Journal List > Infect Chemother > v.50(4) > 1109959

Lee, Chun, Song, Choe, Bang, Kim, Kim, Park, Kim, Park, and Oh: Optimal Timing of Zoster Vaccination After Shingles: A Prospective Study of the Immunogenicity and Safety of Live Zoster Vaccine

Abstract

Background

Zoster vaccination is recommended for people with a history of herpes zoster (HZ), but the most effective timing of vaccine administration after zoster illness is unresolved. This prospective observational study compared the immunogenicity and safety of administering HZ vaccine at 6-12 months and 1-5 years after zoster illness.

Materials and Methods

Blood samples were collected before the administration of live zoster vaccine and 6 weeks after vaccination. Varicella-zoster virus (VZV) IgG concentrations and T-cell responses were assessed by glycoprotein enzyme-linked immunosorbent assay and interferon-γ enzyme-linked immunospot assay (ELISPOT), respectively.

Results

The baseline geometric mean value (GMV) of VZV IgG was higher in the 6-12 months group than in the 1-5 years group (245.5 IU/mL vs. 125.9 IU/mL; P = 0.021). However, the GMV increased significantly in both groups (P = 0.002 in the 6-12 months group; P <0.001 in the 1-5 years group). The results of the ELISPOT assay were not significant for differences of the GMV between baseline and 6-week post-vaccination groups, while the GMV increased significantly in both groups (P = 0.001 in the 6-12 months group; P <0.001 in the 1-5 years group).

Conclusion

The immunogenicity of zoster vaccine may be similar whether administered 6-12 months, or >1 year after zoster illness.

Trial Registration

ClinicalTrials.gov Identifier: NCT02704572

Introduction

Herpes zoster (HZ) is a common disease that affects 25 to 35% of the population [12]. Typically, clinical manifestation presents as a vesicular rash, generally limited to a single dermatome and accompanied by aching pain or burning sensation. The most common HZ complication is postherpetic neuralgia, which is a chronic neuropathic pain persisting >120 days after the onset of rash [3]. Major risk factors are increasing age and unvaccinated persons aged >85 years old who have an approximately 50% risk of developing HZ disease [4]. Moreover, recurrent HZ occurs in approximately 6.2% of persons with a history of the disease [5]. Zoster vaccine reduces the development of HZ and postherpetic neuralgia and is recommended for all immunocompetent adults aged ≥60 years [6]. Zoster vaccine has an immune boosting effect in persons with a history of HZ disease [7] and reduces the rate of recurrence in immunocompetent individuals [8]. As a result, current guidelines recommend the vaccination of persons with a history of zoster [69101112].
However, the timing of zoster vaccination following a HZ episode is unclear. The United States Centers for Disease Control and Prevention Advisory Committee on Immunization Practices does not specify vaccine administration timing for those with a history of zoster [6], while both the Australian Immunization Handbook and Canada's National Advisory Committee on Immunization suggest vaccinating at least 1-year after a HZ episode [910]. Public Health England also recommends that immunocompetent individuals who develop zoster illness should delay zoster vaccination for 1 year [11]. However, these recommendations assume that vaccine efficacy could be reduced because of varicella-zoster virus (VZV) boosted immunity. Therefore, it is necessary to scientifically determine the most effective time for zoster vaccination.
The objective of this study was to compare immune response and safety based on the timing of vaccine administration after HZ illness. Therefore, we conducted a prospective study to assess cellular and humoral immune responses and collected adverse event information related to immunocompetent subjects.

Materials and Methods

1. Subjects and vaccination

This study assessed both the immune response to HZ vaccination at 6 weeks and vaccination safety in relation to administration timing following HZ illness. The study was conducted from January 2016 to December 2016. The study enrolled adults who provided written informed consent and who were aged ≥50 years with a physician confirmed diagnosis of HZ illness within the previous 0.5-5 years. Potential subjects who met any of the following criteria were excluded: 1) known contraindication to live zoster vaccination, 2) previous recipient of zoster vaccine, 3) history of taking immunosuppressive drugs such as cytotoxic chemotherapy or corticosteroids, 4) a CD4 cell count <500/mm3 in the setting of HIV infection, 5) organ transplantation, 6) autoimmune disease, or 7) other conditions that cause an abnormal immune response to vaccination such as fever on the day of vaccination or having received other vaccinations within the prior month.
To analyze immune response and vaccine administration safety, study subjects were divided into one of two groups based on whether they received vaccine at 6-12 months or 1-5 years after zoster illness. Subjects who had a HZ episode 12 months earlier were included in the 6-12 months group. In addition, subgroup analysis was performed that divided the period of 1-5 years into 1-3 years (13-36 months) and 3-5 years (>36 months).
Zostavax® (Merck Sharp & Dohme Corp, Rahway, NJ, USA) is a live, attenuated VZV vaccine that was administered as a one-time subcutaneous injection of 0.65 mL containing 19,400 plaque-forming units of the Oka/Merck strain. Blood samples were collected before and 6 weeks after vaccination; peripheral blood mononuclear cells (PBMCs) and serum were immediately separated and stored in liquid nitrogen and –70°C, respectively.
This study was approved by the institutional review board of Seoul National University Hospital (IRB No. 1511-046-718) and registered at ClinicalTrials.gov (number: NCT02704572).

2. Immunologic assessments

Titers of IgG antibodies against VZV glycoprotein were measured using SERION ELISA classic VZV IgG (gpELISA) (Institute Virion\Serion, Würzburg, Germany) according to the manufacturer's instructions. Briefly, serum samples were diluted (1:1,000) and added in duplicate into a 96-well plate that was coated with VZV-specific antigens and then incubated at 37°C for 1 h. After washing, a solution of anti-human IgG antibody conjugated to alkaline phosphatase was added and incubated at 37°C for 30 min. Following this, it was incubated with a para-nitrophenyl phosphate solution at 37°C for 30 min. Optical density was measured at 405 nm using a VersaMax microplate spectrophotometer (Molecular Devices Corporation, San Jose, CA, USA) and antibody titer was quantified using a lot-specific standard curve based on a 4-parameter logistic function.
The VZV-specific interferon gamma (IFN-γ) enzyme linked immunosorbent spot (ELISPOT) assay was performed using an IFN-γ ELISPOT set (BD Bioscience, San Jose, CA, USA) as previously described [12]. Briefly, the 96-well plates were coated with anti-human IFN-γ monoclonal antibody (BD Bioscience) and then incubated overnight. The wells were blocked for 2 h with culture medium containing 10% fetal bovine serum and complete medium containing an ultraviolet-inactivated preparation of VZV antigen (VR-916, American Type Culture Collection, Manassas, VA, USA).
A 100-μL suspension of PBMCs of 107 PBMCs/mL was added to each well, and plates were incubated for 16 to 20 h at 37°C and 5% CO2 in 95% humidity. Next, 100 μL of biotinylated anti-human IFN-γ antibody were added to each well. Plates were incubated for 2 h at 4°C, washed, and developed with streptavidin-horseradish peroxidase and substrate (AEC Substrate Reagent Set, BD Bioscience). The resulting spots were enumerated by an automated microscope (CRL ImmunoSpot S4Core Analyzer, Cellular Technology ltd. Cleveland, OH, USA). The VZV-specific responses were reported as the number of spot-forming cells (SFCs) per 106 PBMCs based on an SFC count in response to VZV antigen minus the SFC count in response to MRC-5 cell antigen. Samples with a poor response to phytohemagglutinin (<300 SFCs/106 PBMCs) were excluded from the analysis.

3. Safety assessment

We evaluated vaccine safety with a self-reported structured questionnaire and medical interview 6 weeks after vaccination to identify the type and severity of any local and systemic adverse events. Adverse events were graded using United States Food and Drug Administration standard toxicity scale [13]. The relationship between adverse events and vaccination was divided into three categories: unlikely, possible, or likely. A vaccine-related adverse event was defined as having a possible or likely relationship.

4. Statistical analysis

The VZV-specific IgG levels and SFCs of the ELISPOT assay were presented as the geographic mean value (GMV) and geometric mean ratio of values at baseline and 6 weeks after vaccination (the geographic mean fold rise [GMFR]). GMV and GMFR were compared using the Wilcoxon signed-rank test. The Mann-Whitney U test was used to compare other continuous variables and the Chi-square test or Fisher's exact test was used for categorical variables, when appropriate. All tests were two-sided, and P <0.05 was considered significant. All statistical analyses were performed using IBM SPSS Statistics for Windows, version 24.0 (IBM Corp., Armonk, NY, USA).

Results

1. Subjects

A total of 60 subjects were enrolled in the study (Supplementary Fig. 1). One person in the 1-5 years group was excluded due to loss to follow-up and the data for 59 subjects were used in the final analysis. The median age was 63 years (range, 50-84) with 42 subjects (71.2%) <70 years old and 17 (28.8%) ≥70 years old (Table 1). Thirty-nine subjects (66.1%) were female. Participants in each group were not significantly different with respect to age, sex, and underlying disease other than hypertension (P = 0.018) and diabetes mellitus (P = 0.025) which were more frequent in the 6-12 months group (Table 1, Supplementary Table 1). Following zoster illness, 18 and 41 subjects were vaccinated from the 6-12 months and 1-5 years groups, respectively. A 66-year-old male subject with hypertension in the 6-12 months group had disseminated HZ from the buttocks to trunk and neck. Before the study, one subject from each group had a history of two HZ episodes. One subject, from the 6-12 month group, was a 71-year-old female with hypertension whose HZ first developed 10 years ago then relapsed on her left chest 11 months before study enrollment. The subject in the 1-5 years group was a 61-year-old female without underlying medical condition, whose HZ first developed 30 years ago then relapsed on her left flank 4 years before the study.
Table 1

Clinical characteristics of subjects at baseline

ic-50-311-i001
Variable All subjects 6–12 months after zoster (n = 18) 1–5 years after zoster (n = 41) P-value
Age
Median years (range) 63 (50–84) 68 (50–84) 62 (50–79) 0.564
Female sex 39 (66.1) 9 (50.0) 30 (73.2) 0.083
Median months after zoster (range) 25 (7–51) 9 (7–12) 40 (13–51) <0.001
Disseminated zoster 1 (1.7) 1a (5.6) 0 0.305
Underlying disease
Hypertension 9 (15.3) 6 (33.3) 3 (7.3) 0.018
Diabetes mellitus 3 (5.1) 3 (16.7) 0 0.025
Malignancy 1 (1.7) 0 1 (1.4) >0.999
Hypothyroidism 1 (1.7) 1 (5.6) 0 0.305
aHerpes zoster rash started from the left buttock and progressed to the trunk and neck.
The value denotes the subject number (%) unless otherwise indicated.

2. Immunologic assessments

The baseline GMV of VZV-specific IgG was 154.88 IU/mL (95% confidence interval [CI], 123.03-194.98) and was significantly higher in the 6-12 months group than the 1-5 years group (245.47 IU/mL vs. 125.89 IU/mL, P = 0.021) (Table 2). Six weeks after vaccination, the GMVs increased significantly in both groups (P = 0.002 in the 6-12 months group, P <0.001 in the 1-5 years group). The GMVs at 6 weeks were not significantly different between groups (346.74 IU/mL vs. 309.03 IU/mL, P = 0.569). Meanwhile, the GMFR was significantly lower in the 6-12 months group than in the 1-5 years group (1.41 vs. 2.45, P = 0.002). The percent of subjects with a GMFR >4 was lower in the 6-12 months group than in the 1-5 years group but was not statistically significant (5.6% vs. 24.4%, P = 0.146).
Table 2

Varicella-zoster virus specific immune response

ic-50-311-i002
Variable Vaccination timing after zoster illness P-value
6–12 months 1–5 years
No. Value No. Value
ELISA for IgG (IU/mL)
Baseline GMV 18 245.47 (144.54–416.87) 41 125.89 (100.00–158.49) 0.021
Post-vaccination GMV 18 346.74 (229.09–537.03) 41 309.03 (251.19–371.54) 0.569
GMFR 18 1.41 (1.17–1.74) 41 2.45 (1.95–3.09) 0.002
GMFR >4 18 1 (5.6) 41 10 (24.4) 0.146
IFN-γ ELISPOT (SFC)
Baseline GMV 16 7.76 (3.72–16.60) 35 9.12 (6.17–13.80) 0.673
Post-vaccination GMV 16 26.30 (15.14–45.71) 35 38.02 (28.84–51.29) 0.184
GMFR 16 3.39 (2.04–5.62) 35 4.17 (2.88–6.03) 0.507
GMFR >4 16 7 (43.8) 35 15 (42.9) 0.952
ELISA, enzyme-linked immunosorbent assay; GMV, geometric mean value; GMFR, geometric mean fold-rise; INF-γ ELISPOT, interferon-γ enzyme-linked immunosorbent spot; SFC, spot forming cell per 106 peripheral blood mononuclear cells.
Data represent the geometric mean (95% confidence interval) or No. (%).
The IFN-γ ELISPOT assay baseline GMV was 8.71 SFCs per 106 PBMCs (95% CI, 6.17-12.59) for the total study population. Six weeks after vaccination, the GMV significantly increased to 33.88 SFCs per 106 PBMC cells (95% CI, 26.30-43.65, P <0.001). There was no significant difference in the GMV at baseline and 6 weeks after vaccination between groups and the GMVs increased significantly in both groups (P = 0.001 in the 6-12 months group, P <0.001 in the 1-5 years group) (Table 2). There were no significant differences between groups in the GMFR (3.39 vs. 4.17, P = 0.507), and the number of subjects with a GMFR >4 (43.8% vs. 42.9%, P = 0.952). Due to the lack of observed cells, eight samples could not be analyzed, resulting in the discrepancy of sample numbers when using ELISA and ELISPOT.
Subgroups were analyzed by dividing subjects into groups based on zoster vaccine administration time following zoster illness (6-12 months, 1-3 years, and >3 years), and this review showed that boosting effect in humoral and cellular immunity was comparable between the groups (Fig. 1).
Figure 1

Varicella-zoster virus (VZV) specific immune responses according to zoster vaccine administration timing after zoster illness. The study population was divided into a before 1-year group (6-12 months), 1-3 years group (13-36 months), and >3 years group (>36 months). (A) Bars indicate geometric means and 95% confidence intervals (CI) for titers of enzyme-linked immunosorbent assay against VZV glycoproteins (gpELISA). (B) Bars indicate geometric means and 95% CIs for interferon-γ enzyme-linked immunospot (ELISPOT) counts, measured as spot-forming cells per 106 peripheral blood mononuclear cells.

aP <0.01.
ic-50-311-g001

3. Safety assessment

There were three cases of vaccine related adverse events during the study period: one person from each group had mild pain and tenderness at the injection site, and one subject from the 6-12 months group had mild nausea and vomiting. There were no cases of a zoster-like rash during the study period.

Discussion

This prospective study showed that 6 weeks after the administration of live zoster vaccine to subjects with a 6-12 month or 1-5 year history of having had zoster illness experienced a significant increase of anti-VZV IgG levels measured by gpELISA and IFN-γ ELISPOT. In addition, the anti-VZV IgG levels and IFN-γ ELISPOT counts at 6 weeks were comparable between the groups, although baseline IgG levels were higher in the 6-12 months group than in the 1-5 years group.
Reactivation of VZV is known to be related to declining VZV-specific cell-mediated immunity [1415]. VZV-specific cellular immunity was reported to be induced similarly by zoster illness and zoster vaccination [16]. In the present study, cellular immunity was measured by the interferon-γ ELISPOT, which was significantly increased by zoster vaccination and was comparable 6 weeks after vaccination in both groups. These findings suggest that both groups had a similar likelihood of developing HZ after zoster vaccination.
Humoral immunity is less emphasized since the level of VZV-specific IgG is maintained with age [15], but some studies have reported the role of VZV-specific IgG in zoster illness. Gilbert et al. speculated that the increased fold change in gpELISA titers 6 weeks after vaccination correlates with zoster vaccine efficacy [17], and Levin et al. suggested that VZV-specific IgG correlates with cell-mediated immunity [18]. In the present study, the GMFR of IgG in gpELISA was higher in the >1-year group, although this result seems to be influenced by the higher baseline IgG titers in the 6-12 months group. However, as there was no significant difference in the absolute IgG titer after vaccination and the number of patients with a GMFR increase of >4 (43.8% in the 6-12 months group vs. 42.9% in the 1-5 years group), the difference in vaccine efficacy between the two groups seems not to be definite.
Analysis of both cellular and humoral responses to zoster vaccination reveals that vaccination administered ≥1 year after zoster illness may have similar immunogenicity responses. Our sub-analyses of groups vaccinated 1-3 years and 3-5 years after HZ also suggest that a vaccination delay of >3-years after zoster illness does not provide a cellular and humoral immune response advantage.
Previous prospective and retrospective observational studies suggest that patients who had a zoster illness within the previous year had a low rate of recurrence (0.4-0.8%) [1920]. These findings are compatible with our results showing that the anti-VZV IgG level was maintained until 1 year after zoster illness. Similarly, Tseng et al. reported that the recurrence rate of HZ was 0.1% during a 2-year follow-up of immunocompetent persons regardless of vaccination history [8]. However, the recurrence rates of HZ increased to 5.3% after a 20-year follow-up in long-term cohort studies [21]. Based on these epidemiological findings and data from the present study, the low recurrence rate in subjects with a recent HZ history might be associated with an immunologic boosting effect caused by the original zoster illness.
This study has several limitations. First, only the VZV-specific IFN-γ ELISPOT was used to evaluate VZV-associated cell-mediated immunity. Other methods, such as an intracellular cytokine assay, may show more details about the CD4/CD8 cell response and effector/memory cells. Second, confounding factors such as age, gender, and underlying disease might affect the results of this study because the number of subjects was small. However, a sub-analysis dividing study groups into those aged <70, and those ≥70 years of age, showed similar results (data not shown). Third, a reported diagnosis or history of HZ could be erroneous. However, one of the reasons for recommending zoster vaccination after HZ is that the previous illness may have been misdiagnosed. In addition, diagnosis of HZ is usually not made by laboratory tests in actual practice. Therefore, it may be reasonable to apply the results of this study to clinical practice. Fourth, long-term clinical data about HZ recurrence were not available. A large-scale study is needed to confirm the results of this study.
In conclusion, the immunogenicity of zoster vaccination administered 6-12 months after zoster illness may be similar to that administered >1 year after zoster illness.

Notes

Presentation The results of this study were selected for a poster presentation at the ID Week 2017 in San Diego, CA, October 4-8, 2017.

Funding This work was supported by a grant (No. 03-2015-0360) from the Seoul National University Hospital (SNUH) Research Fund, Seoul, Korea.

Conflict of Interest No conflicts of interest.

Author Contributions

  • Conceptualization: WBP, NJK, MO.

  • Data curation: JYC, WBP, KHS, PGC, JHB, ESK.

  • Formal analysis: EL, WBP, HBK, SWP, NJK, MO.

  • Writing - original draft: EL, JYC, WBP.

  • Writing - review & editing: EL, JYC, WBP, PGC, KHS, JHB, ESK, HBK, SWP, NJK, MO.

References

1. Bowsher D. The lifetime occurrence of herpes zoster and prevalence of post-herpetic neuralgia: a retrospective survey in an elderly population. Eur J Pain. 1999; 3:335–342.
crossref
2. van Hoek AJ, Gay N, Melegaro A, Opstelten W, Edmunds WJ. Estimating the cost-effectiveness of vaccination against herpes zoster in England and Wales. Vaccine. 2009; 27:1454–1467.
crossref
3. Schmader K, Gnann JW Jr, Watson CP. The epidemiological, clinical, and pathological rationale for the herpes zoster vaccine. J Infect Dis. 2008; 197:Suppl 2. S207–S215.
crossref
4. Cohen JI. Clinical practice: Herpes zoster. N Engl J Med. 2013; 369:255–263.
5. Yawn BP, Wollan PC, Kurland MJ, St Sauver JL, Saddier P. Herpes zoster recurrences more frequent than previously reported. Mayo Clin Proc. 2011; 86:88–93.
crossref
6. Dooling KL, Guo A, Patel M, Lee GM, Moore K, Belongia EA, Harpaz R. Recommendations of the advisory committee on immunization practices for use of herpes zoster vaccines. MMWR Morb Mortal Wkly Rep. 2018; 67:103–108.
crossref
7. Mills R, Tyring SK, Levin MJ, Parrino J, Li X, Coll KE, Stek JE, Schlienger K, Chan IS, Silber JL. Safety, tolerability, and immunogenicity of zoster vaccine in subjects with a history of herpes zoster. Vaccine. 2010; 28:4204–4209.
crossref
8. Tseng HF, Chi M, Smith N, Marcy SM, Sy LS, Jacobsen SJ. Herpes zoster vaccine and the incidence of recurrent herpes zoster in an immunocompetent elderly population. J Infect Dis. 2012; 206:190–196.
crossref
9. National Centre for Immunisation Research and Surveillance. The Australian immunisation handbook. 10th ed. Accessed 6 December, 2018. Available at: http://www.practiceassist.com.au/PracticeAssist/media/ResourceLibrary/Programs%20and%20National%20Schemes/Immunise-The-Australian-Immunisation-Handbook-10th-Edition.pdf.
10. National Advisory Committee on Immunization. Update on the Use of Herpes Zoster Vaccine. Accessed December 6, 2018. Available at: https://www.canada.ca/en/public-health/services/publications/healthy-living/update-use-herpes-zoster-vaccine.html?_ga=2.252998144.881335399.1517230152-1438438172.1517230152.
11. Public Health England. Vaccination against shingles: 2015/16, Information for healthcare professionals. Accessed 6 December, 2018. Available at: https://www.gov.uk/government/uploads/system/uploads/attachment_data/file/503099/PHE_Shingles_advice_for_health_professionals_2015-16__February2016_V4.pdf.
12. Kim JW, Min CK, Mun YC, Park Y, Kim BS, Nam SH, Koh Y, Kwon JH, Choe PG, Park WB, Kim I. Varicella-zoster virus-specific cell-mediated immunity and herpes zoster development in multiple myeloma patients receiving bortezomib- or thalidomide-based chemotherapy. J Clin Virol. 2015; 73:64–69.
crossref
13. Food and Drug Administration (FDA). Toxicity grading scale for healthy adult and adolescent volunteers enrolled in preventive vaccine clinical trials. Rockville, MD: Botswana Government Print;2007.
14. Oxman MN. Immunization to reduce the frequency and severity of herpes zoster and its complications. Neurology. 1995; 45:12 Suppl 8. S41–S46.
crossref
15. Levin MJ, Oxman MN, Zhang JH, Johnson GR, Stanley H, Hayward AR, Caulfield MJ, Irwin MR, Smith JG, Clair J, Chan IS, Williams H, Harbecke R, Marchese R, Straus SE, Gershon A, Weinberg A. Veterans Affairs Cooperative Studies Program Shingles Prevention Study Investigators. Varicella-zoster virus-specific immune responses in elderly recipients of a herpes zoster vaccine. J Infect Dis. 2008; 197:825–835.
crossref
16. Weinberg A, Zhang JH, Oxman MN, Johnson GR, Hayward AR, Caulfield MJ, Irwin MR, Clair J, Smith JG, Stanley H, Marchese RD, Harbecke R, Williams HM, Chan IS, Arbeit RD, Gershon AA, Schödel F, Morrison VA, Kauffman CA, Straus SE, Schmader KE, Davis LE, Levin MJ. US Department of Veterans Affairs (VA) Cooperative Studies Program Shingles Prevention Study Investigators. Varicella-zoster virus-specific immune responses to herpes zoster in elderly participants in a trial of a clinically effective zoster vaccine. J Infect Dis. 2009; 200:1068–1077.
crossref
17. Gilbert PB, Gabriel EE, Miao X, Li X, Su SC, Parrino J, Chan IS. Fold rise in antibody titers by measured by glycoprotein-based enzyme-linked immunosorbent assay is an excellent correlate of protection for a herpes zoster vaccine, demonstrated via the vaccine efficacy curve. J Infect Dis. 2014; 210:1573–1581.
crossref
18. Levin MJ, Schmader KE, Pang L, Williams-Diaz A, Zerbe G, Canniff J, Johnson MJ, Caldas Y, Cho A, Lang N, Su SC, Parrino J, Popmihajlov Z, Weinberg A. Cellular and humoral responses to a second dose of herpes zoster vaccine administered 10 years after the first dose among older adults. J Infect Dis. 2016; 213:14–22.
crossref
19. Helgason S, Sigurdsson JA, Gudmundsson S. The clinical course of herpes zoster: a prospective study in primary care. Eur J Gen Pract. 1996; 2:12–16.
crossref
20. Donahue JG, Choo PW, Manson JE, Platt R. The incidence of herpes zoster. Arch Intern Med. 1995; 155:1605–1609.
crossref
21. Ragozzino MW, Melton LJ 3rd, Kurland LT, Chu CP, Perry HO. Population-based study of herpes zoster and its sequelae. Medicine (Baltimore). 1982; 61:310–316.
crossref

SUPPLEMENTARY MATERIALS

Supplementary Table 1

Underlying diseases of subjects at baseline

Supplementary Figure 1

Flow chart of study design.
TOOLS
ORCID iDs

Eunyoung Lee
https://orcid.org/0000-0001-8280-3605

June Young Chun
https://orcid.org/0000-0001-9345-6645

Kyoung-Ho Song
https://orcid.org/0000-0002-4517-3840

Pyoeng Gyun Choe
https://orcid.org/0000-0001-6794-7918

Ji Hwan Bang
https://orcid.org/0000-0002-7628-1182

Eu Suk Kim
https://orcid.org/0000-0001-7132-0157

Hong Bin Kim
https://orcid.org/0000-0001-6262-372X

Sang Won Park
https://orcid.org/0000-0002-0550-1897

Nam Joong Kim
https://orcid.org/0000-0001-6793-9467

Wan Beom Park
https://orcid.org/0000-0003-0022-9625

Myoung-don Oh
https://orcid.org/0000-0002-2344-7695

Similar articles