Journal List > J Rheum Dis > v.24(4) > 1064333

Kwon, Lee, Yang, Choi, Park, Lee, Song, and Lee: Risk of Herpes Zoster in Patients with Rheumatoid Arthritis Undergoing Biologic Disease-Modifying Therapy

Abstract

Objective

Rheumatoid arthritis (RA) patients suffer from an increased risk of herpes zoster (HZ) partially due to immunosuppressant medications. This study investigated HZ in RA patients treated with biologic disease-modifying antirheumatic drugs (bDMARDs), as compared with conventional DMARDs (cDMARDs).

Methods

This retrospective case series study assembled record information of 277 RA patients who received bDMARDs after failure of at least one cDMARDs at Seoul National University Hospital between August 2003 and February 2015. Following capture of baseline information and identification of HZ episodes, crude HZ incidence rates per 100 patient-years (95% confidence intervals) were calculated.

Results

For 718 treatment courses, 277 (38.6%) comprised cDMARDs, 66 (9.2%) infliximab, 175 (24.4%) etanercept, 95 (13.2%) adalimumab, 9 (1.3%) golimumab, 41 (5.7%) rituximab, 31 (4.3%) abatacept, and 24 (3.3%) tocilizumab. There were 37 HZ episodes, 16 during cDMARD treatment courses, and 21 accompanying bDMARDs, two with infliximab, eight with etanercept, five with adalimumab, and three each with rituximab and abatacept. The crude HZ incidence rate per 100 patient-years was 2.4 (1.4∼3.9) for cDMARDs, 2.2 (0.3∼7.9) for infliximab, 1.8 (0.8∼3.6) for etanercept, 3.7 (1.2∼8.4) for adalimumab, 3.9 (0.8∼11.0) for rituximab, and 8.5 (1.8∼23.1) for abatacept.

Conclusion

We conclude that bDMARDs do not always increase the risk of HZs in RA patients, although HZ rates vary for different bDMARDs.

REFERENCES

1. Geraldino-Pardilla L, Bathon JM. Management of rheumatoid arthritis: synovitis. Hochberg MC, Silman AJ, Smolen JS, Weinblatt ME, Weisman MH, editors. Hochberg Rheumatology. 6th ed.Philadelphia: Elsevier;2015. p. 802–8.
crossref
2. Law SC, Street S, Yu CH, Capini C, Ramnoruth S, Nel HJ, et al. T-cell autoreactivity to citrullinated autoantigenic peptides in rheumatoid arthritis patients carrying HLA-DRB1 shared epitope alleles. Arthritis Res Ther. 2012; 14:R118.
crossref
3. Mitchell DM, Spitz PW, Young DY, Bloch DA, McShane DJ, Fries JF. Survival, prognosis, and causes of death in rheumatoid arthritis. Arthritis Rheum. 1986; 29:706–14.
crossref
4. Wolfe F, Mitchell DM, Sibley JT, Fries JF, Bloch DA, Williams CA, et al. The mortality of rheumatoid arthritis. Arthritis Rheum. 1994; 37:481–94.
crossref
5. Symmons DP, Jones MA, Scott DL, Prior P. Longterm mortality outcome in patients with rheumatoid arthritis: early presenters continue to do well. J Rheumatol. 1998; 25:1072–7.
6. Hayward AR, Herberger M. Lymphocyte responses to varicella zoster virus in the elderly. J Clin Immunol. 1987; 7:174–8.
crossref
7. Cohen JI. Clinical practice: Herpes zoster. N Engl J Med. 2013; 369:255–63.
8. Smitten AL, Choi HK, Hochberg MC, Suissa S, Simon TA, Testa MA, et al. The risk of herpes zoster in patients with rheumatoid arthritis in the United States and the United Kingdom. Arthritis Rheum. 2007; 57:1431–8.
crossref
9. Schmajuk G, Trivedi AN, Solomon DH, Yelin E, Trupin L, Chakravarty EF, et al. Receipt of disease-modifying antirheumatic drugs among patients with rheumatoid arthritis in Medicare managed care plans. JAMA. 2011; 305:480–6.
crossref
10. Wolfe F, Michaud K, Chakravarty EF. Rates and predictors of herpes zoster in patients with rheumatoid arthritis and non-inflammatory musculoskeletal disorders. Rheumatology (Oxford). 2006; 45:1370–5.
crossref
11. McDonald JR, Zeringue AL, Caplan L, Ranganathan P, Xian H, Burroughs TE, et al. Herpes zoster risk factors in a national cohort of veterans with rheumatoid arthritis. Clin Infect Dis. 2009; 48:1364–71.
crossref
12. Veetil BM, Myasoedova E, Matteson EL, Gabriel SE, Green AB, Crowson CS. Incidence and time trends of herpes zoster in rheumatoid arthritis: a population-based cohort study. Arthritis Care Res (Hoboken). 2013; 65:854–61.
crossref
13. Winthrop KL, Baddley JW, Chen L, Liu L, Grijalva CG, Delzell E, et al. Association between the initiation of antitumor necrosis factor therapy and the risk of herpes zoster. JAMA. 2013; 309:887–95.
crossref
14. Sakai R, Komano Y, Tanaka M, Nanki T, Koike R, Nagasawa H, et al. Time-dependent increased risk for serious infection from continuous use of tumor necrosis factor antagonists over three years in patients with rheumatoid arthritis. Arthritis Care Res (Hoboken). 2012; 64:1125–34.
15. Winthrop KL, Yamanaka H, Valdez H, Mortensen E, Chew R, Krishnaswami S, et al. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014; 66:2675–84.
crossref
16. Yun H, Xie F, Delzell E, Chen L, Levitan EB, Lewis JD, et al. Risks of herpes zoster in patients with rheumatoid arthritis according to biologic disease-modifying therapy. Arthritis Care Res (Hoboken). 2015; 67:731–6.
crossref
17. Antonelli MA, Moreland LW, Brick JE. Herpes zoster in patients with rheumatoid arthritis treated with weekly, low-dose methotrexate. Am J Med. 1991; 90:295–8.
crossref
18. Strangfeld A, Listing J, Herzer P, Liebhaber A, Rockwitz K, Richter C, et al. Risk of herpes zoster in patients with rheumatoid arthritis treated with anti-TNF-alpha agents. JAMA. 2009; 301:737–44.
19. Galloway JB, Mercer LK, Moseley A, Dixon WG, Ustianowski AP, Helbert M, et al. Risk of skin and soft tissue infections (including shingles) in patients exposed to anti-tumour necrosis factor therapy: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis. 2013; 72:229–34.
crossref
20. Serac G, Tubach F, Mariette X, Salmon-Céron D, Ravaud P, Lioté F, et al. Risk of herpes zoster in patients receiving an-ti-TNF-α in the prospective French RATIO registry. J Invest Dermatol. 2012; 132:726–9.
crossref
21. Mélet J, Mulleman D, Goupille P, Ribourtout B, Watier H, Thibault G. Rituximab-induced T cell depletion in patients with rheumatoid arthritis: association with clinical response. Arthritis Rheum. 2013; 65:2783–90.
crossref
22. Lavielle M, Mulleman D, Goupille P, Bahuaud C, Sung HC, Watier H, et al. Repeated decrease of CD4+ T-cell counts in patients with rheumatoid arthritis over multiple cycles of rituximab treatment. Arthritis Res Ther. 2016; 18:253.
crossref
23. Schiff M, Weinblatt ME, Valente R, van der Heijde D, Citera G, Elegbe A, et al. Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: two-year efficacy and safety findings from AMPLE trial. Ann Rheum Dis. 2014; 73:86–94.
crossref
24. Ghoreschi K, Jesson MI, Li X, Lee JL, Ghosh S, Alsup JW, et al. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol. 2011; 186:4234–43.
crossref
25. Schiff MH, Kremer JM, Jahreis A, Vernon E, Isaacs JD, van Vollenhoven RF. Integrated safety in tocilizumab clinical trials. Arthritis Res Ther. 2011; 13:R141.
crossref

Table 1.
Baseline characteristics of study patients at the time point of RA diagnosis
Variable Patient (n=277)
Age at RA diagnosis (yr) 46.8 (13.6)
Sex (female), n (%) 596 (83.0)
Underlying disease, n (%)  
Diabetes milletus 75 (10.5)
Hypertension 198 (27.7)
Malignancy 32 (4.5)
Laboratory findings  
RF, titer (IU/mL) (n=273) 80.4 (99.2)
Anti-CCP antibody, titer (IU/mL) (n=289) 124.2 (166.8)
ESR (mm/h) (n=253) 49.4 (34.7)
CRP (mg/dL) (n=271) 2.4 (4.1)
Conventional DMARD after RA diagnosis, n (%)  
Methotrexate 201 (75.0)
Leflunomide 46 (17.2)
Hydroxychloroquine 126 (47.0)
Sulfasalazine 88 (32.8)
Corticosteroids 215 (80.2)
Prednisolone, dose* (mg/d) 5.4 (4.5)

Values are presented as mean (standard deviation) or number (%). RA: rheumatoid arthritis, RF: rheumatoid factor, CCP: cyclic citrullinated peptide, ESR: erythrocyte sedimentation rate, CRP: C-reactive protein, DMARD: disease-modifying antirheumatic drug.

* Corticosteroids calculated by prednisolone equivalent dose.

Table 2.
Comparison of characteristics and demographics at the time point of starting between cDMARD and bDMARD treatment period
Variable cDMARDs period (n = 277) bDMARDs (post-cDMARDs period)
Total bDMARDs (n = 441) Anti-TNF α antibodies (n = 170) Infliximab (n = 66) Adalimumab (n = 95) Golimumab (n = 9) Etanercept (n= 175) Rituximab (n=41) Abatacept (n = 31) Tocilizumab (n = 24)
Age (yr) 49.7 (12.7) 52.9 (13.2) 53.4 (13.0) 56.2 (12.4) 51.4 (13.3) 53.7 (10.7) 51.8 (12.8) 55.1 (12.4) 51.8 (14.3) 54.6 (18.0)
Sex (female), n (%) 229 (82.7) 367 (83.2) 145 (85.3) 57 (86.4) 80 (84.2) 8 (88.9) 144 (82.3) 34 (82.9) 26 (83.9) 18 (75.0)
Treatment duration (yr) 2.4 (2.3) 1.8 (1.9) 1.4 (1.5) 1.4 (1.4) 2.5 (2.3) 1.4 (1.6) 1.2 (1.4) 1.9 (1.6) 1.1 (1.2) 0.8 (0.5)
Laboratory findings                    
ESR (mm/h) 49.4 (34.7) 56.5 (28.7) 54.1 (26.8) 55.2 (28.4) 54.5 (26.5) 41.5 (11.3) 57.8 (28.7) 71.5 (32.3) 43.5 (23.7) 54.4 (30.0)
CRP (mg/dL) 2.4 (4.1) 2.6 (2.8) 2.2 (2.5) 2.4 (2.6) 2.2 (2.4) 0.6 (0.86) 2.9 (3.1) 3.9 (3.6) 1.8 (1.9) 2.0(1.8)
Concomitant cDMARD, n (%)                    
Methotrexate 201 (75.0) 289 (65.5) 123 (72.4) 50 (75.8) 68 (71.6) 5 (55.6) 113 (64.6) 25 (61.0) 15 (48.4) 12 (50.0)
Leflunomide 46 (17.2) 28 (6.4) 14 (8.2) 8 (12.1) 6 (6.3) 0 (0) 10 (5.7) 3 (7.3) 1 (3.2) 0 (0)
Hydroxychloroquine 126 (47.0) 41 (9.3) 12 (7.1) 7 (10.6) 5 (5.3) 0 (0) 24 (13.7) 3 (7.3) 2 (6.5) 0 (0)
Sulfasalazine 88 (32.8) 40 (9.1) 9 (5.3) 4 (6.1) 5 (5.3) 0 (0) 22 (12.6) 8 (19.5) 1 (3.2) 0 (0)
Prednisolone 215 (80.2) 378 (85.7) 147 (86.5) 63 (95.5) 80 (84.2) 4 (44.4) 149 (85.1) 37 (90.2) 27 (87.1) 18 (75.0)
Prednisolone, dose (mg/d) 5.4 (4.5) 5.3 (3.3) 5.2 (3.1) 6.1 (2.8) 4.7 (3.0) 2.1 (3.4) 5.0 (3.0) 6.7 (4.4) 5.7 (4.2) 4.7 (3.5)

Values are presented as mean (standard deviation) or number (%). n: the number of treatment episodes receiving cDMARD and/or bDMARD therapies. cDMARD: conventional disease-modifying antirheumatic drug, bDMARD: biologic disease-modifying antirheumatic drug, TNF: tumor necrosis factor, ESR: erythrocyte sedimentation rate, CRP: C-reactive protein.

* Anti-TNFa antibodies including infliximab, adalimumab, and golimumab.

Oral corticosteroid calculated by predinisolone equivalent dose.

Table 3.
Crude incidence rates of herpes zoster events per 100 patient-years
Treatment Observed patient-years HZ cases Crude incidence rates (95% CI)
cDMARDs (n=277) 662.1 16 2.4 (1.4∼3.9)
bDMARDs (n=441) 808.1 21 2.6 (1.6∼4.0)
Anti-TNFα antibodies* (n=170) 236.9 7 2.9 (2.2∼6.0)
Infliximab (n=66) 89.8 2 2.2 (0.3∼7.9)
Adalimumab (n=95) 135.9 5 3.7 (1.2∼8.4)
Golimumab (n=9) 11.2 0 0.0 (0∼28.5)
Etanercept (n=175) 440.3 8 1.8 (0.8∼3.6)
Rituximab (n=41) 77.1 3 3.9 (0.8∼11.0)
Abatacept (n=31) 35.5 3 8.5 (1.8∼23.1)
Tocilizumab (n=24) 18.3 0 0.0 (0∼18.5)

n: the number of treatment episodes receiving cDMARDs and/or bDMARDs therapies. HZ: herpes zoster, CI: confidence interval, cDMARDs: conventional disease-modifying antirheumatic drugs, bDMARD: biologic disease-modifying antirheumatic drug, TNF: tumor necrosis factor.

* Anti-TNFα antibodies including infliximab, adalimumab, and golimumab.

Table 4.
Clinical characteristics of herpes zoster cases in bDMARD users
Patient no. Age (yr) Sex Disease duration at HZ occurrence (yr) Biologics MTX (mg/wk) PD*(mg/d) N'th biologics at HZ occurrence Total number of biologics use
1 39 F 7.2 ETA 10 1.25 1 2
2 78 F 9.0 ADA 12.5 0 2 4
3 54 F 1.4 RTX 0 5 2 2
4 45 F 4.4 ADA 15 0 1 3
5 52 F 7.3 ABT 15 0 2 3
6 57 F 9.1 IFX 15 2.5 1 1
7 50 F 18.3 ETA 17.5 2.5 1 2
8 56 F 7.8 RTX 0 5 2 2
9 37 F 14.8 ETA 15 5 1 1
10 41 F 12.4 ETA 0 0 1 1
11 42 F 12.7 ETA 0 0 1 1
12 51 F 1.1 ADA 15 5 1 2
13 59 F 2.2 ETA 0 5 1 1
14 66 M 2.1 ETA 10 2.5 1 1
15 74 F 12.1 IFX 15 5 2 2
16 75 F 18.0 ADA 12.5 5 1 1
17 42 F 8.3 ETA 0 2.5 2 2
18 56 F 13.3 ABT 0 0 2 2
19 65 F 22.2 RTX 12.5 5 3 3
20 73 F 3.1 ABT 0 5 2 2
21 51 F 2.1 ADA 15 1.5 1 1

bDMARD: biologic disease-modifying antirheumatic drug, HZ: herpes zoster, MTX: methotrexate, PD: prednisolone, M: male, F: female, ETA: etanercept, ADA: adalimumab, RTX: rituximab, ABT: abatacept, IFX: infliximab.

* Oral corticosteroid calculated by predinisolone equivalent dose.

TOOLS
Similar articles