells (PBMCs) with an anti -HLA-A2 -positive monoclonal antibody (MAb) (Incstar), followed by fluorescein isothiocyanate -conjugated sheep anti -mouse IgG second-layer MAb and flow-cytometric analysis.
 
Synthetic HBV peptides and antigens. Peptides corresponding to the sequence of core 18 -27; envelope 183 -91, 335 -43, 338 -47, and 348 -57; and polymerase 455 -63, 502 -10, 575 -83, 655 -63, and 816 -24 regions of HBV genotype D were synthesized (Chiron Mimotopes). Hepatitis B core antigen (HBcAg) and HBsAg were produced in Escherichia coli strain K802 and were 90% pure. HBsAg was supplied by Rhein Biotech, and HBcAg was provided by G. Borisova (University of Latvia).
 
Production of T cell lines and intracellular IFN-Ei staining. PBMCs were seeded at a concentration of 3 A 106 cells/mL and stimulated with 1 E mol/L relevant peptide (the 10 peptides listed above). Recombinant interleukin (IL) -2 (10 IU/mL) (Boehringer Mannheim) was added on day 4 of cell culture. Cells were restimulated on day 10, with 1 E mol/L relevant peptide (5 h), the last 4 h with 10 Eg/mL brefeldin A (Sigma-Aldrich). Cells were stained with anti-CD8+ (Pharmingen), permeabilized with Cytofix/Cytoperm (Pharmingen), stained with anti -IFN-Ei mAb (R&D Systems), and analyzed by use of a FACScan flow cytometer with CELLQuest software (Becton Dickinson).
 
For enumeration of HBV-specific CD4+ T cells, PBMCs were suspended at a concentration of 3 A 106 cells/mL in RPMI 1640 and 5% human serum and stimulated with 1 E mol/L HBcAg or HBsAg for 6 -16 h, with addition of brefeldin A after 1 h. Cells were washed, stained with anti-CD4+ phycoerythrin (Pharmingen), and subjected to intracellular cytokine staining with anti -IFN-Ei or anti -IL-2 mAb (R&D Systems). For all intracellular cytokine staining experiments, responses were calculated by subtracting background production of IFN-Ei or IL-2 in a negative control well without peptide or antigen restimulation. Stimulation with anti-CD3 and anti-CD28 MAb (1 E mol/L each) or phorbol 12-myristate 13-acetate (3 ng/mL) and ionomycin (100 ng/mL), for 6 h in the presence of brefeldin A, was used as a positive control.
 
Statistical analysis. In the cross-sectional study of HIV-negative and HIV-positive patients, the nonparametric Fisher's exact test was used to compare the number of patients with any HBV-specific CD8+ T cell response detectable. The nonparametric Mann-Whitney U (Wilcoxon rank sum) test was used to compare the groups, with the number and size of T cell responses for each patient taken into account.
 
RESULTS
 
Reduction of HBV-specific CD8+ T cell responses in HIV-positive, HBV-immune patients. We initially conducted a cross-sectional study of HIV-positive and HIV-negative patients with natural immunity to HBV, either with or without a history of acute symptomatic infection. All patients were screened for CD8+ T cells producing IFN-Ei in response to a panel of 10 peptides representing frequently recognized HLA-A2 -restricted HBV epitopes, and results were calculated after subtraction of background staining in wells without peptide restimulation. In 3 HLA-A2 -positive, HIV-negative patients who had successfully resolved acute symptomatic HBV infection 3 -5 years before, we found peptide-specific production of IFN-Ei for 6 or 7 of the 10 epitopes tested (figure 1 and table 2). We found that HIV-negative patients who had resolved HBV infection without prior symptoms of acute infection also had detectable CD8+ T cell responses to at least 1 HLA-A2 -restricted epitope in 6 of 11 cases (figure 1 and table 2). In some patients (e.g., N9, whose last risk exposure to HBV was >10 years before), responses had a level of multispecificity and expansion potential similar to those seen in patients with symptomatic acute infection, who have been the basis of all previous immunological studies of patients resolving HBV infection.
 
In a similar group of 12 HAART-naive, HIV-positive patients who also had serological evidence of past HBV infection without a history of symptoms, HBV-specific CD8+ T cell responses were markedly diminished. This group was matched with the HIV-negative, HBV-immune group for sex, age, ethnic origin, and likely route of acquisition of HBV infection, and all were antiretroviral naive. HBV-specific CD8+ T cell responses were undetectable after 10 days of specific peptide stimulation, by both intracellular cytokine staining (figure 1) and tetramer staining (data not shown), apart from 1 low-level core 18 -27 response in P16. In an HIV-positive patient with a history of acute symptomatic hepatitis B (P10; 8 years before), intracellular cytokine staining for IFN-Ei after 10 days of in vitro stimulation showed recognition of 4 of 10 epitopes tested. The proportion of patients with any HBV-specific CD8+ T cell response and the total number and magnitude of responses was significantly greater for the whole HIV-negative group than for the whole HIV-positive group (P = .018, Fischer's exact test, and P = .006, Mann-Whitney U test, respectively). These differences remained significant in a comparison of the closely matched subsets in the HIV-negative and HIV-positive groups who resolved HBV infection without symptoms (P = .027, for the proportion of patients with at least 1 response, Fisher's exact test; P = .01, for the number and size of responses, Mann-Whitney U test) (figure 1).
 
Intracellular cytokine staining directly ex vivo, in a subgroup of 2 HBV-immune patients who resolved symptomatic infection and 2 HBV-immune patients who resolved asymptomatic infection, from both the HIV-negative and HIV-positive groups, confirmed that responses were detectable in all of the HIV-negative patients tested. In the HIV-positive group, responses were detected directly ex vivo in only 1 of the HIV-positive patients (in P10, who had a history of acute symptomatic HBV infection 8 years before, but not in P11, who had acute infection >15 years before, or in the patients with asymptomatic infection). In all patients studied by both methods, the breadth of the HBV-specific immune responses seen after direct ex vivo expansion was similar to that seen after in vitro expansion.
 
HBV-specific CD8+ T cell responses in HIV-positive patients receiving antiretroviral therapy. To investigate any potential for reconstitution of HBV-specific CD8+ T cell responses, we initially examined responses in 2 patients who had resolved HBV infection without symptoms and were already receiving treatment for HIV infection. P12 started zidovudine and lamivudine at a CD4+ T cell count nadir of 240 cells/E L; at the time that samples were obtained, he had been maintained on this regimen for 3 years (at which time his CD4+ T cell count was 350 cells/E L and his HIV load was 500 copies/mL). P2 had been receiving HAART for 6 months, but lamivudine was substituted for didanosine 1 month before samples were obtained. His CD4+ T cell count increased from a nadir of 160 cells/E L to 700 cells/E L, and his HIV load was suppressed to <50 copies/mL. Both of these patients had CD8+ T cell responses to 3 of the 10 HBV-specific, HLA-A2 -restricted epitopes tested, which, after 10 days of in vitro expansion, were present at a magnitude similar to that seen in the HIV-negative, HBV-immune patients. This contrasted with the lack of responses detectable in 11 of the 12 untreated HIV-positive patients who had an equivalent pattern of HBV immunity and no history of symptomatic acute infection. HBV-specific CD8+ T cell responses were detectable in the treated HIV-positive patients but not in the untreated HIV-positive, HBV-immune patients tested directly ex vivo.
 
Reconstitution of HBV-specific CD8+ T cell responses on longitudinal study of patients starting HAART. The cross-sectional data suggested a decrease in HBV-specific CD8+ T cell responses in HIV-positive, HBV-immune patients and a possible reconstitution of such responses in those receiving HAART. We therefore longitudinally studied the impact of HAART on HBV-specific immune responses. Four patients who had resolved HBV infection (without a history of asymptomatic acute infection) and were starting HAART for progressive HIV infection were studied prospectively, and sequential blood samples were obtained before and during therapy. The drug regimens used are shown in table 1 and figure 3A and were selected by the clinician independently of participation in the present study. These patients were screened for functionally active HBV-specific CD8+ T cell responses on 2 -6 occasions by use of the same panel of 10 peptides applied in the cross-sectional studies.
 
The temporal relationship between increase in CD4+ T cell count, suppression of HIV load, and reconstitution of HBV-specific CD8+ T cell responses for each of the 4 patients is presented in figure 3A. Patients were screened up to 2 years after starting HAART, and, 3 years after starting HAART, samples were again obtained from any patients with reconstitution of HBV-specific CD8+ T cell responses. In 2 patients, the increase in CD4+ T cell count and suppression of HIV load were accompanied by detection of HBV-specific CD8+ T cell responses by 24 weeks of antiretroviral treatment. The epitopes detected were from the envelope group; 1 patient also recovered a response to core 18 -27. HBV-specific CD8+ T cell responses were still detectable when samples were again obtained from P1 and P7, 3 years after starting HAART, and, in P1, expanded to higher frequencies than at earlier sampling times. This is compatible with the finding of persistent CD8+ T cell responses in the patients studied cross-sectionally 6 months to 3 years after starting antiretroviral therapy.
 
Direct ex vivo analysis of P7 revealed reconstitution of HBV-specific CD8+ T cell responses at week 24, which is consistent in specificity and timing with the in vitro data, and a simultaneous decrease in levels of CD8+ specific for an HIV-1 -specific, HLA-A2 -restricted epitope (gag 77 -85; data not shown). This patient was also tested directly ex vivo for HBcAg-specific CD4+ T cell responses, by intracellular IFN-Ei staining, which revealed a parallel increase in HBV-specific CD4+ and CD8+ T cell responsiveness.
 
Parallel between reconstitution of HBV-specific CD4+ T cell responses and HAART-induced increases in total CD4+ T cells. To further investigate the potential role that enhanced CD4+ T cell help plays in recovering HBV-specific CD8+ T cell responses in these patients, we screened all the patients being longitudinally studied for HBV-specific CD4+ T cell responses at each time point. In light of recent data highlighting the potential importance of IL-2 -producing CD4+ T cells in maintaining adequate antiviral CD8+ T cell responses (reviewed in [21]), patients were tested for the presence of CD4+ T cells able to produce IFN-Ei and/or IL-2 on stimulation with HBcAg and HBsAg. The assay used to measure CD4+ intracellular production of IFN-Ei and/or IL-2, with or without stimulation with HBcAg or HBsAg, is shown in figure 4A. As illustrated by the fluorescence-activated cell sorter dot-plot profiles for this treated patient (P1; week 150), responses to overnight antigenic stimulation were easily detectable in some cases (IL-2 -positive CD4+ responding to HBcAg). In other examples (IFN-Ei -positive CD4+ responding to HBcAg and HBsAg), the responding populations were only slightly greater than background levels but could be confirmed by their ability to increase after a 10-day antigen-specific in vitro expansion.
 
The temporal relationship between CD4+ T cell responses and changes in CD4+ T cell count and HIV load is shown in figure 4B. All 4 HAART-treated patients developed detectable CD4+ T cell responses to both HBcAg and HBsAg from week 6 onward, which persisted for the duration of follow-up (at least 6 months in 2 patients and at least 3 years in the other 2). Three of the 4 patients were given lamivudine-containing combinations; P5, who also developed strong HBV-specific CD4+ T cell responses, was not treated with any drug with anti-HBV activity. All patients achieved adequate viral suppression and good total CD4+ T cell count recovery/increases, but the patient with the highest CD4+ T cell count nadir, lowest starting HIV load, and most rapid suppression of HIV load (P1) had the most effective reconstitution of both CD4+ and CD8+ T cell responses.
 
Figure 4C summarizes the lack of CD4+ T cell responses to HBcAg and HBsAg in 5 HBV-immune patients with HIV-related CD4+ lymphopenia (CD4+ T cell count <350 cells/E L) before the start of antiretroviral therapy. This contrasts with the HBV-specific CD4+ T cell responses in 6 HIV-negative patients who had also resolved asymptomatic HBV infection. The peak responses seen in the HIV-positive patients receiving HAART were comparable in frequency and magnitude to those seen in the HIV-negative patients.
 
AUTHOR DISCUSSION
In the present study, we explored the effect of HIV immunodepletion and HAART-related immune restoration on reshaping immune responses to HBV infection. We observed a decrease in functionally active HBV-specific CD8+ T cell responses in association with HIV infection in HBV-immune patients. Accumulating data suggest that residual virus is kept under tight control by an ongoing immune response in such patients [11, 12, 22]. Thus, the reduction in HBV-specific T cell responses observed in the present study provides a mechanism that could contribute to the increased risk of reactivation of hepatitis B surface antigenemia seen in HIV infection and other situations of clinical immunodepletion [23, 24].
 
To date, immunological studies of HBV immunity after acute infection have only included patients presenting with symptomatic infection. However, a large proportion of patients are found to have immunity to HBV without having had any prior symptoms of acute infection. Although the duration since the initial exposure to HBV was not always known, a number of HIV-negative patients in the potentially heterogeneous group in the present study had preserved HBV-specific responses over the course of many years. Although we cannot exclude the possibility that HIV-positive patients had a longer interval between resolution of HBV infection and the time that samples were obtained, the efficient long-term preservation of HBV-specific CD8+ T cell responses makes this unlikely to account for their lack of responsiveness.
 
Data describing the impact of HIV on responses to other viruses suggest that the persistence of detectable responses is related to the magnitude of antigen-specific response generated by each virus. For example, Epstein-Barr virus (EBV) and cytomegalovirus (CMV) are both associated with stable high-frequency memory cytotoxic T lymphocyte (CTL) populations, which, although reduced by HIV infection, remain easily detectable [18, 25]. Since the responses generated in association with control of HBV infection are typically lower in frequency than those to viruses such as EBV and CMV, it is not surprising that few remain detectable in HIV-positive patients. A recent study [26] including long-term follow-up of HIV-positive patients revealed maintenance of numbers but functional impairment of the EBV-specific CD8+ T cell response associated with subtle increases in EBV load and progression to non-Hodgkin lymphoma. Thus, careful long-term assessment of HIV-positive, HBV-immune patients would be required to examine the virological impact of the impaired HBV-specific CD8+ T cell response in this group of patients. A reduction of HBV-specific CD8+ T cell responses after HIV infection would be consistent with the demonstrated ability of HIV to induce apoptosis of CTLs of unrelated specificities through FasL-mediated [27] and tumor necrosis factor -mediated [28] counterattack. Similarly, data from murine models show that heterologous viral infections can quantitatively and qualitatively alter the memory pool of existing antiviral CD8+ [29, 30].
 
Our previous study of HIV-positive patients with ongoing HBV infection revealed the potential to recover some HBV-specific T cell responses in patients receiving HAART, in association with the reduction in HBV load induced by anti-HBV agents [20]. By contrast, in the present study, we investigated a group of patients in whom HBV load was already efficiently suppressed, to dissect the potential contribution of HAART-mediated immune reconstitution to the restoration of HBV-specific T cell responses. There is evidence from studies of the herpes viridae (EBV, CMV, and Kaposi sarcoma -associated herpesvirus) that HAART can restore specific T cell frequencies [17, 18, 31, 32], and this correlates with a decrease in end-organ disease [4, 33, 34]. The 2 patients studied cross-sectionally and 2 of the 4 patients studied longitudinally restored functionally active HBV-specific CD8+ T cell responses that had expansion potential after in vitro stimulation. That CD8+ T cell responses were reconstituted only in a proportion of these HBV-immune patients is in accordance with our findings in HIV-negative patients without a history of symptomatic HBV infection and are in line with those of a study of the impact of HIV infection and antiretroviral therapy on responses to Mycobacterium avium complex [35].
 
An important contribution to the reconstitution of HBV-specific CD8+ T cell responses is likely to be a restoration of CD4+ T cell help associated with the increasing CD4+ T cell counts. T cell help is known to be important for maintaining functionally active CD8+ T cell responses [36], and recent data suggest that IL-2 -producing CD4+ T cells play a key role in antiviral immunity [21, 37]. In both of the patients with restoration of HBV-specific CD8+ T cell responses, we observed a concomitant reconstitution of IFN-Ei -positive and IL-2 -positive CD4+ T cell responses to HBcAg and HBsAg after the start of HAART. These HBV-specific CD4+ T cell responses were similarly observed in a patient receiving an antiretroviral combination that did not include lamivudine or other drugs with anti-HBV activity. Importantly, we demonstrated the potential for long-term maintenance of both CD4+ and CD8+ T cells during prolonged follow-up of 2 patients, in contrast to the rapid but short-lived effects of lamivudine on HBV-specific T cell function reported in patients with ongoing HBV infection [20, 38]. There is a temporal correlation between the reconstitution of T cell responses observed in the present study and that seen in other studies of HAART-mediated reconstitution of T cell function (reviewed in [39]), with initial reconstitution at `3 months and subsequent reconstitution continuing gradually for at least 2 years [40]. These findings should be extended to a larger group of patients, since the present study was not powered to detect an effect of CD4+ T cell count nadir, magnitude of changes in CD4+ T cell count, or HIV load on the extent of HBV-specific T cell reconstitution. However, it is worth noting that the patient with the most effective reconstitution of HBV-specific CD4+ and CD8+ T cell responses was the one who started HAART with the highest CD4+ T cell count nadir and the lowest HIV load and who achieved the most efficient containment of HIV to undetectable levels.
 
The findings of the present study could be extrapolated to suggest that, in patients treated for HBV/HIV coinfection, reconstitution of HBV-specific T cell responses may involve 2 distinct components: the reconstitution of responses associated with reduction in HBV load [20], as seen in treatment of HBV monoinfection [41, 42], and the more gradual, sustained reconstitution associated with the prolonged suppression of HIV viremia and reconstitution of HBV-specific CD4+ T cell responses demonstrated here. Thus, antiretroviral therapy can lead to an increase in functional CD4+ and CD8+ T cell responses to HBV infection, supporting the potential of HAART to reconstitute immune responses to clinically important pathogens.
 
References
 
1. Thio CL, Seaberg EC, Skolasky R Jr, et al. HIV-1, hepatitis B virus, and risk of liver-related mortality in the Multicenter Cohort Study (MACS). Lancet 2002; 360:1921 -6. First citation in article | PubMed
 
2. Puoti M, Spinetti A, Ghezzi A, et al. Mortality for liver disease in patients with HIV infection: a cohort study. J Acquir Immune Defic Syndr 2000; 24:211 -7. First citation in article | PubMed
 
3. Martin-Carbonero L, Soriano V, Valencia E, Garcia-Samaniego J, Lopez M, Gonzalez-Lahoz J. Increasing impact of chronic viral hepatitis on hospital admissions and mortality among HIV-infected patients. AIDS Res Hum Retroviruses 2001; 17:1467 -71. First citation in article | PubMed
 
4. Palella FJ Jr, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338:853 -60. First citation in article | PubMed
 
5. Hadler SC, Judson FN, O'Malley PM, et al. Outcome of hepatitis B virus infection in homosexual men and its relation to prior human immunodeficiency virus infection. J Infect Dis 1991; 163:454 -9. First citation in article | PubMed
 
6. Scharschmidt BF, Held MJ, Hollander HH, et al. Hepatitis B in patients with HIV infection: relationship to AIDS and patient survival. Ann Intern Med 1992; 117:837 -8. First citation in article | PubMed
 
7. Colin JF, Cazals-Hatem D, Loriot MA, et al. Influence of human immunodeficiency virus infection on chronic hepatitis B in homosexual men. Hepatology 1999; 29:1306 -10. First citation in article | PubMed
 
8. Gilson RJ, Hawkins AE, Beecham MR, et al. Interactions between HIV and hepatitis B virus in homosexual men: effects on the natural history of infection. AIDS 1997; 11:597 -606. First citation in article | PubMed
 
9. Thimme R, Wieland S, Steiger C, et al. CD8+ T cells mediate viral clearance and disease pathogenesis during acute hepatitis B virus infection. J Virol 2003; 77:68 -76. First citation in article | PubMed
 
10. Maini MK, Boni C, Lee CK, et al. The role of virus-specific CD8+ cells in liver damage and viral control during persistent hepatitis B virus infection. J Exp Med 2000; 191:1269 -80. First citation in article | PubMed
 
11. Penna A, Artini M, Cavalli A, et al. Long-lasting memory T cell responses following self-limited acute hepatitis B. J Clin Invest 1996; 98:1185 -94. First citation in article | PubMed
 
12. Rehermann B, Ferrari C, Pasquinelli C, Chisari FV. The hepatitis B virus persists for decades after patients' recovery from acute viral hepatitis despite active maintenance of a cytotoxic T-lymphocyte response. Nat Med 1996; 2:1104 -8. First citation in article | PubMed
 
13. Francisci D, Baldelli F, Papili R, Stagni G, Pauluzzi S. Prevalence of HBV, HDV and HCV hepatitis markers in HIV-positive patients. Eur J Epidemiol 1995; 11:123 -6. First citation in article | PubMed
 
14. Penna A, Chisari FV, Bertoletti A, et al. Cytotoxic T lymphocytes recognize an HLA-A2 -restricted epitope within the hepatitis B virus nucleocapsid antigen. J Exp Med 1991; 174:1565 -70. First citation in article | PubMed
 
15. Nayersina R, Fowler P, Guilhot S, et al. HLA A2 restricted cytotoxic T lymphocyte responses to multiple hepatitis B surface antigen epitopes during hepatitis B virus infection. J Immunol 1993; 150:4659 -71. First citation in article | PubMed
 
16. Rehermann B, Fowler P, Sidney J, et al. The cytotoxic T lymphocyte response to multiple hepatitis B virus polymerase epitopes during and after acute viral hepatitis. J Exp Med 1995; 181:1047 -58. First citation in article | PubMed
 
17. Wilkinson J, Cope A, Gill J, et al. Identification of Kaposi's sarcoma -associated herpesvirus (KSHV) -specific cytotoxic T-lymphocyte epitopes and evaluation of reconstitution of KSHV-specific responses in human immunodeficiency virus type 1 -infected patients receiving highly active antiretroviral therapy. J Virol 2002; 76:2634 -40. First citation in article | PubMed
 
18. Dalod M, Dupuis M, Deschemin JC, et al. Broad, intense anti -human immunodeficiency virus (HIV) ex vivo CD8+ responses in HIV type 1 -infected patients: comparison with anti -Epstein-Barr virus responses and changes during antiretroviral therapy. J Virol 1999; 73:7108 -16. First citation in article | PubMed
 
19. Kostense S, Otto SA, Knol GJ, et al. Functional restoration of human immunodeficiency virus and Epstein-Barr virus -specific CD8+ T cells during highly active antiretroviral therapy is associated with an increase in CD4+ T cells. Eur J Immunol 2002; 32:1080 -9. First citation in article | PubMed
 
20. Lascar RM, Gilson RJ, Lopes AR, Bertoletti A, Maini MK. Reconstitution of hepatitis B virus (HBV) -specific T cell responses with treatment of human immunodeficiency virus/HBV coinfection. J Infect Dis 2003; 188:1815 -9. First citation in article | Full Text | PubMed
 
21. Day CL, Walker BD. Progress in defining CD4 helper cell responses in chronic viral infections. J Exp Med 2003; 198:1773 -7. First citation in article | PubMed
 
22. Maini MK, Boni C, Ogg GS, et al. Direct ex vivo analysis of hepatitis B virus -specific CD8+ T cells associated with the control of infection. Gastroenterology 1999; 117:1386 -96. First citation in article | PubMed
 
23. Chazouilleres O, Mamish D, Kim M, et al. "Occult" hepatitis B virus as source of infection in liver transplant recipients. Lancet 1994; 343:142 -6. First citation in article | PubMed
 
24. Waite J, Gilson RJ, Weller IV, et al. Hepatitis B virus reactivation or reinfection associated with HIV-1 infection. AIDS 1988; 2:443 -8. First citation in article | PubMed
 
25. Komanduri KV, Donahoe SM, Moretto WJ, et al. Direct measurement of CD4+ and CD8+ T-cell responses to CMV in HIV-1 -infected subjects. Virology 2001; 279:459 -70. First citation in article | PubMed
 
26. van Baarle D, Hovenkamp E, Callan MF, et al. Dysfunctional Epstein-Barr virus (EBV) -specific CD8+ T lymphocytes and increased EBV load in HIV-1 infected individuals progressing to AIDS-related non-Hodgkin lymphoma. Blood 2001; 98:146 -55. First citation in article | PubMed
 
27. Xu XN, Screaton GR, McMichael AJ. Virus infections: escape, resistance, and counterattack. Immunity 2001; 15:867 -70. First citation in article | PubMed
 
28. Herbein G, Mahlknecht U, Batliwalla F, et al. Apoptosis of CD8+ T cells is mediated by macrophages through interaction of HIV gp120 with chemokine receptor CXCR4. Nature 1998; 395:189 -94. First citation in article | PubMed
 
29. Selin LK, Lin MY, Kraemer KA, et al. Attrition of T cell memory: selective loss of LCMV epitope -specific memory CD8 T cells following infections with heterologous viruses. Immunity 1999; 11:733 -42. First citation in article | PubMed
 
30. McNally JM, Zarozinski CC, Lin MY, Brehm MA, Chen HD, Welsh RM. Attrition of bystander CD8 T cells during virus-induced T-cell and interferon responses. J Virol 2001; 75:5965 -76. First citation in article | PubMed
 
31. Rinaldo CR Jr, Huang XL, Fan Z, et al. Anti -human immunodeficiency virus type 1 (HIV-1) CD8+ T-lymphocyte reactivity during combination antiretroviral therapy in HIV-1 -infected patients with advanced immunodeficiency. J Virol 2000; 74:4127 -38. First citation in article | PubMed
 
32. Casazza JP, Betts MR, Picker LJ, Koup RA. Decay kinetics of human immunodeficiency virus -specific CD8+ T cells in peripheral blood after initiation of highly active antiretroviral therapy. J Virol 2001; 75:6508 -16. First citation in article | PubMed
 
33. Komanduri KV, Viswanathan MN, Wieder ED, et al. Restoration of cytomegalovirus-specific CD4+ T-lymphocyte responses after ganciclovir and highly active antiretroviral therapy in individuals infected with HIV-1. Nat Med 1998; 4:953 -6. First citation in article | PubMed
 
34. Ledergerber B, Telenti A, Egger M. Risk of HIV related Kaposi's sarcoma and non-Hodgkin's lymphoma with potent antiretroviral therapy: prospective cohort study. Swiss HIV Cohort Study. BMJ 1999; 319:23 -4. First citation in article | PubMed
 
35. Havlir DV, Schrier RD, Torriani FJ, Chervenak K, Hwang JY, Boom WH. Effect of potent antiretroviral therapy on immune responses to Mycobacterium avium in human immunodeficiency virus -infected subjects. J Infect Dis 2000; 182:1658 -63. First citation in article | Full Text | PubMed
 
36. Kalams SA, Walker BD. The critical need for CD4 help in maintaining effective cytotoxic T lymphocyte responses. J Exp Med 1998; 188:2199 -204. First citation in article | PubMed
 
37. Younes SA, Yassine-Diab B, Dumont AR, et al. HIV-1 viremia prevents the establishment of interleukin 2 -producing HIV-specific memory CD4+ T cells endowed with proliferative capacity. J Exp Med 2003; 198:1909 -22. First citation in article | PubMed
 
38. Boni C, Penna A, Bertoletti A, et al. Transient restoration of anti-viral T cell responses induced by lamivudine therapy in chronic hepatitis B. J Hepatol 2003; 39:595 -605. First citation in article | PubMed
 
39. Carcelain G, Debre P, Autran B. Reconstitution of CD4+ T lymphocytes in HIV-infected individuals following antiretroviral therapy. Curr Opin Immunol 2001; 13:483 -8. First citation in article | PubMed
 
40. Notermans DW, Pakker NG, Hamann D, et al. Immune reconstitution after 2 years of successful potent antiretroviral therapy in previously untreated human immunodeficiency virus type 1 -infected adults. J Infect Dis 1999; 180:1050 -6. First citation in article | PubMed
 
41. Boni C, Bertoletti A, Penna A, et al. Lamivudine treatment can restore T cell responsiveness in chronic hepatitis B. J Clin Invest 1998; 102:968 -75. First citation in article | PubMed
 
42. Boni C, Penna A, Ogg GS, et al. Lamivudine treatment can overcome cytotoxic T-cell hyporesponsiveness in chronic hepatitis B: new perspectives for immune therapy. Hepatology 2001; 33:963 -71. First citation in article | PubMed
 
 
 
 
 
  icon paper stack View Older Articles   Back to Top   www.natap.org