Open access

Prevalence of Reactivation of Hepatitis B Virus DNA Replication in Rheumatoid Arthritis Patients

Written By

Yukitomo Urata, Ryoko Uesato and Dai Tanaka

Submitted: 11 November 2010 Published: 01 August 2011

DOI: 10.5772/20972

From the Edited Volume

DNA Replication - Current Advances

Edited by Herve Seligmann

Chapter metrics overview

3,630 Chapter Downloads

View Full Metrics

1. Introduction

1.1. Hepatitis B virus and rheumatoid arthritis

More than one-third of the worldwide population is infected with hepatitis B virus (HBV), and 350 million individuals have chronic HBV infection [1], with 75% of those living in the Southeast Asia and Western Pacific regions. HBV infection is a leading cause of cirrhosis and hepatocellular carcinoma (HCC) [2] and is estimated to be responsible for 500,000–700,000 deaths annually. Reactivation of hepatitis B in patients undergoing immunosuppressive therapy is a clinically important complication [3-5]. Hepatitis B reactivation can be transient and clinically silent but is often severe and results in acute hepatic failure.

Two clinical scenarios contribute to the reactivation of hepatitis B. The first occurs in patients with chronic hepatitis B. Fulminant HBV has been reported in hepatitis B surface antigen (HBsAg)-positive patients with rheumatoid arthritis (RA) taking tumor necrosis factor agents (TNFA) [6, 7].

Second, reactivation of hepatitis B occurs in patients with resolved hepatitis B. In these patients, low levels of HBV replication have been shown to persist in the liver and in peripheral blood mononuclear cells for decades [8-10], and reactivation occurs after transplantation, immunosuppressive therapy, and allogeneic and autologous hematopoietic stem-cell transplantation, with the reappearance of HBsAg [11-15]. Reactivation of hepatitis B can occur in RA patients with resolved hepatitis B who are on immunosuppressive therapy, including corticosteroids (CS), methotrexate (MTX) [16], and TNFA [17,18], and can result in fulminant or lethal hepatitis [4]. Optimal management practices for this group of patients are unclear [9].

We performed this study to determine the rate of reactivation of HBV DNA replication in RA patients with resolved hepatitis B.

Advertisement

2. Materials and methods

2.1. Patients and methods

In our departments, 516 patients who were treated for RA between January 2008 and August 2009 fulfilled the American College of Rheumatology 1987 revised criteria for RA. All patients were evaluated for HBV markers, including HBsAg, anti-hepatitis B surface antibody (anti-HBs), and anti-hepatitis B core antibody (anti-HBc). HBV markers were detected using commercial enzyme immunoassays (HBsAg: ARCHITECT HBsAg QT, anti-HBs: ARCHITECT Anti-HBs, and anti-HBc: ARCHITECT Anti-HBc; Abbott Laboratories, Wiesbaden, Germany). If patients were HBsAg-positive or HBsAg-negative and anti-HBs- and/or anti-HBc-positive, HBV DNA levels were assessed. Sensitivity was 2 log copies/mL. When negative HBV DNA results were obtained, measurements were repeated every 3 months, and if HBV DNA became positive, measurements were repeated every month. Medications, including biologic agents, were generally not discontinued, irrespective of HBV DNA levels. All study protocols were approved by the ethics committees of the participating centers, and all patients provided written informed consent before enrolment.

2.2. Quantification of HBV DNA in blood by real-time PCR

HBV DNA levels were quantified using the automated COBAS TaqMan HBV Test version 2 0 (Roche, Basel, Switzerland). Samples were pretreated using the COBAS AmpliPrep System for amplification and quantification by real-time PCR and were analyzed using the COBAS TaqMan gene analyzer [19].

2.3. Statistical analysis

The Fisher’s exact test, Student’s t-test, and Mann–Whitney U test were used to compare baseline patient characteristics between subgroups. Two-tailed values of p ≤ 0.05 were regarded as significant. Cox regression hazard analyses were used to separately investigate the influence of biologic agents, MTX, CS, and disease-modifying antirheumatic drugs (DMARDs) on reactivation of HBV DNA replication. To identify the relative important of these factors, we performed a stepwise forward elimination multiple logistic regression model. All analyses were performed using JMP version 8.0 software (JMP Japan, Tokyo, Japan).

Advertisement

3. Results

Background characteristics of the 516 patients are listed in Table 1. Seven patients were HBsAg-positive, while 157 were HBsAg-negative and anti-HBs- and/or anti-HBc-positive (30.4%). No resolved hepatitis B patients were positive for HBV DNA at baseline.

Subjects were followed for 18 months, and HBV DNA became positive (3.44 log copies/mL) in 13 of 157 patients (8.3%), whereas hepatic function remained normal in all cases (Table 1). Details of patients developing reactivation of HBV DNA replication are listed in Table 2; 1 patient developed reactivation of HBV DNA replication twice, 10 patients showed HBV DNA replication during biologic agent therapy [etanercept (ETN), n = 8; abatacept, n = 2; adalimumab, n = 1; infliximab, n = 1; tocilizumab, n = 1; and rituximab, n = 1], whereas 3 patient showed replication without biologic agent therapy. Types of DMARDs and immunosuppressants used for RA treatment during the study and numbers of patients being administered each pharmacotherapy are shown in Table 3. In 2 of the 13 patients, HBV DNA became negative without therapy. In 10 of the 13 patients, HBV DNA became negative with entecavir therapy (mean, 3.3 months). In the remaining 1 patient, after HBV DNA became positive, she suddenly died due to unknown causes.

Exploratory analysis was conducted on factors that were potentially associated with HBV replication development (Table 3). Among resolved hepatitis B patients who did and did not

Baseline demographic, clinical, and laboratory characteristics HBV replication (+) HBV replication (-) P value
n 13 144
Age, years (mean) 66.6 ± 10.7 (67.6) 64.9 ± 11.8 (66.2) 0.670
Female, n 8 (61.5%) 114 (77.9%) 0.505
RA duration, years 8.0 ± 7.7 (4.7) 7.6 ± 9.0 (4.0) 0.241
CRP, mg/dL 0.92 ± 2.46 (0.09) 1.04 ± 2.11 (0.20) 0.218
ESR, mm/h 26.0 ± 30.0 (13.0) 26.1 ± 26.8 (15.0) 0.476
IgM RF, IU/mL 46.2 ± 34.0 (49.3) 88.0 ± 151.2 (24.5) 0.791
AST, U/L 25.5 ± 6.5 (27.0) 27.9 ± 16.4 (23.0) 0.688
ALT, U/L 19.9 ± 6.8 (20.5) 26.0 ± 19.2 (19.0) 0.959
IgG, mg/dL 1454 ± 573 (1382) 1432 ± 450 (1358) 0.604
Neutrophil count 3326 ± 1567 (2722) 4462 ± 2302 (3868) 0.063
Lymphocyte count 1503 ± 425 (1431) 1732 ± 813 (1562) 0.323

Table 1.

Values are given as mean ± standard deviation (median)RA, rheumatoid arthritis; CRP, C-reactive protein; ESR, erythrocyte sedimentation rate; Ig, immunoglobulin; RF, rheumatoid factor; AST, aspartate aminotransferase; ALT, alanine aminotransferaseComparison of hepatitis B virus (HBV) replication-positive and HBV replication-negative patients for baseline demographic, clinical, and laboratory characteristics

develop reactivation of HBV DNA replication, a significant difference was noted between the use of biologic agents (76.9% vs. 36.1%, respectively; p = 0.006), ETN (61.5% vs. 22.2%, respectively; p = 0.005), MTX (76.9% vs. 46.5%, respectively; p = 0.044), high-dose CS (15.4% vs. 1.4%; p = 0.035), and tacrolimus hydrate (30.8% vs. 5.6%; p = 0.010). Cox regression hazard analysis also revealed that biologic agent and ETN use can be as predictors for reactivation of HBV DNA replication. The hazard ratio (HR) for use of a biologic agent and etanercept was 10.9 (p = 0.008) and 6.9 (p = 0.001), respectively. Age at presentation, duration of RA, male gender, use of MTX and CS, dose of MTX and CS, levels of alanine aminotransferase and aspartate aminotransferase, immunoglobulin G level, neutrophil counts, and lymphoid cell counts were not associated with the reactivation of HBV DNA replication. The four variables extracted from the stepwise analysis were then entered as predictors of HBV DNA replication in a multivariate logistic regression model to determine their independent importance. The results of this model are shown in table 4. Predictive capacity was recognized for the use of tacrolimus hydrate only.

A recent study investigated 244 HBsAg-negative lymphoma patients receiving cytotoxic chemotherapy [13]. Reactivation of hepatitis B developed following therapy in 8 of these 244 patients (3.3%). Patients appeared to have a greater tendency to develop fulminant hepatic failure (3 of 8 patients, 37.5%). Direct DNA sequencing results confirmed that all 8 patients showed reactivation of hepatitis B from resolved hepatitis B. These patients were initially HBsAg-negative, and HBsAb- and/or HBcAb-positive, and serum liver enzyme levels were not elevated. At the time of hepatitis B reactivation, these patients became HBsAg positive. This change was associated with a more than 100-fold increase in serum HBV DNA levels, which occurred before the elevation of serum transaminases [13].

CD4+ T-helper cells may contribute to the control of HBV infection primarily by facilitating the induction and maintenance of HBV-specific cytotoxic T lymphocytes (CTL). MTX and

Table 2.

RA, rheumatoid arthritis; MTX, methotrexate; DMARDs, disease-modifying antirheumatic drugs; ALT, alanine aminotransferaseaThis patient sequentially received 3 biologic agents: infliximab, etanercept, and tocilizumab.bThis patient had HBV-DNA reactivation twice.cThis patient sequentially received 2 biologic agents: etanercept, and abatacept.Demographic, clinical, and laboratory characteristics of patients with HBV replication

Variables Number of patientsa P value HR (95% CI)
HBV replication
(+)
HBV replication
(-)
Total 13 144
Biologic agent 10 (76.9%) 52 (36.1%) 0.006 2.1 (1.5–3.1)
Adalimumab 1 (7.7%) 8 (5.6%) 0.550 1.4 (0.2–10.2)
Etanercept 8 (61.5%) 32 (22.2%) 0.005 2.8 (1.6–4.7)
Infliximab 1 (7.7%) 17 (11.8%) 1.000 0.7 (0.1–4.5)
Tocilizumab 1 (7.7%) 7 (4.9%) 0.507 1.6 (0.2–11.9)
Abatacept 2 (15.4%) 3 (20.8%) 0.055 7.4 (1.4–40.3)
Rituximab 1 (7.7%) 0 0.08
Methotrexate 10 (76.9%) 67 (46.5%) 0.044 1.7 (1.2–2.3)
mean dose 7.1 ± 1.9 mg/week 6.8 ± 1.9 mg/week 0.707
Corticosteroids 6 (46.2%) 57 (39.6%) 0.770 1.2 (0.6–2.2)
mean dose 12.7 ± 15.6 mg/day 5.7 ± 5.0 mg/day 0.533
High dose of corticosteroids
(≥0.5mg·kg-1·day-1)
2 (15.4%)
2 (1.4%) 0.035 11.1 (1.7–72.3)
Sulfasalazine 1 (7.7%) 36 (25.0%) 0.303 0.3 (0.0–2.1)
Bucillamine 3 (23.1%) 29 (20.1%) 0.729 1.1 (0.4–3.3)
Tacrolimus hydrate 4 (30.8%) 8 (5.6%) 0.010 5.5 (1.9–15.9)
Sodium aurothiomalate 1 (7.7%) 5 (3.5%) 0.410 2.2 (0.3–17.6)
Leflunomide 1 (7.7%) 2 (1.4%) 0.230 5.5 (0.5–57.1)
d-penicillamine 0 2 (1.4%) 1.000
Actarit 0 1 (0.7%) 1.000
Auranofin 0 7 (4.9%) 1.000
Cyclosporine 0 1 (0.7%) 1.000
Minocycline hydrochloride 0 2 (1.4%) 1.000
Cyclophosphamide 0 1 (0.7%) 1.000

Table 3.

Values are given as the number of patients taking a drug; patients can take more than one drug and can switch to another biologic agentHR, hazard ratio; 95% CI, 95% confidence intervalNumber of patients using concomitant drugs related to rheumatoid arthritis during the study [comparing hepatitis B virus (HBV) replication-positive patients with HBV replication-negative patients]

Odds Ratio
(95% CI)
p value
Tacrolimus hydrate 11.1 (2.0-50.6) 0.0015
Sulfasalazine 0.3 (0.0-1.7) 0.2604
Abatacept 1.5 (0.1-17.4) 0.7726
immunoglobulin G 1.9 (0.0-160.3) 0.7572

Table 4.

5% CI, 95% confidence intervalLogistic regression model predicting hepatitis B virus replication in rheumatoid arthritis patients

tacrolimus hydrate may inhibit the function of CTL that controls HBV proliferation, and trigger reactivation of HBV-DNA replication [20, 21]. CS has shown to have direct stimulatory effects on HBV replication, in addition to indirect effects mediated via generalized immune system suppression [4].

TNF is a proinflammatory cytokine that plays a key role in host responses to several types of infection and other stimuli [22]. Various observations have strongly implicated TNF in the pathogenesis of RA and ankylosing spondylitis (AS), and increased TNF production propagates rheumatoid synovitis, promotes osteoclast formation, and results in characteristic bone and joint destruction [23]. TNFA significantly affects the current treatment of RA and AS [24] but is associated with adverse reactions such as reactivation of tuberculosis [25]. Studies regarding the safety of TNFA with chronic viral infection are limited. Several theories exist regarding how TNF inhibitors reactivate hepatitis B. Elevated TNF levels are seen in both the serum and hepatocytes of patients with chronic hepatitis B [26], and are secreted by HBV-specific CTL [27]. TNF has biological activity and an amino acid sequence similar to lymphotoxin, which inhibits HBV replication [28]. Infected cells are also reported to be selectively killed by TNF [33]. TNF acts to suppress HBV DNA replication by reducing intracellular HBV transcription [29]. Animal studies have shown that TNF-knockout mice have defects in the proliferative capacity of HBV-specific CTL [30], suggesting that TNF plays a role in clearing or controlling HBV [30, 31]. Moreover, HBV-specific CTL inhibits HBV gene expression by secreting antiviral cytokines, such as interferon γ and TNF, and inducing apoptosis in HBV-infected hepatocytes [32, 33].

With increasing use of biologic agents such as TNFA, anti-IL-6 receptor, anti-CD20 [34], and anti-CD28, reactivation of HBV DNA replication in patients with resolved HBV will likely increase, particularly in endemic areas. Among patients who are scheduled to receive MTX, CS, tacrolimus hydrate, and biologic agents, patients who are HBsAg negative should be further screened for anti-HBc and anti-HBs.

Conflict of interest statement

None

Advertisement

Acknowledgments

We would like to thank the members of the Contract Research Organization for their assistance in collecting patient data.

References

  1. 1. AS Lok Mc Mahon. B. J. Chronic hepatitis. B. update 2009 Hepatology 2009; 50 61 2 .
  2. 2. Di Bisceglie A. M. Hepatitis B. hepatocellular carcinoma. Hepatology 2009 49: S56 60 .
  3. 3. Hoofnagle JH. Reactivation of hepatitis B. Hepatology 2009 49 :S156 65 .
  4. 4. Calabrese L. H. Zein N. N. Vassilopoulos D. Hepatitis B. virus . H. B. V. reactivation with. immunosuppressive therapy. in rheumatic. diseases assessment. preventive strategies. Ann Rheum Dis 2006 65 983 9 .
  5. 5. 20 Zingarelli S. Airò P. Frassi M. Bazzani C. Scarsi M. Puoti M. Prophylaxis and therapy of HBV infection in 20 patients treated with disease modifying antirheumatic drugs or with biological agents for rheumatic diseases. Reumatismo 2008; 60 22 7 .
  6. 6. Ostuni P. Botsios C. Punzi L. Sfriso P. Todesco S. Hepatitis B. reactivation in. a. chronic hepatitis. B. surface antigen. carrier with. rheumatoid arthritis. treated with. infliximab low dose. methotrexate Ann Rheum Dis 2003 62 686 7 .
  7. 7. Cansu D. U. Kalifoglu T. Korkmaz C. Short-term course of chronic hepatitis B and C under treatment with etanercept associated with different disease modifying antirheumatic drugs without antiviral prophylaxis. J Rheumatol 2008 35 421 4 .
  8. 8. Zoulim F. New insight on hepatitis B virus persistence from the study of intrahepatic viral cccDNA. J Hepatol 2005 42 302 8 .
  9. 9. Rehermann B. Ferrari C. Pasquinelli C. Chisari F. V. 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 .
  10. 10. Yuki N. Nagaoka T. Yamashiro M. Mochizuki K. Kaneko A. Yamamoto K. Omura M. et al. Long-term histologic and virologic outcomes of acute self-limited hepatitis B. Hepatology 2003 37 1172 9 .
  11. 11. Yeo W. Johnson P. J. Diagnosis prevention. management of. hepatitis B. virus reactivation. during anticancer. therapy Hepatology 43 209 20 2006
  12. 12. Kitano K. Kobayashi H. Hanamura M. Furuta K. Ueno M. Rokuhara A. et al. Fulminant hepatitis after allogenic bone marrow transplantation caused by reactivation of hepatitis B virus with gene mutations in the core promoter region. Eur J Haematol 77 255 8 2006
  13. 13. Hui CK, Cheung WW, Zhang HY, Au WY, Yueng YH, Leung AY, et al. Kinetics and risk of de novo hepatitis B infection in HBsAg-negative patients undergoing cytotoxic chemotherapy. Gastroenterology 2006 131 59 68 .
  14. 14. Sera T. Hiasa Y. Michitaka K. Konishi I. Matsuura K. Tokumoto Y. et al. Anti-H Anti-HBs-positive liver failure due to hepatitis B virus reactivation induced by rituximab. Intern Med 2006 45 721 4 .
  15. 15. Esteve M. Saro C. González-Huix F. Suarez F. Forné M. Viver J. M. Chronic hepatitis. B. reactivation following. infliximab therapy. in Crohn’s. disease patients. need for. primary prophylaxis. Gut 2004 53 1363 5
  16. 16. Ito S. Nakazono K. Murasawa A. Mita Y. Hata K. Saito N. ea al. Development of fulminant hepatitis B (precore variant mutant type) after the discontinuation of low-dose methotrexate therapy in a rheumatoid arthritis patient. Arthritis Rheum 2001 44 339 42 .
  17. 17. Montiel P. M. Solis J. A. Chirinos J. A. a. Casis B. Sánchez F. Rodríguez S. Hepatitis B. virus reactivation. during therapy. with etanercept. in an. H. Bs Ag-negative anti-H Bs-positive. patient Liver Int 2008 28 718 20 .
  18. 18. Madonia S. Orlando A. Scimeca D. Olivo M. Rossi F. Cottone M. Occult hepatitis. B. infliximab-induced H. B. V. reactivation Inflamm Bowel Dis 2007 13 508 9 .
  19. 19. Sasaki S. Sato S. Kano Y. Akaike A. Omura T. Sato T. et al. Validity of. C. O. B. A. S. Taq Man. H. B. V. Test 2 20 Igaku to Yakugaku 2009; 61: 787-795 (in Japanese)
  20. 20. Kuwano K. Arai S. The inhibitory effect 506 FK506 on cytotoxic T-lymphocyte killing. Immunol Lett. 1994; 43 153 7 .
  21. 21. Strauss G. Osen W. Debatin K. M. Induction of apoptosis and modulation of activation and effector function in T cells by immunosuppressive drugs. Clin Exp Immunol. 2002 128 255 66 .
  22. 22. Bradley J. R. T. N. F-mediated inflammatory. disease J. Pathol 2008 214 149 60
  23. 23. Harris ED Jr. Rheumatoid arthritis. Pathophysiology and implications for therapy. N Engl J Med 1990 322 1277 89 .
  24. 24. Gibbons LJ, Hyrich KL. Biologic therapy for rheumatoid arthritis: clinical efficacy and predictors of response. BioDrug 2009 23 111 24 .
  25. 25. Winthrop KL. Risk and prevention of tuberculosis and other serious opportunistic infections associated with the inhibition of tumor necrosis factor. Nat Clin Pract Rheumatol 2006; 2: 602-10.
  26. 26. Daniels H. M. Meager A. Eddleston A. L. Alexander G. J. Williams R. Spontaneous production of tumour necrosis factor alpha and interleukin-1 beta during interferon-alpha treatment of chronic HBV infection. Lancet 1990 335 875 7 .
  27. 27. Fang J. W. Shen W. W. Meager A. Lau J. Y. Activation of the tumor necrosis factor-alpha system in the liver in chronic hepatitis B virus infection. Am J Gastroenterol 1996 91 748 53 .
  28. 28. Stoop JN, Woltman AM, Biesta PJ, Kusters JG, Kuipers EJ, Janssen HL, van der Molen RG. Tumor necrosis factor alpha inhibits the suppressive effect of regulatory T cells on the hepatitis B virus-specific immune response. Hepatology 2007 46 699 705 .
  29. 29. Wong GH, Goeddel DV. Tumour necrosis factors alpha and beta inhibit virus replication and synergize with interferons. Nature 1986 323 819 22 .
  30. 30. Kasahara S. Ando K. Saito K. Sekikawa K. Ito H. Ishikawa T. Ohnishi H. et al. Lack of tumor necrosis factor alpha induces impaired proliferation of hepatitis B virus-specific cytotoxic T lymphocytes. J Virol 200377 77 2469 76 .
  31. 31. Ganem D. Prince A. M. Hepatitis B. virus infection--natural. history clinical consequences. N Engl J Med 2004 350 1118 29 .
  32. 32. Guidotti L. G. Ishikawa T. Hobbs M. V. Matzke B. Schreiber R. Chisari F. V. Intracellular inactivation of the hepatitis B virus by cytotoxic T lymphocytes. Immunity 19964 4 25 36 .
  33. 33. Marinos G. Naoumov N. V. Rossol S. Torre F. Wong P. Y. Gallati H. Portmann B. et al. Tumor necrosis factor receptors in patients with chronic hepatitis B virus infection. Gastroenterology 1995 108 1453 63 .
  34. 34. Yeo W. Chan T. C. Leung N. W. Lam W. Y. Mo F. K. Chu M. T. Chan H. L. et al.. Hepatitis B. virus reactivation. in lymphoma. patients with. prior resolved. hepatitis B. undergoing anticancer. therapy with. or without. rituximab J Clin Oncol 200927 27 605 11 .

Written By

Yukitomo Urata, Ryoko Uesato and Dai Tanaka

Submitted: 11 November 2010 Published: 01 August 2011