You are on page 1of 7

Vaccine 33 (2015) 789–795

Contents lists available at ScienceDirect

Vaccine
journal homepage: www.elsevier.com/locate/vaccine

Comparison of intramuscular and subcutaneous administration of a


herpes zoster live-attenuated vaccine in adults aged ≥50 years:
A randomised non-inferiority clinical trial
Javier Diez-Domingo a,b , Thomas Weinke c , Juan Garcia de Lomas d , Claudius U. Meyer e ,
Isabelle Bertrand f , Cécile Eymin f , Stéphane Thomas f , Christine Sadorge f,∗
a
FISABIO-Public Health, Avda Cataluna 21, 46020 Valencia, Spain
b
Universidad Católica de Valencia, ‘San Vicente Martir’, Valencia, Spain
c
Klinikum Ernst von Bergmann, Charlottenstr. 72, 14467 Potsdam, Germany
d
Department of Microbiology, University of Valencia, School of Medicine, Avda Blasco Ibañez 17, 46010 Valencia, Spain
e
Pediatric Immunlogy, University Medical Center of the Johannes Gutenberg University, Ober Zahlbacher Str. 63, 55128 Mainz, Germany
f
Sanofi Pasteur MSD, 162 avenue Jean Jaurès, CS 50712, 69367 Lyon Cedex 07, France

a r t i c l e i n f o a b s t r a c t

Article history: Zostavax® is a live, attenuated varicella zoster virus (VZV) vaccine developed specifically for the preven-
Received 24 September 2014 tion of HZ and PHN in individuals aged ≥50 years. During the clinical development of Zostavax, which was
Received in revised form mainly in the US, the vaccine was administrated by the subcutaneous (SC) route. In Europe, many health-
10 December 2014
care professionals prefer administering vaccines by the intramuscular (IM) route. This was an open-label,
Accepted 11 December 2014
randomised trial conducted in 354 subjects aged ≥50 years. The primary objectives were to demonstrate
Available online 30 December 2014
that IM administration is both non-inferior to SC administration in terms of 4-week post-vaccination geo-
metric mean titres (GMTs), and elicits an acceptable geometric mean fold-rise (GMFR) of antibody titres
Keywords:
Herpes zoster
measured by glycoprotein enzyme-linked immunosorbent assay. Pre-specified non-inferiority was set as
Shingles the lower bound of the 95% confidence interval (CI) of the GMT ratio (IM/SC) being >0.67. An acceptable
Varicella zoster virus vaccine GMFR for the IM route was pre-specified as the lower bound of its 95% CI being >1.4. Description of the
Intramuscular administration route VZV immune response using the interferon-gamma enzyme-linked immunospot (IFN-␥ ELISPOT) assay
Sub-cutaneous administration route and of the safety were secondary objectives.
Randomised controlled trial Participants were randomised to IM or SC administration (1:1). The baseline demographics were
comparable between groups; mean age: 62.6 years (range: 50.0–90.5). The primary immunogenicity
objectives were met (per protocol analysis): GMT ratio (IM/SC): 1.05 (95% CI: 0.93–1.18); GMFR: 2.7
(2.4–3.0). VZV immune response using IFN-␥ ELISPOT were comparable between groups. Frequencies
of systemic adverse events were comparable between groups. Injection-site reactions were less fre-
quent with IM than SC route: erythema (15.9% versus 52.5%), pain (25.6% versus 39.5%) and swelling
(13.6% versus 37.3%), respectively. In adults aged ≥50 years, IM administration of Zostavax elicited sim-
ilar immune responses to SC administration and was well tolerated, with fewer injection-site reactions
than with SC administration.
© 2014 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).

1. Introduction latent in the spinal ganglia and cranial sensory ganglia after pri-
mary VZV infection. The primary infection is seen as varicella
Herpes zoster (HZ), commonly known as shingles, results from or chickenpox [1–3]. HZ is characterised by a unilateral, painful,
the reactivation of varicella zoster virus (VZV), which remains vesicular rash that is usually limited to a single dermatome
[4].
In Europe, almost everybody is at risk of HZ, since more than 95%
∗ Corresponding author. 69367 Lyon Cedex 07, France. Tel.: +33 (0)437 284 579; of adults aged ≥40 years are VZV seropositive following varicella
fax: +33 (0)437 284 451. and 90% of individuals become VZV seropositive before adolescence
E-mail addresses: diez jav@gva.es (J. Diez-Domingo), tweinke@klinikumevb.de
[5,6]. There is a sharp increase in the incidence of HZ at the age of 50,
(T. Weinke), jglomas@gmail.com (J. Garcia de Lomas), meyer@uni-mainz.de
(C.U. Meyer), ibertrand@spmsd.com (I. Bertrand), ceymin@spmsd.com (C. Eymin),
with 2/3 of HZ cases occurring in adults aged ≥50 [7]. The lifetime
sthomas@spmsd.com (S. Thomas), csadorge@spmsd.com (C. Sadorge). risk of zoster is estimated to be 10–30% with incidence increasing

http://dx.doi.org/10.1016/j.vaccine.2014.12.024
0264-410X/© 2014 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
790 J. Diez-Domingo et al. / Vaccine 33 (2015) 789–795

with age, affecting up to 50% of people who live up to 85 years disease (e.g. human immunodeficiency virus, cancer) or medical
[7–10]. treatment (e.g. chemotherapy, transplant recipients).
The most frequent and debilitating complication of HZ is post- The study was carried out in accordance with the principles of
herpetic neuralgia (PHN), a neuropathic pain syndrome that can Good Clinical Practice and the ethical principles of the Declaration
persist for months, years or even decades after the HZ rash has of Helsinki. The study protocol was approved by the appropri-
gone [11,12]. The frequency and severity of PHN increases with ate institutional review boards and regulatory agencies. Written,
age, occurring in between 25% and 50% of patients with HZ aged informed consent was obtained from all subjects before study
≥50 years old [13–16]. Although the mechanisms responsible for entry.
maintaining VZV in its latent state and for its reactivation are not ClinicalTrials.gov identifier: NCT01391546 and EudraCT identi-
fully understood, evidence suggests that cell-mediated immunity fier: 2009-012458-19.
(CMI) is critical [17–19]. The age-related decline in VZV-specific
CMI has been shown to have temporal association with an increase 2.2. Study design
in the incidence and severity of HZ and its complications in older
adults [20–25]. This was an open-label, randomised, comparative, study con-
HZ and particularly PHN are debilitating and have a damaging ducted in seven centres in Germany and three in Spain from June
impact on multiple aspects of patients’ lives. They also have an 2011 to September 2012 to compare IM and SC administration of
impact on their physical functionality by interfering with activities one dose of Zostavax in adults aged ≥50 years. The subjects were
of daily living [26–32]. randomised using an electronic case report form (e-CRF). Alloca-
Zostavax® (commercialised in Europe by Sanofi Pasteur MSD, tion schedules were generated using a 1:1 ratio with permuted
manufactured by Merck Sharpe and Dohme) is a live, attenuated blocks of 4–6. The randomisation was stratified by ‘ELISPOT’ site
VZV vaccine developed specifically for the prevention of HZ and (see below) or not and by age with three strata: 50–59, 60–69 and
PHN in individuals aged ≥50 years [33]. The vaccine boosts VZV- ≥70 years (because the immune response has been shown to be
specific cell-mediated immunity (CMI); this boosted CMI controls age-dependent) [34,45,46]. There were no protocol amendments
reactivation of the latent VZV and/or replication and, therefore, after the start of subject recruitment.
prevents herpes zoster or attenuates its severity [17,34–39]. Blood samples were taken at visit 1, before vaccination, and at
In the large-scale Shingles Prevention Study (SPS) involving visit 2, 4 weeks after vaccination (between 28 and 35 days). The
more than 38,000 subjects aged ≥60 years, vaccine efficacy for humoral immune response was assessed using the glycoprotein
the prevention of HZ was 63.9% (95% confidence interval (CI): enzyme-linked immunosorbent assay (gpELISA) assay to measure
55.5–70.9) in individuals aged 60–69 years, and 37.6% (95% CI: the VZV antibody titre in all subjects and the CMI response was
25.0–48.1) in those aged ≥70 years [40,41]. The vaccine efficacy for assessed for a sub-set of subjects enrolled at designated sites,
the reduction in PHN was 65.7% (95% CI: 20.4–86.7) in those aged the ‘ELISPOT’ sites, using the interferon-gamma (IFN-␥)-enzyme-
60–69 years and 66.8% (95% CI: 43.3–81.3) for those aged ≥70 years. linked immunospot (ELISPOT) assay [47,48].
Zostavax has also been shown to reduce the burden of HZ-related Between visit 1 and 2, the participants were given a diary card
interference with daily living activities and the impact on health- to record their temperature if they were febrile (oral tempera-
related quality of life [42]. In the Zostavax Efficacy and Safety Trial ture ≥38.3 ◦ C), occurrence of any solicited injection-site (erythema,
(ZEST), involving more than 20,000 subjects aged 50–59 years, the swelling and pain) adverse reactions (Days 0–4) and any unsolicited
vaccine significantly reduced the risk of developing HZ by 69.8% injection-site adverse reactions, varicella, varicella-like rashes, HZ
(95% CI: 54.1–80.6) [43]. In Europe, Zostavax is indicated for the and zoster-like rashes and other systemic adverse events (AEs)
prevention of HZ and PHN in adults aged ≥50 years [44]. (Days 0–28). They were also asked to report any serious AEs (SAEs)
During clinical development, which was mainly done in the that occurred at any time during the study.
US, the vaccine was administered by the subcutaneous (SC) route.
The preference for the administration route can vary between 2.3. Study vaccine
healthcare professionals and between countries; in some European
countries the preferred administration route is intramuscular (IM). The study vaccine, Zostavax, was administered in the deltoid
The objectives of this trial were to compare the immunogenic- region of the upper arm, by IM (needle: 25 mm, 23 gauge) or
ity, safety and tolerability of Zostavax when administered by the SC (needle: 16 mm, 25 gauge) according to the randomisation.
IM route versus the SC route, to support and facilitate the use of Use of an anaesthetic cream or patch was not allowed. Each
the vaccine in Europe by offering the choice for the administration dose of 0.65 mL contained ≥19,400 PFU of varicella-zoster virus,
route. Oka/Merck strain (live, attenuated) (Lot Numbers: WL00040507;
WL00046785).

2. Methods 2.4. Objectives

2.1. Study population 2.4.1. Co-primary objectives


The two co-primary objectives were to demonstrate that IM
Healthy adults aged ≥50 years with a history of varicella or res- administration of Zostavax in healthy adults aged ≥50 years is
ident for >30 years in a country with endemic VZV infection were non-inferior to SC administration in terms of the 4-week post-
eligible. Subjects were excluded if they had previously been vac- vaccination VZV antibody geometric mean titre (GMT) measured
cinated with any VZV-containing vaccine or had previously been with gpELISA and that it induces an acceptable geometric mean
diagnosed with HZ. In addition, were excluded: any subjects with a fold rise [GMFR] of VZV antibody titres (gpELISA) from pre- to 4-
history of a febrile episode (≥38.3 ◦ C) in the 72 h prior to study vac- week post-vaccination. Non-inferiority was defined as the lower
cination, those who had received any live vaccine within 28 days bound of the two-sided 95% confidence interval (CI) around the
of study vaccination or inactivated vaccine within 14 days of study post-vaccination GMT ratio (IM group/SC group) being greater than
vaccination or immunoglobulins or other blood products within 5 2/3 (i.e. ruling out a decrease of ≥1.5-fold). An acceptable GMFR
months before vaccination and those who were taking systemic was defined as the lower bound of the two-sided 95% CI around
antiviral therapy or had an immune deficiency associated with the GMFR being greater than 1.4 in the IM group based on a

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
J. Diez-Domingo et al. / Vaccine 33 (2015) 789–795 791

Screened
N=359
(ELISPOT: N=228)
Screen failures, n=5
Met exclusion criteria (4) lost
to follow-up (1)
Randomised
N=354
(ELISPOT: N=228)

IM group SC group
N=177 N=177
(ELISPOT: N=115) (ELISPOT: N=113)

Past history of HZ, not vaccinated and no


blood drawn, n=1 No pre- or post-vaccination
ELISPOT results, n=2

No pre- or post-vaccination
ELISPOT results, n=1

ELISPOT FAS n=113 FAS and Safety Set FAS and Safety Set ELISPOT FAS n=111
n=176 n=177

Protocol deviation * which Immunosuppressive treatment


invalidated ELISPOT results, n=2 before & during the trial, n=1

ELISPOT PPS n=111 PPS n=175 PPS n=177 ELISPOT PPS n=111

* one subject exposed to VZV and one subject with a rash of interest following vaccination

Fig. 1. Disposition of study participants. IM: intramuscular; SC: subcutaneous; BS: blood sample; PPS: per protocol set; FAS: full analysis set.

minimum increase that was established using the results from 2.6. Statistics
the SPS trial [41]. Success of the trial required that both of the
co-primary objectives were satisfied. SAS® (V9.1.3; SAS Institute Inc., Cary, North Carolina, USA) was
used to perform the statistical analyses. The immunogenicity anal-
yses were performed on the per protocol set (PPS) that included all
2.4.2. Secondary objectives randomised participants who received the study vaccine and had
The secondary immunological objectives were to describe the at least one valid (either pre- or post-vaccination) immunogenicity
gpELISA GMT and GMFR in both groups and the IFN-␥ ELISPOT GMC evaluation for VZV antibody (PPS) or VZV ELISPOT count (ELISPOT
and GMFR in both groups in the ELISPOT sub-set at 4-weeks post- PPS). The gpELISA GMT ratio (IM group/SC group) and the corre-
vaccination. The secondary safety objectives were to describe and sponding 95% CI were calculated using a longitudinal data analysis
compare the safety profiles of Zostavax administered by the IM and model including pre- and post-vaccination log-transformed titres
SC routes. as response variables and age at vaccination, group and visit (pre-
and post-vaccination) as covariates [50]. This model handles miss-
ing data and provides the same results as an ANCOVA model when
2.5. Sample size calculation
there are no subjects with missing data. The CI around the GMFR
in the IM group was calculated using Student’s t distribution for
It was planned to randomise 177 subjects to each group
paired samples on log-transformed data.
(N = 354). Assuming that 90% of the participants would be evaluable,
The safety analyses were performed on all participants who
this sample size would give 159 evaluable subjects per group for
received the study vaccine and who had safety follow-up (safety
the main analysis (per protocol). If the true GMT ratio (IM group/SC
analysis set). Data were analysed according to the administration
group) was 1.0 (with a standard deviation of 1.1 on a natural log
route actually used for the vaccine. Descriptive statistics and risk
scale) and the true GMFR was 2.0 (with a standard deviation of 1.0
differences between the IM and SC groups for the comparison of
on a natural log scale), the study would have an overall power of
the safety data are provided.
90% based on a 2.5% one-sided alpha-error rate.
Descriptive analyses were performed for the ELISPOT subgroup.
The number of the subjects to be enrolled in the subgroup was 3. Results
decided, based on previous experience, so that it would be possi-
ble to provide a reliable estimate of the post-vaccination ELISPOT 3.1. Subject disposition and baseline characteristics
GMC [49]. The planned sample size was 80 evaluable subjects per
group, giving 114 randomised subjects per group, assuming that A total of 354 of the 359 screened subjects were randomised
70% of the subjects would be evaluable for the ELISPOT per protocol to receive ZOSTAVAX by the IM route (N = 177; ELISPOT sub-
analysis. set N = 115) or SC route (N = 177; ELISPOT sub-set N = 113); 353

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
792 J. Diez-Domingo et al. / Vaccine 33 (2015) 789–795

Table 1
Demographic characteristics of subjects in the randomised set.

IM group (N = 177) SC group (N = 177) Total (N = 354)

Age at vaccination (years)a


Mean (±SD) 62.6 (±8.3) 62.6 (±8.5) 62.6 (±8.4)
Median (min; max) 61.1 (50.0; 90.5) 61.1 (50.1; 86.1) 61.1 (50.0; 90.5)

Age group (years) n (%)


50–59 81 (45.8) 82 (46.3) 163 (46.0)
60–69 56 (31.6) 55 (31.1) 111 (31.4)
≥70 40 (22.6) 40 (22.6) 80 (22.6)

Gender n (%)
Male 79 (44.6) 80 (45.2) 159 (44.9)
Female 98 (55.4) 97 (54.8) 195 (55.1)
a
Data missing for one subject in the IM group.

Table 2
Pre- and 4-week post-vaccination GMTs and GMFRs of VZV antibody titres (gpELISA units/mL; per protocol seta ).

IM route group (N = 175) SC route group (N = 177)

n GMT (95% CI) n GMT (95% CI)

Pre-vaccination 175 144.6 (125.3–166.9) 176 158.9 (137.9–183.1)


Post-vaccination 168 395.3 (350.6–445.6) 173 391.7 (348.9–439.7)

IM route group (N = 175) SC route group (N = 177)

n GMFR (95% CI) n GMFR (95% CI)

Post-/pre-vaccination 168 2.7 (2.4–3.0) 172 2.5 (2.2–2.8)

CI, confidence interval; GMFR, geometric mean fold-rise; GMT, geometric mean titre; gpELISA, glycoprotein enzyme-linked immunosorbent assay; IM, intramuscular; N,
number of subjects; n, number of subjects with available data; SC, subcutaneous; VZV, varicella-zoster virus.
a
The per protocol analysis set for the primary outcome included all subjects with at least one valid for pre- or post-vaccination immunogenicity evaluation.

subjects received the study vaccine (Fig. 1). One randomised subject 100
in the IM group belonging to the ELISPOT subset was not vaccinated 90
due to past history of herpes zoster, which was an exclusion criteria,
80
and no blood sample was provided, thus the full analysis set (FAS)
and the safety analysis set included 353 subjects (IM: n = 176; SC: 70

n = 177). In addition, in the IM group, one subject received immuno- 60


suppressive therapy prior to vaccination and during the trial. Thus
Subjects (%)

50
352 subjects (IM: n = 175; SC: n = 177) were included in the per
40
protocol set (PPS). Four subjects were excluded from the ELISPOT
randomised sub-set: the subject with a past history of herpes zoster 30
and three other subjects because there were no pre- and post- 20
vaccination ELISPOT results (due to loss of sample during transfer to
10
the laboratory for testing; IM: n = 1; SC: n = 2); thus the ELISPOT FAS
included 224 subjects (IM: n = 113; SC: n = 111). Two additional sub- 0
1 10 100 1,000 10,000
jects were excluded from the IM ELISPOT sub-set due to exposure
Log anti VZV titre (units/mL)
to VZV and rash of interest following vaccination; these protocol
deviations invalidated their post-vaccination ELISPOT results; thus Fig. 2. Reverse cumulative distribution curve of VZV antibody titres pre-vaccination
the ELISPOT PPS included 222 subjects (IM: n = 111; SC: n = 111). and 4-week post vaccination with ZOSTAVAX (IM or SC route) measured by gpELISA
The two groups were comparable in terms of age and gender (per-protocol set). The solid lines show the results for the subjects in the IM group
and the dashed lines show those for subjects in the SC group. IM: intramuscular; SC:
(Table 1). The baseline characteristics for the ELISPOT randomised
subcutaneous; VZV: varicella-zoster virus.
subset were comparable to those of the overall randomised
set.
3.2.2. Secondary immunogenicity objectives
The pre-vaccination and 4-week post-vaccination VZV antibody
3.2. Immunogenicity GMTs and GMFRs were comparable between groups (Table 2 and
Fig. 2). There was a trend for these values to decrease with increas-
3.2.1. Primary immunogenicity objectives ing age (Table 3). The pre-vaccination and 4-week post-vaccination
Both primary immunogenicity objectives were met. The 4-week ELISPOT GMCs and the ELISPOT GMFR were comparable between
post-vaccination VZV antibody GMT in the IM group was non- groups (Table 4).
inferior to that in the SC group (lower bound of the 95% CI of
the GMT ratio (IM/SC) >0.67). The estimated GMT ratio (IM/SC) 3.3. Safety
was 1.05 (95% CI: 0.93–1.18) (p < 0.001). In the IM group, the esti-
mated GMFR of the VZV antibody titres from pre-vaccination to Fewer participants in the IM group experienced at least one
4-week post-vaccination was 2.7 (95% CI: 2.4–3.0) which allowed AE compared with those in the SC group (47.2% versus 69.5%,
to conclude that it was acceptable (lower bound of the 95% CI >1.4) respectively; Table 5). The rates of systemic AEs were compara-
(Table 2). ble between the administration routes (23.3% versus 22.6% for IM

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
J. Diez-Domingo et al. / Vaccine 33 (2015) 789–795 793

Table 3
Pre- and 4-week post-vaccination geometric mean titres (GMTs) and geometric mean fold-rises (GMFR; post/pre-vaccination) of VZV antibody titres (gpELISA units/mL) by
age group – per protocol set.

IM group (N = 175) SC group (N = 177)

Pre-vaccination Post-vaccination Pre-vaccination Post-vaccination

50–59 years old


n 79 74 82 79
GMT [95% CI] 133.9 [108.9;164.7] 446.1 [368.1;540.6] 164.8 [134.3;202.3] 454.1 [390.1;528.5]
GMFR [95% CI] 3.2 [2.7;3.9] 2.8 [2.3;3.4]

60–69 years old


n 56 55 54 54
GMT [95% CI] 154.5 [117.4;203.3] 394.7 [321.1;485.2] 158.7 [121.4;207.4] 338.6 [273.1;419.8]
GMFR [95% CI] 2.5 [2.0;3.1] 2.2a [1.8;2.6]

≥70 years old


n 40 39 40 40
GMT [95% CI] 153.4 [112.8;208.5] 314.8 [250.7;395.3] 147.7 [108.1;201.9] 356.3 [268.9;471.9]
GMFR [95% CI] 2.1 [1.8;2.5] 2.4 [1.9;3.0]
a
Data for one subject missing: n = 53; CI, confidence interval; VZV, varicella-zoster virus.

Table 4
Pre- and 4-week post-vaccination GMC and GMFR of VZV INF-γ ELISPOT counts (ELISPOT per protocol set).

IM route group (N = 111) SC route group (N = 111)


a
n GMC (95% CI) na GMC (95% CI)

Pre-vaccination 101 64.3 (49.6–83.4) 96 58.4 (42.6–80.2)


Post-vaccination 103 209.8 (175.2–251.3) 105 195.7 (161.9–236.6)

IM route group (N = 111) SC route group (N = 111)

na GMFR (95% CI) na GMFR (95% CI)

Post-/pre-vaccination 93 3.3 (2.8–3.9) 90 3.4 (2.7–4.3)

CI, confidence interval; GMC, geometric mean count; GMFR, geometric mean fold-rise; IM, intramuscular; INF, interferon; N, number of participants; n, number of participants
with available data; SC, subcutaneous; VZV, varicella-zoster virus.
a
Missing data for pre-vaccination time points are due to non-evaluable samples. Missing data for post-vaccination time points are due to protocol deviations and thus
results were excluded from the per protocol analyses.

Table 5 70
-36.6
Summary of safety (safety analysis set). (-45.4; -27.2)
p<0.001
IM group (N = 176) SC group (N = 177) 60

n (%) n (%) -14.0


(-23.5; -4.2) -23.7
50 p=0.005 (-32.3; -14.8)
AE (Days 0–28) 83 (47.2) 123 (69.5) p<0.001
Vaccine-related AE (Days 0–28) 68 (38.6) 118 (66.7)
% of subjects

Injection site reaction (Days 0–28) 60 (34.1) 114 (64.4) 40


Solicited injection site reaction 58 (33.0) 110 (62.1)
(Days 0–4)
30
Unsolicited injection site 9 (5.1) 14 (7.9)
reaction (Days 0–28)
20
Systemic AE (Days 0–28) 41 (23.3) 40 (22.6)
-4.5
Vaccine-related systemic AE 12 (6.8) 13 (7.3) (-9.3; -0.5)
(Days 0–28) 10

Injection site rash (Days 0–28) 0 0


Non-injection site rash (Days 0–28) 1 (0.6) 0 0
Erythema Pain Swelling Pruritus
SAE 1 (0.6) 2 (1.1)
Vaccine-related SAE 0 0
Fig. 3. Percentage of subjects (and 95% CI) with injection site adverse reactions from
Withdrawal due to an AE 0 0 Days 0 to 28 (safety analysis set) for the IM group (N = 176; grey bars) and the SC
group (N = 177; white bars). The numbers above the bars are the percentage risk
AE, adverse event; IM, intramuscular; N, number of participants; n (%), number (per-
differences between the IM and SC groups with their 95% confidence intervals. p
centage) of participants presenting the AE at least once; SAE, serious adverse event;
values were calculated for the solicited adverse reactions only, in compliance with
SC, subcutaneous.
the protocol; pruritus was an unsolicited adverse reaction and therefore no p value
was calculated.
and SC groups, respectively; Table 5). Injection-site reactions were
significantly less frequent in the IM group than in the SC group
for: erythema (15.9% versus 52.5%; p < 0.001), pain (25.6% versus by PCR testing, the investigator considered the event to be possi-
39.5%; p = 0.005), swelling (13.6% versus 37.3%; p < 0.001) and pru- bly related to the study vaccine. Three subjects experienced SAEs
ritus (1.7% versus 6.2%), respectively (Fig. 3). One subject in the (IM Group: obstructive hernia (n = 1); SC Group: humerus fracture
IM group reported a zoster-like rash (right thoracic dermatome) of (n = 1), deep venous thrombosis (n = 1)), none of which were con-
mild intensity that occurred 12 days post vaccination and lasted sidered to be vaccine-related by the investigators (Table 5). None
6 days. Although no specimen was obtained for VZV confirmation of the subjects withdrew from the trial because of an AE.

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
794 J. Diez-Domingo et al. / Vaccine 33 (2015) 789–795

4. Discussion Authors’ contributions

This study was designed to assess the immunogenicity and All authors participated in the implementation and/or conduct
safety of IM administration of Zostavax compared with SC adminis- of the study including substantial contributions to conception and
tration. The results show that the immune response to IM Zostavax design, the gathering of the data, or analysis and interpretation of
is similar to that to SC Zostavax. In addition, IM administration the data. All the authors revised the manuscript critically for impor-
results in significantly fewer injection-site reactions with a compa- tant intellectual content and approved the final version before
rable rate of systemic AEs, as has been described for other vaccines submission.
[51,52].
The estimated GMFR, obtained using the gpELISA assay, was Financial disclosure
comparable to those observed in previous studies that evaluated
immune response at 4-week post-vaccination in adults aged ≥50 Sanofi Pasteur MSD funded this clinical trial and was involved
years [34,45,46]. Unlike our trial, these three trials had only two in all stages of the study conduct and analysis. Sanofi Pasteur MSD
age groups: 50–59 years and >60 years, therefore we could only also funded the development and publication costs for the present
compare the 50–59 age groups in which the values ranged from manuscript. All authors had full access to the data and the cor-
2.4 to 3.9. As expected and as already reported in other stud- responding author had final responsibility for submission of the
ies, the estimated GMFRs were numerically higher in those aged manuscript.
50–59 years (3.2 in the IM group and 2.8 in the SC group) com-
pared with those who were older (60–69 years: 2.5 and 2.2; ≥70
Conflicts of interest
years: 2.1 and 2.4, in the IM and SC groups, respectively). The
lower immunological response observed with age is due to the phe-
JDD has been and is the principal investigator in trials sponsored
nomenon of immunosenescence, or ageing of the immune system
by Sanofi Pasteur MSD, GSK, Merck, Baxter, Novartis and Pfizer. His
[53,54].
institutions have received research grants from Sanofi Pasteur MSD,
The estimated GMFRs obtained using the IFN-␥ ELISPOT assay
Pfizer and Baxter. He has received grants for attending meeting and
in a subset of subjects (N = 228) enrolled at 4 of the 10 study sites
has been a member of advisory boards for GSK, Pfizer and Sanofi
were comparable between the two groups: 3.3 in IM group and 3.4
Pasteur MSD.
in SC group. The IFN-␥ ELISPOT was used in two previous studies in
TW has received honoraria for lecturing and consulting activities
older populations (≥60 years) [49,55]. In one of these studies, the
from Novartis Vaccines and Sanofi Pasteur MSD.
estimated GMFR was 2.1 at 6-weeks post-vaccination and 2.7 and
JGDL and CUM have no potential conflicts of interest to declare.
2.1 at 2 and 6-weeks post-vaccination, respectively, in the other
IB, CE, ST and CS are employed by Sanofi Pasteur MSD, the com-
study [49,55].
pany that commercialises the herpes zoster live-attenuated vaccine
The safety profile of Zostavax in the SC group was compara-
(Zostavax® ) in Europe.
ble to the known safety profile of the product [56]. In the IM
group, there were fewer injection-site reactions than in the SC
group. Lower rates of injection-site reactions were also reported Acknowledgements
in two studies in healthy children that compared the IM and
SC administration of varicella vaccine, Varivax® and a combina- The authors would like to thank the subjects who participated in
tion paediatric vaccine, ProQuad® , which contain the same VZV the trial, as well as the following investigators for their contribution
strain [57,58]. A literature review of clinical trials reported that in to the study: Rolf Dominicus (Dülmen, Germany); Christine Gri-
two trials comparing immunogenicity or safety of pneumococcal gat, Martina Lilie (Hamburg, Germany); Dorothee Kieninger-Baum
vaccines in adults and in four trials comparing immunogenic- (Mainz, Germany); Wiefried Steinborn (Straubing, Germany); Peter
ity or safety of influenza vaccines administered via IM or SC Kindermann (Riesa, Germany); Enrique Guinot, Vincenta Pineda
routes in adults, the immunogenicity and systemic safety were (Valencia, Spain).
at least as good by IM administration compared with SC, and They also would like to thank Xavier Cornen, Sylvie Dard, Nicolas
that reactogenicity was generally better with IM administration Goulleret and Elodie Turpaud (Sanofi Pasteur MSD), Martina König
[51]. (THEOREM Clinical Research GmbH & Co. KG) for their contribu-
One possible limitation of this study was the open-label design tion to the conduct of the study; Janie Parino and Paula Annunziato
for the routes of administration. However, randomisation was per- (Merck & Co., Inc.) for their input during the study design and for
formed using an eCRF thus minimising any subject selection bias. their critical review of the study results and Laura Malette and Bar-
In addition, the immunogenicity assays were performed on blinded bara E. Meyer (PPD Vaccine and Biologics, LLC, Wayne, PA, USA)
samples so that the laboratory staff who performed the immuno- for overseeing the gpELISA assays and Julie Bowman (ViraCor-IBT
genicity assays (including primary criteria) were unaware of the Laboratories Inc, Lenexa, KS, USA) for overseeing the IFN-␥ ELISPOT
vaccination administration route. The present study had an excel- assay.
lent completion rate, with only two randomised subjects (2/354; The authors take full responsibility for the content of this
0.6%) not completing the trial. manuscript and thank Margaret Haugh from MediCom Consult,
Villeurbanne, France (funded by Sanofi Pasteur MSD) for providing
medical writing and editorial assistance.
5. Conclusion
References
In adults aged ≥50 years, IM administration of Zostavax elicited
similar immune responses to SC administration and was well [1] Arvin AM. Varicella-zoster virus: molecular virology and virus–host interac-
tions. Curr Opin Microbiol 2001;4:442–9.
tolerated with fewer injection-site reactions than with SC admin- [2] Gauthier A, Breuer J, Carrington D, Martin M, Remy V. Epidemiology and cost
istration. These data support the administration of vaccine using of herpes zoster and post-herpetic neuralgia in the United Kingdom. Epidemiol
either SC or IM and thus could allow healthcare professionals the Infect 2009;137:38–47.
[3] Johnson RW, Wasner G, Saddier P, Baron R. Herpes zoster and posther-
flexibility to choose their preferred administration route in routine petic neuralgia: optimizing management in the elderly patient. Drugs Aging
clinical practice. 2008;25:991–1006.

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.
J. Diez-Domingo et al. / Vaccine 33 (2015) 789–795 795

[4] Gnann Jr JW, Whitley RJ. Clinical practice. Herpes zoster. N Engl J Med study of a refrigerator-stable formulation of Zostavax. Clin Vaccine Immunol
2002;347:340–6. 2008;15:314–9.
[5] Nardone A, de Ory F, Carton M, Cohen D, van Damme P, Davidkin I, et al. The [35] Levin MJ, Barber D, Goldblatt E, Jones M, LaFleur B, Chan C, et al. Use of a live
comparative sero-epidemiology of varicella zoster virus in 11 countries in the attenuated varicella vaccine to boost varicella-specific immune responses in
European region. Vaccine 2007;25:7866–72. seropositive people 55 years of age and older: duration of booster effect. J Infect
[6] Sengupta N, Taha Y, Scott FT, Leedham-Green ME, Quinlivan M, Breuer Dis 1998;178(Suppl. 1):S109–12.
J. Varicella-zoster-virus genotypes in East London: a prospective study in [36] Levin MJ, Murray M, Rotbart HA, Zerbe GO, White CJ, Hayward AR. Immune
patients with herpes zoster. J Infect Dis 2007;196:1014–20. response of elderly individuals to a live attenuated varicella vaccine. J Infect
[7] Pinchinat S, Cebrian-Cuenca AM, Bricout H, Johnson RW. Similar herpes zoster Dis 1992;166:253–9.
incidence across Europe: results from a systematic literature review. BMC Infect [37] Levin MJ, Smith JG, Kaufhold RM, Barber D, Hayward AR, Chan CY, et al. Decline
Dis 2013;13:170. in varicella-zoster virus (VZV)-specific cell-mediated immunity with increas-
[8] Brisson M, Edmunds WJ, Law B, Gay NJ, Walld R, Brownell M, et al. Epidemiology ing age and boosting with a high-dose VZV vaccine. J Infect Dis 2003;188:
of varicella zoster virus infection in Canada and the United Kingdom. Epidemiol 1336–44.
Infect 2001;127:305–14. [38] Oxman MN. Immunization to reduce the frequency and severity of herpes
[9] Hope-Simpson RE. The nature of herpes zoster: a long-term study and a new zoster and its complications. Neurology 1995;45:S41–6.
hypothesis. Proc R Soc Med 1965;58:9–20. [39] Trannoy E, Berger R, Hollander G, Bailleux F, Heimendinger P, Vuillier D, et al.
[10] Schmader K. Herpes zoster in older adults. Clin Infect Dis 2001;32:1481–6. Vaccination of immunocompetent elderly subjects with a live attenuated Oka
[11] Mick G, Hans G. Postherpetic neuralgia in Europe: the scale of the problem and strain of varicella zoster virus: a randomized, controlled, dose–response trial.
outlook for the future. J Clin Gerontol Geriat 2013;4:102–8. Vaccine 2000;18:1700–6.
[12] Johnson RW, Rice AS. Clinical practice. Postherpetic neuralgia. N Engl J Med [40] Oxman MN, Levin MJ. Vaccination against herpes zoster and postherpetic neu-
2014;371:1526–33. ralgia. J Infect Dis 2008;197(Suppl. 2):S228–36.
[13] Dworkin RH, Schmader KE. The epidemiology and natural history of herpes [41] Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, et al. A
zoster and postherpectic neuralgia. In: Watson CPN, Gershon AA, editors. Pain vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N
research and clinical management. 11th ed. Amsterdam, The Netherlands: Else- Engl J Med 2005;352:2271–84.
vier Science; 2001. p. 39–64. [42] Schmader KE, Johnson GR, Saddier P, Ciarleglio M, Wang WW, Zhang JH, et al.
[14] Johnson RW, Wasner G, Saddier P, Baron R. Postherpetic neuralgia: Effect of a zoster vaccine on herpes zoster-related interference with functional
epidemiology, pathophysiology and management. Expert Rev Neurother status and health-related quality-of-life measures in older adults. J Am Geriatr
2007;7:1581–95. Soc 2010;58:1634–41.
[15] Scott FT, Leedham-Green ME, Barrett-Muir WY, Hawrami K, Gallagher WJ, John- [43] Schmader KE, Levin MJ, Gnann Jr JW, McNeil SA, Vesikari T, Betts RF, et al.
son R, et al. A study of shingles and the development of postherpetic neuralgia Efficacy, safety, and tolerability of herpes zoster vaccine in persons aged 50–59
in East London. J Med Virol 2003;70(Suppl. 1):S24–30. years. Clin Infect Dis 2012;54:922–8.
[16] Yawn BP, Saddier P, Wollan PC, St Sauver JL, Kurland MJ, Sy LS. A population- [44] EMA. ZOSTAVAX SmPC; 2014. http://www.ema.europa.eu/ema/index.jsp?curl=
based study of the incidence and complication rates of herpes zoster before pages/medicines/human/medicines/000674/human med 001185.jsp&mid=
zoster vaccine introduction. Mayo Clin Proc 2007;82:1341–9. WC0b01ac058001d124 [accessed 5.12.14].
[17] Frazer IH, Levin MJ. Paradigm shifting vaccines: prophylactic vaccines against [45] Arnou R, Fiquet A, Thomas S, Sadorge C. Immunogenicity and safety of
latent varicella-zoster virus infection and against HPV-associated cancer. Curr ZOSTAVAX® approaching expiry potency in individuals aged ≥50 years. Hum
Opin Virol 2011;1:268–79. Vaccin 2011;7:1060–5.
[18] Levin MJ, Schmader KE, Gnann JW, McNeil SA, Vesikari T, Betts RF, et al. [46] Kerzner B, Murray AV, Cheng E, Ifle R, Harvey PR, Tomlinson M, et al. Safety
Varicella-zoster virus-specific antibody responses in 50–59-year-old recipients and immunogenicity profile of the concomitant administration of ZOSTAVAX
of zoster vaccine. J Infect Dis 2013;208:1386–90. and inactivated influenza vaccine in adults aged 50 and older. J Am Geriatr Soc
[19] Oxman MN. Zoster vaccine: current status and future prospects. Clin Infect Dis 2007;55:1499–507.
2010;51:197–213. [47] Krah DL, Cho I, Schofield T, Ellis RW. Comparison of gpELISA and neutralizing
[20] Arvin A. Aging, immunity, and the varicella-zoster virus. N Engl J Med antibody responses to Oka/Merck live varicella vaccine (Varivax) in children
2005;352:2266–7. and adults. Vaccine 1997;15:61–4.
[21] Aw D, Silva AB, Palmer DB. Immunosenescence: emerging challenges for an [48] Smith JG, Levin M, Vessey R, Chan IS, Hayward AR, Liu X, et al. Measurement
ageing population. Immunology 2007;120:435–46. of cell-mediated immunity with a Varicella-Zoster Virus-specific interferon-
[22] Berger R, Florent G, Just M. Decrease of the lymphoproliferative response to gamma ELISPOT assay: responses in an elderly population receiving a booster
varicella-zoster virus antigen in the aged. Infect Immun 1981;32:24–7. immunization. J Med Virol 2003;70(Suppl. 1):S38–41.
[23] Hayward AR, Herberger M. Lymphocyte responses to varicella zoster virus in [49] Vermeulen JN, Lange JM, Tyring SK, Peters PH, Nunez M, Poland G, et al. Safety,
the elderly. J Clin Immunol 1987;7:174–8. tolerability, and immunogenicity after 1 and 2 doses of zoster vaccine in healthy
[24] Miller AE. Selective decline in cellular immune response to varicella-zoster in adults >/=60 years of age. Vaccine 2012;30:904–10.
the elderly. Neurology 1980;30:582–7. [50] Liang KY, Zeger SL. Longitudinal data analysis of continuous and discrete
[25] Weinberg A, Lazar AA, Zerbe GO, Hayward AR, Chan IS, Vessey R, et al. Influence responses for pre-post designs. Sankhya 2000;62:134–48.
of age and nature of primary infection on varicella-zoster virus-specific cell- [51] Ajana F, Sana C, Caulin E. Are there differences in immunogenicity and safety
mediated immune responses. J Infect Dis 2010;201:1024–30. of vaccines according to the injection method? Med Mal Infect 2008;38:
[26] Bouhassira D, Chassany O, Gaillat J, Hanslik T, Launay O, Mann C, et al. 648–57.
Patient perspective on herpes zoster and its complications: an observational [52] Herzog C. Influence of parenteral administration routes and additional factors
prospective study in patients aged over 50 years in general practice. Pain on vaccine safety and immunogenicity: a review of recent literature. Expert
2012;153:342–9. Rev Vaccines 2014;13:399–415.
[27] Drolet M, Brisson M, Schmader KE, Levin MJ, Johnson R, Oxman MN, et al. The [53] Pawelec G. Hallmarks of human “immunosenescence”: adaptation or dysreg-
impact of herpes zoster and postherpetic neuralgia on health-related quality ulation? Immun Ageing 2012;9:15.
of life: a prospective study. CMAJ 2010;182:1731–6. [54] Weinberger B, Herndler-Brandstetter D, Schwanninger A, Weiskopf D,
[28] Gavazzi G, Krause KH. Ageing and infection. Lancet Infect Dis 2002;2:659–66. Grubeck-Loebenstein B. Biology of immune responses to vaccines in elderly
[29] Johnson RW, Bouhassira D, Kassianos G, Leplege A, Schmader KE, Weinke T. persons. Clin Infect Dis 2008;46:1078–84.
The impact of herpes zoster and post-herpetic neuralgia on quality-of-life. BMC [55] Levin MJ, Oxman MN, Zhang JH, Johnson GR, Stanley H, Hayward AR, et al.
Med 2010;8:37. Varicella-zoster virus-specific immune responses in elderly recipients of a her-
[30] Pickering G, Leplege A. Herpes zoster pain, postherpetic neuralgia, and quality pes zoster vaccine. J Infect Dis 2008;197:825–35.
of life in the elderly. Pain Pract 2011;11:397–402. [56] EMA. ZOSTAVAX SmPC; 2013. http://www.ema.europa.eu/ema/index.jsp?curl=
[31] Schmader KE, Sloane R, Pieper C, Coplan PM, Nikas A, Saddier P, et al. The impact pages/medicines/human/medicines/000674/human med 001185.jsp&mid=
of acute herpes zoster pain and discomfort on functional status and quality of WC0b01ac058001d124 [accessed 18.7.13].
life in older adults. Clin J Pain 2007;23:490–6. [57] EMA. ProQuad SmPC; 2014. http://www.ema.europa.eu/ema/index.jsp?curl=
[32] Weinke T, Edte A, Schmitt S, Lukas K. Impact of herpes zoster and post-herpetic pages/medicines/human/medicines/000622/human med 000997.jsp&mid=
neuralgia on patients’ quality of life: a patient-reported outcomes survey. Z WC0b01ac058001d124 [accessed 5.12.14].
Gesundh Wiss 2010;18:367–74. [58] Gillet Y, Habermehl P, Thomas S, Eymin C, Fiquet A. Immunogenicity and
[33] Levin MJ. Immune senescence and vaccines to prevent herpes zoster in older safety of concomitant administration of a measles, mumps and rubella vaccine
persons. Curr Opin Immunol 2012;24:494–500. (M-M-RvaxPro) and a varicella vaccine (VARIVAX) by intramuscular or subcu-
[34] Gilderman LI, Lawless JF, Nolen TM, Sterling T, Rutledge RZ, Fernsler DA, et al. A taneous routes at separate injection sites: a randomised clinical trial. BMC Med
double-blind, randomized, controlled, multicenter safety and immunogenicity 2009;7:16.

Downloaded for Mahasiswa KTI UKDW (mhsktiukdw01@gmail.com) at Universitas Kristen Duta Wacana from ClinicalKey.com by Elsevier on April 07, 2019.
For personal use only. No other uses without permission. Copyright ©2019. Elsevier Inc. All rights reserved.

You might also like