Anda di halaman 1dari 8

Vaccine 30 (2012) 2556–2563

Contents lists available at SciVerse ScienceDirect

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

Comparison of safety and immunogenicity of two doses of investigational


hepatitis B virus surface antigen co-administered with an immunostimulatory
phosphorothioate oligodeoxyribonucleotide and three doses of a licensed
hepatitis B vaccine in healthy adults 18–55 years of age!,!!,!,!!
Scott A. Halperin a,∗ , Brian Ward b , Curtis Cooper c , Gerald Predy d , Francisco Diaz-Mitoma e ,
Marc Dionne f , Joanne Embree g , Allison McGeer h , Paul Zickler i , Karl-Heinz Moltz j , René Martz k ,
Ingo Meyer l , Shelly McNeil a , Joanne M. Langley a , Eduardo Martins m , William L. Heyward m ,
J. Tyler Martin m
a
Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, and Capital Health, Halifax, Canada
b
McGill University Health Centre-Vaccine Study Centre, Montreal, Canada
c
Ottawa Hospital, Ottawa, Canada
d
Alberta Health Services, Edmonton, Canada
e
Northern Ontario Research Institute, Sudbury, Canada
f
Institut national de santé publique du Québec, Beauport, Canada
g
University of Manitoba, Winnipeg, Canada
h
Mount Sinai Hospital, Toronto, Canada
i
TASC Research Services, Surrey, Canada
j
APEX GmbH, München, Germany
k
Clinical Research Hamburg, Hamburg, Germany
l
Momentum Pharma Services, Hamburg, Germany
m
Dynavax Technologies Corporation, Berkeley, United States

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

Article history: Background: The currently licensed aluminum-hydroxide-adjuvanted hepatitis B vaccines require three
Received 19 September 2011 doses over a 6-month period to achieve high rates of protection in adults. We compared tolerability and
Received in revised form immunogenicity of two doses of an investigational hepatitis B vaccine using hepatitis B surface antigen
23 December 2011
adjuvanted with an immunostimulatory phosphorothioate oligodeoxyribonucleotide (HBV-ISS) to three
Accepted 28 January 2012
doses of a licensed alum-adjuvanted vaccine (HBV-Eng).
Available online 9 February 2012
Methods: In this randomized, observer-blind study, healthy adults received two doses of HBV-ISS at 0
and 4 weeks or three doses of HBV-Eng at 0, 4, and 24 weeks. The primary immunogenicity endpoint was
Keywords:
Hepatitis B vaccine
the seroprotection rate (antibody ≥ 10 mIU/mL) 8 weeks after the second dose of HBV-ISS compared to 4
Immunostimulatory sequences weeks after the third dose of HBV-Eng.
Randomized trial Results: A total of 2415 participants were randomized in a ratio of 3:1 to HBV-ISS (n = 1809) and HBV-Eng
(n = 606). The percentage of subjects exhibiting a seroprotective immune response at the primary time
point was significantly higher (95.1%) for HBV-ISS than for HBV-Eng (81.1%). Superiority of the seropro-
tective rates for HBV-ISS was demonstrated at all time points measured. Geometric mean concentrations

! Additional investigators and study sites: Ben Lasko, Manna Research, Toronto, Canada; Randy Hart, White Hills Medical Clinic, St. John’s, Canada; Dennis O’Keefe,
Commonwealth Medical Clinic, Mount Pearl, Canada; Martyn Chilvers, Sarnia Institute of Clinical Research, Sarnia, Canada; Randy MacKinnon, Island Clinical Trials,
Charlottetown, Canada; Isabelle Schenkenberger, Klinische Forschung Berlin, Berlin, Germany; Regina Sennewald, Pharos GmbH, Ulm, Germany; Werner Weber,
Pharos GmbH, Ulm, Germany; Gabriele Illies, SMO.MD GmbH, Magdeburg, Germany; and Olga Maus, Zentrum für Therapiestudien der Innomed GmbH, Leipzig, Germany.
!! Presented in part at the Digestive Disease Week Meeting, Chicago, Illinois, May 30–June 4, 2009 and the 48th Annual Meeting of the Infectious Diseases
Society of America, Vancouver, British Columbia, Canada, October 21–24, 2010.
! Drs. Martin, Heyward, and Martins were employees of Dynavax Technologies during the planning, implementation, and/or analysis of the study (Dr. Martins’
current address is Gilead Sciences Inc, Foster City, CA). All other authors had no financial interest in the vaccine or its manufacturer but received research funding to
undertake the study.
!! The study was funded by Dynavax Technologies Corporation.
∗ Corresponding author at: Canadian Center for Vaccinology, Dalhousie University, IWK Health Centre, 5850/5980 University Avenue, Halifax, Nova Scotia, B3K6R8,
Canada. Tel.: +1 902 470 8141; fax: +1 902 470 7232.
E-mail address: scott.halperin@dal.ca (S.A. Halperin).

0264-410X/$ – see front matter © 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.vaccine.2012.01.087
S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563 2557

were also significantly higher in the HBV-ISS group at all time points measured except at week 28
(24 weeks post-second dose of HBV-ISS and 4 weeks post-third dose HBV-ISS) at which time the anti-
body concentrations were similar. Both vaccines were welltolerated although injection-site reactions
were reported at a higher rate in HBV-ISS recipients. A short, two-dose regimen of HBV-ISS induced a
superior antibody response than a three-dose regimen of a licensed hepatitis B vaccine and was well
tolerated.
© 2012 Elsevier Ltd. All rights reserved.

1. Introduction alum-adjuvanted, licensed hepatitis B vaccine in healthy adoles-


cents and adults.
Hepatitis B virus (HBV) infection has a worldwide distribu-
tion and can cause a serious, potentially fatal liver infection. HBV 2. Methods
can be transmitted vertically from mother to newborn, horizon-
tally amongst family members, through sexual contacts, and via
exchange of blood (injection drug use, blood transfusion) [1]. 2.1. Vaccines
Disease severity ranges from subclinical to severe hepatitis with
hepatic failure and death [1]. After acute infection, 1–10% of adults The experimental vaccine (HBV-ISS) consisted of 3 mg 1018
and 30–90% of newborns become chronic HBV carriers and some ISS (sequence 5# -TGACTGTGAACGTTCGAGATGA-3# ) in combination
progress to hepatic cirrhosis and hepatocellular carcinoma [2]. The with 20 !g of yeast-derived recombinant HBsAg (manufactured
World Health Organization estimates that over 2 billion people for Dynavax Technologies by Rentschler Biotechnologie GmbH,
have been infected with HBV worldwide and over 350 million peo- Laupheim, Germany) per 0.5 mL dose; a single lot was used for
ple are chronic carriers. Approximately 4.5 million new cases occur the study. The control, comparator vaccine (HBV-Eng) contained
and 620,000 people die of acute or chronic HBV infections each year 20 !g of recombinant HBsAg vaccine per 1.0 mL with 0.5 mg of
[3–6]. aluminum hydroxide adjuvant (Engerix-B® , GlaxoSmithKline, Rix-
An effective vaccine against HBV has been available for 30 ensart, Belgium). To preserve the blind, sterile saline placebo was
years and is recommended for universal vaccination [7]. The first given as a third dose to participants allocated to the HBV-ISS group
generation HBV vaccines involved isolating hepatitis B surface anti- so that both groups received injections at 0, 4, and 24 weeks.
gen (HBsAg) from plasma of chronically infected patients [8]. By
1986, safe and effective recombinant, yeast-derived HBsAg vac- 2.2. Study population
cines were developed [9]. Universal implementation of hepatitis
B vaccine programs led to a dramatic decrease in new cases of Healthy adolescents between 11 and 18 years of age in Canada
HBV infection and a subsequent decrease in hepatocellular car- and adults between 18 and 55 years of age in Canada and Germany
cinoma [10]. For maximum immunogenicity, three doses of the were eligible to participate if they had no history of HBV infec-
alum-adjuvanted vaccine must be given with a minimum inter- tion or immunization and were seronegative for HBsAg, antibody
val of 1 month between the first two doses and at least 4 months to HBsAg (anti-HBsAg), and antibody to hepatitis B core antigen
between the second and third doses [11–13]. Other approved dos- (anti-HBcAg). Individuals were excluded if they were pregnant
ing schedules that achieve adequate rates of seroprotection include or breast-feeding, were unwilling to use effective contraception
a 2, 4, 6 months schedule for infants, an accelerated adult schedule during the study, had an autoimmune disease or any clinically sig-
(0, 1, 2 months with a fourth dose at 12 months), and a two-dose nificant acute or debilitating chronic illness, were at high risk for
schedule (0, 6 months) for adolescents [14–16]. Completion of a recent exposure to hepatitis B, hepatitis C, or human immunode-
vaccination schedule over 6 months can pose problems for ado- ficiency virus, had received blood products or immunoglobulins in
lescents, individuals with predictably poor rates of compliance, the previous 3 months, had sensitivity to any component of any
and individuals who need rapid protection (travelers to high-risk of the vaccines, had ever received an injection of DNA plasmids or
areas, health care workers) [3,14,17,18]. Despite their success, the oligonucleotides, were immunosuppressed or receiving systemic
current licensed vaccines are less immunogenic in older adults corticosteroids, or had received any vaccines in the previous month.
≥40 years of age, males, smokers, and the obese (persons with
body mass index ≥30 kg/m2 ) [19–21]. As well, certain individu- 2.3. Study design and procedures
als respond poorly or not at all to vaccination even after repeated
vaccination courses (so called hyporesponders and nonresponders) The study was a participant-and observer-blinded, random-
[22]. ized, controlled, multicenter study. Participants provided written
Immunostimulatory sequences (ISS) are bacterial DNA or syn- informed consent prior to any study procedure. The study was
thetic oligonucleotides [23,24] that stimulate the innate immune approved by the Research Ethics Boards of all participating institu-
system through toll-like receptor-9 (TLR-9). Activation of the tions in Canada and by a central Research Ethics Board in Germany.
TLR-9 pathway results in the production of interferon gamma Participants were randomly allocated to HBV-ISS or HBV-Eng by
and inducers of interferon gamma (interferon alpha and beta, a center-specific, computer-generated list produced by a contract
interleukin-12, interleukin-18) that drive the adaptive immune research organization. Randomization was in a 3:1 ratio (HBV-
response toward a T-helper cell 1 phenotype [25–35]. 1018 ISS is a ISS:HBV-Eng) with a block size of 4 stratified by age (11–39 years
22-mer phosporothioate oligodeoxyribonucleotide that has broad vs. 40–55 years). Participants randomized to the HBV-Eng group
immunostimulatory activity in vitro and in vivo in multiple species, received three doses at 0, 4, and 24 weeks; participants random-
including humans [36]. 1018 ISS administered with HBsAg was pre- ized to receive HBV-ISS received two doses at 0 and 4 weeks and
viously demonstrated to be immunogenic and well tolerated in placebo at 24 weeks.
healthy adults in a dose-ranging phase 1 study [37] and in young Vaccines were given as intramuscular injections into the del-
adults in a phase 2 study using a 0-, 8-week schedule [38]. The toid muscle with a 1–1½ in. (2 in. in obese participants), 25-gauge
purpose of this study was to compare the tolerability and immuno- needle, alternating the arm with each injection. Participants were
genicity of recombinant HBsAg adjuvanted with 1018 ISS with an monitored by study staff for 30 min after each injection for
2558 S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563

immediate reactions. Participants measured their oral temperature and had any post-baseline safety data. When treatment groups
daily with a supplied digital thermometer and assessed selected were compared based on categorical variables, a contingency table
injection-site reactions (erythema, swelling, and pain) and sys- analysis was used with the Cochran–Mantel–Haenszel technique
temic adverse events daily (headache, fatigue, malaise, and fever); to control for categorical age. Continuous variables (except for age)
all unsolicited adverse events were collected and tabulated by body were tested with an analysis of variance (ANOVA) model contain-
system. Adverse events were either measured (fever, erythema, ing fixed effects for treatment and age. The model for age contained
and swelling) or categorized as Grade 1 (mild, no interference only treatment as a fixed effect. All data analyses were performed
with activity), Grade 2 (moderate, some interference with activ- using Statistical Analysis Systems (SAS Version 8.2 or later, SAS
ity), or Grade 3 (severe, significant interference or preventing Institute, Cary, NC, USA).
daily activity). Erythema and swelling were defined as Grade 0 A sample size of 1176 in the HBV-ISS group and 392 in the HBV-
(<25 mm), Grade 1 (25–50 mm), Grade 2 (51–100 mm), and Grade Eng group (total 1568 participants) was calculated for the primary
3 (>100 mm). Fever was defined as Grade 0 (<38 ◦ C), Grade 1 immunogenicity outcome to have 90% power to demonstrate non-
(≥38 ◦ C and <38.5 ◦ C), Grade 2 (≥38.5 ◦ C and <39.5 ◦ C), and Grade inferiority in SPR following immunization with HBV-ISS compared
3 (≥39.5 ◦ C). Adverse events were recorded by participants in a to HBV-Eng. A total sample size of 2400 (1800 HBV-ISS, 600 HBV-
standardized diary for 7 days postvaccination and were collected Eng) was planned to ensure sufficient safety data on the HBV-ISS
by study personnel at weeks 0 and 4 (time of the first two vac- cohort.
cinations), 12, 24 (time of third vaccination), and 28; telephone
reminders were performed 3 days after each injection.
Blood was collected by venipuncture before and 4 weeks after 3. Results
dose 1 (at the time of dose 2); 4 and 8 weeks post-dose 2 (weeks
8 and 12); and before and 4 weeks post dose 3 (weeks 24 and 3.1. Demographics and participant disposition
28). Anti-HBsAg was measured at each of these time points by
the hepatitis B enhanced chemiluminescence immunoassay (Hep A total of 2910 individuals were screened for participation in the
B ECi, Ortho Clinical Diagnostics, Raritan, NJ, USA); all testing was study between 12 December 2006 and 12 July 2007 (Fig. 1). Of those,
done in a blinded fashion on code-labeled, matched pre- and post- 482 were excluded because of pre-existing serologic markers for
immunization sera. All anti-HBsAg levels were reported as mIU/mL. HBV infection or immunization, failure to return for enrollment, or
Anti-nuclear antibodies and anti-single-stranded and anti-double- withdrawal of consent. A total of 2428 participants were random-
stranded DNA were measured before the first injection and at week ized. Although individuals between 11 and 18 years of age were
28 (4 weeks post dose 3). eligible to be enrolled in Canada, only 13 of the 2428 participants
enrolled were in this age cohort because most individuals of that
2.4. Data analysis and statistical considerations age had already been immunized with a hepatitis B vaccine as part
of universal immunization programs. For this reason, the analysis
The primary outcome for the study was the seroprotection focused only on the 2415 participants 18–55 years of age. The mean
rate (SPR; proportion of participants with an anti-HBsAg level age of participants was 39.9 years in the HBV-ISS group (n = 1809)
≥10 mIU/mL) measured 4 weeks post-dose 3 (week 28) in partici- and 39.8 years in the HBV-Eng group (n = 606) (Table 1). A history
pants who received HBV-Eng on a 0-, 4-, 24-week schedule and 8 of a medical or surgical condition or event was reported by 82.1% of
weeks post-dose 2 (week 12) in participants who received HBV-ISS HBV-ISS participants and 80.4% of HBV-Eng participants; the dis-
on a 0-, 4-week schedule (time of maximum response based on ear- tribution of conditions by organ system was similar between the
lier studies). The hypothesis tested was that the SPR post-dose 2 for two groups.
HBV-ISS would be noninferior to the SPR post-dose 3 for HBV-Eng. A total of 96.6% of randomized participants between 18 and 55
Noninferiority was declared if the lower limit of the 95% confidence years of age completed the study, 96.5% (1746/1809) in the HBV-ISS
interval for the difference in SPR between HBV-ISS and HBV-Eng group and 97.0% (588/606) in the HBV-Eng group. Of the 63 partici-
was above −10%; superiority was declared if the lower limit of the pants who did not complete the study in the HBV-ISS group, loss to
95% confidence interval for the difference in SPR between HBV-ISS follow up (n = 30) and withdrawal of consent (n = 18) were the most
and HBV-Eng was above 0%. Additional outcomes included com- common reasons. Other reasons included noncompliance with the
parisons of SPR 4 weeks after the three-dose HBV-Eng schedule protocol (n = 3), protocol violation (n = 2), pregnancy (n = 6), with-
and the two-dose HBV-ISS schedule, SPR at each visit when speci- drawal by the sponsor because the participant was planning to be
mens were obtained, and geometric mean antibody concentrations out of the country for an extended period (n = 1), and seizure dis-
(GMCs) using the same comparisons as with SPR. order (n = 1); there were two withdrawals related to an adverse
Safety outcomes (solicited and unsolicited adverse events) event. There were 18 participants in the HBV-Eng group who did
and laboratory measures of autoimmunity (antinuclear antibod- not complete the study; reasons included loss to follow up (n = 10),
ies [ANA] and anti-double-stranded DNA [anti-ds-DNA]) were also non-compliance (n = 2), withdrawal of consent (n = 2), pregnancy
compared. Adverse events were tabulated by day and severity; the (n = 1), withdrawal by investigator (n = 1), and an adverse event
maximum severity reported was used for each time period. The (n = 2).
proportion of participants having an adverse event was calculated
by vaccine group, observation period, and severity.
The analysis population for immunogenicity outcomes was the 3.2. Adverse events
per-protocol population defined as participants who met the eligi-
bility criteria, did not violate the protocol in a substantial manner, The safety analysis was performed on all 2415 enrolled and ran-
received all protocol-specified study injections, had anti-HBsAg domized participants. Pain at the injection site was reported by
measurements and all injections within the specified day ranges, 35.3–38.6% of HBV-ISS recipients after one of the two doses and
and had an anti-HBsAg measurement at the time of their primary 21.6–33.7% of HBV-Eng recipients after one of the three doses; pain
endpoint. The intent-to-treat population was defined as partici- was reported in progressively fewer vaccinees with subsequent
pants who received at least one study injection and had at least one doses after either vaccine (Table 2). Severe pain was reported by
post-baseline anti-HBsAg level. The safety population was defined 1.7% or fewer vaccine recipients after any dose. Injection-site ery-
as enrolled participants who received at least one study injection thema and swelling was infrequent, reported by fewer than 5% of
S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563 2559

Fig. 1. Participant disposition.

either vaccine group; severe erythema or swelling was reported by solicited adverse events were no different than those reported by
0.3% or fewer recipients of either vaccine at any dose. the HBV-ISS group after their saline placebo injection (dose 3).
There were no differences in systemic adverse events between Severe systemic adverse events were infrequent; 1.2% or fewer par-
the vaccine groups (Table 2). After the second dose, rates of these ticipants reported severe fatigue, 1.3% or fewer severe headache,

Table 1
Summary of participant demographics and baseline characteristics.

Characteristic HBV-ISS (n = 1809) HBV-Eng (n = 606) Total (n = 2415)

Age, years
Mean (range) 39.9 (18–55) 39.8 (18–55) 39.9 (18–55)
18–39, n (%) 818 (45.2) 275 (45.4) 1093 (45.3)
40–55, n (%) 991 (54.8) 331 (54.6) 1322 (54.7)
Gender, n (%)
Male 852 (47.1) 262 (43.2) 1114 (46.1)
Female 957 (52.9) 344 (56.8) 1301 (53.9)
Race, n (%)
White 1690 (93.4) 556 (91.7) 2246 (93.0)
Black 39 (2.2) 20 (3.3) 59 (2.4)
Asian 43 (2.4) 22 (3.6) 65 (2.7)
First Nations or Inuit 16 (0.9) 3 (0.5) 19 (0.8)
Native Hawaiian or Pacific Islander 1 (0.1) 0 (0.0) 1 (0.0)
Other 20 (1.1) 5 (0.8) 25 (1.0)
Weight (kg)
n 1806 605 2411
Mean (range) 80.30 (43.0–173.0) 80.79 (39.3–179.0) 80.42 (39.3–179.0)
Height (cm)
n 1807 605 2412
Mean (range) 171.00 (139.7–228.6) 170.76 (147.0–196.0) 170.94 (139.7–228.6)
Body mass index
n 1806 605 2411
Mean (range) 27.41 (15.0–58.1) 27.62 (16.4–63.2) 27.46 (15.0–63.2)
Smoker, n (%)
Yes 654 (36.2) 224 (37.0) 878 (36.4)
No 1155 (63.8) 382 (63.0) 1537 (63.6)
2560 S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563

Table 2
Summary of solicited reactions within 7 days of each injection.

Reaction First injection Second injection Third injection

0 weeks 4 weeks 24 weeks

HBV-ISS HBV-Eng HBV-ISS HBV-Eng Placebo HBV-Eng


n = 1809 % (95% CI) n = 606 % (95% CI) n = 1797 % (95% CI) n = 604 % (95% CI) n = 1768 % (95% CI) n = 598 % (95% CI)

Injection site 40.3 (38.0–42.6) 34.2 (30.4–38.1) 36.2 (34.0–38.5) 25.5 (22.1–29.2) 7.6 (6.4–9.0) 21.9 (18.7–25.4)
Pain 38.6 (36.3–40.9) 33.7 (29.9–37.6) 35.3 (33.1–37.6) 25.2 (21.8–28.8) 7.2 (6.1–8.5) 21.6 (18.3–25.1)
Milda 34.2 (32.0–36.4) 29.4 (25.8–33.2) 28.0 (26.0–30.2) 20.7 (17.5–24.1) 5.5 (4.5–6.7) 17.1 (14.1–20.3)
Moderateb 4.1 (3.3–5.2) 4.1 (2.7–6.0) 6.4 (5.3–7.6) 3.6 (2.3–5.5) 0.5 (0.2–1.0) 2.8 (1.7–4.5)
Severec 0.3 (0.1–0.6) 0.2 (0.0–0.9) 0.9 (0.5–1.4) 0.8 (0.7–1.9) 1.2 (0.8–1.9) 1.7 (0.8–3.1)
Redness 4.1 (3.3–5.2) 0.5 (0.1–1.4) 2.9 (2.2–3.8) 1.0 (0.4–2.2) 0.3 (0.1–0.7) 0.7 (0.2–1.7)
≥2.5 cm to ≤5.0 cm 4.0 (3.1–5.0) 0.5 (0.1–1.4) 2.6 (1.9–3.5) 0.7 (0.2–1.7) 0.3 (0.1–0.7) 0.5 (0.1–1.4)
≥5.1 cm to ≤10.0 cm 0.2 (0.0–0.5) 0.0 (0.0–0.6) 0.2 (0.1–0.6) 0.0 (0.0–0.6) 0.0 (0.0–0.2) 0.0 (0.0–0.6)
>10.0 cm 0.0 (0.0–0.2) 0.0 (0.0–0.6) 0.1 (0.0–0.4) 0.3 (0.0–1.2) 0.0 (0.0–0.2) 0.2 (0.0–0.9)
Swelling 2.3 (1.6–3.1) 0.7 (0.2–1.7) 1.5 (1.0–2.2) 0.5 (0.1–1.4) 0.2 (0.0–0.5) 0.5 (0.1–1.5)
≥2.5 cm to ≤5.0 cm 2.2 (1.5–2.9) 0.7 (0.2–1.7) 1.2 (0.7–1.8) 0.2 (0.0–0.9) 0.1 (0.0–0.4) 0.3 (0.0–1.2)
≥5.1 cm to ≤10.0 cm 0.1 (0.0–0.4) 0.0 (0.0–0.6) 0.2 (0.1–0.6) 0.0 (0.0–0.6) 0.1 (0.0–0.3) 0.0 (0.0–0.6)
10.0 cm 0.0 (0.0–0.2) 0.0 (0.0–0.6) 0.1 (0.0–0.4) 0.3 (0.0–1.2) 0.0 (0.0–0.2) 0.2 (0.0–0.9)
Systemic 26.9 (24.9–29.0) 29.2 (25.6–33.0) 21.3 (19.4–23.2) 18.5 (15.5–21.9) 12.8 (11.31–14.5) 14.5 (11.8–17.6)
Fatigue 17.4 (15.7–19.2) 16.7 (13.8–19.9) 13.8 (12.2–15.4) 12.1 (9.6–15.0) 7.9 (6.6–9.2) 10.0 (7.7–12.7)
Mild 11.9 (10.4–13.5) 10.4 (8.1–13.1) 9.4 (8.1–10.8) 7.0 (5.1–9.3) 4.7 (3.8–5.8) 5.0 (3.4–7.1)
Moderate 4.4 (3.5–5.4) 5.6 (3.9–7.8) 3.7 (2.9–4.6) 4.1 (2.7–6.0) 2.6 (1.9–3.4) 4.3 (2.9–6.3)
Severe 1.2 (0.7–1.8) 0.7 (0.2–1.7) 0.7 (0.4–1.2) 1.0 (0.4–2.1) 0.6 (0.3–1.0) 0.7 (0.2–1.7)
Headache 16.8 (15.1–18.6) 19.3 (16.2–22.7) 12.7 (11.2–14.4) 12.4 (9.9–15.3) 9.0 (7.7–10.4) 9.5 (7.3–12.2)
Mild 10.6 (9.2–12.1) 11.6 (9.1–14.4) 8.1 (6.9–9.4) 7.8 (5.8–10.2) 5.2 (4.2–6.3) 5.4 (3.7–7.5)
Moderate 5.2 (4.2–6.3) 6.4 (4.6–8.7) 3.7 (2.9–4.7) 4.0 (2.6–5.8) 3.0 (2.3–3.9) 3.5 (2.2–5.3)
Severe 1.0 (0.6–1.6) 1.3 (0.6–2.6) 0.9 (0.6–1.5) 0.7 (0.2–1.7) 0.8 (0.4–1.3) 0.7 (0.2–1.7)
Malaise 9.2 (7.9–10.6) 8.9 (6.8–11.5) 7.6 (6.4–8.9) 6.4 (4.6–8.7) 4.3 (3.4–5.4) 6.4 (4.5–8.6)
Mild 5.5 (4.5–6.7) 5.3 (3.6–7.4) 5.0 (4.0–6.1) 4.3 (2.8–6.2) 2.3 (1.7–3.1) 3.2 (1.9–4.9)
Moderate 3.1 (2.3–4.0) 2.8 (1.6–4.4) 2.0 (1.4–2.8) 1.7 (0.8–3.0) 1.4 (0.9–2.1) 2.7 (1.5–4.3)
Severe 0.6 (0.3–1.0) 0.8 (0.3–1.9) 0.7 (0.3–1.2) 0.5 (0.1–1.4) 0.6 (0.3–1.0) 0.5 (0.1–1.5)

n = 1783 n = 597 n = 1763 n = 591 n = 1675 n = 561


Fever 1.1 (0.6–1.7) 1.8 (0.9–3.3) 1.4 (0.9–2.1) 1.9 (0.9–3.3) 1.3 (0.8–2.0) 1.8 (0.9–3.3)
≥38.0 ◦ C to <38.5 ◦ C 0.6 (0.3–1.0) 1.0 (0.4–2.2) 0.5 (0.2–1.0) 1.2 (0.5–2.4) 0.8 (0.4–1.3) 0.9 (0.3–2.1)
≥38.5 ◦ C to <39.5 ◦ C 0.4 (0.2–0.8) 0.7 (0.2–1.7) 0.8 (0.4–1.3) 0.3 (0.0–1.2) 0.5 (0.2–0.9) 0.7 (0.2–1.8)
≥39.5 ◦ C 0.1 (0.0–0.4) 0.2 (0.0–0.9) 0.1 (0.0–0.4) 0.3 (0.0–1.2) 0.1 (0.0–0.3) 0.2 (0.0–1.0)
a
No interference with activity.
b
Some interference with activity.
c
Significant interference, prevents daily activity.

and 0.8% or fewer severe malaise. Only one participant (HBV-Eng) discontinued after developing a pulmonary embolism 42 days after
reported a fever of 40 ◦ C or higher. the second dose of HBV-ISS. There were also two discontinuations
Unsolicited adverse events were reported by 60.5% of HBV- in the HBV-Eng group: a 49-year-old woman discontinued after
ISS recipients and 62.0% of HBV-Eng recipients (data not shown). increasing ocular pressure and blurred vision related to glaucoma
Intercurrent infections were the most commonly reported adverse 4 days after the first dose and a 35-year-old woman discontinued
event, reported by 32.5% of HBV-ISS recipients and 30.7% of HBV- because of a flare of arthritis in her hands 9 days after the second
Eng recipients. Musculoskeletal complaints were reported by 14.8% dose. No deaths occurred during the study.
and 14.0% of HBV-ISS and HBV-Eng recipients, respectively. Other There were no significant changes in ANA and anti-ds-DNA,
common reports included gastrointestinal complaints (11.0% vs. two markers of autoimmunity, in recipients of either vaccine. ANA
14.2%), respiratory complaints (10.2% vs. 9.1%), and nervous system titers were considered “normal” at levels ≤1:160 IU/mL. At base-
disorders (dizziness, headache; 14.0% vs. 13.9%); there were no dif- line, 89.3% (1616/1804) of participants in the HBV-ISS group and
ferences between the vaccine groups for any unsolicited adverse 89.3% (541/605) of participants in the HBV-Eng group had nor-
event. mal ANA titers. Postvaccination, at 28 weeks, 91.9% and 91.4%
Overall, 41 participants reported 47 serious adverse events of participants in the HBV-ISS and HBV-Eng groups, respectively,
including 28 (1.5%) participants in the HBV-ISS group (33 events) had normal ANA titers. From baseline to week 28, 31 (1.7%) par-
and 13 (2.1%) participants in the HBV-Eng group (14 events). ticipants in the HBV-ISS group and 8 (1.3%) participants in the
Pulmonary embolism (3 participants) and jaw fracture, breast can- HBV-Eng group had a twofold ANA increase, 10 (0.6%) and 6
cer, depression, and pneumothorax (2 participants each) were (1.0%), respectively, had a fourfold increase; and 1 (0.1%) and 0
reported by more than one participant. An autoimmune illness (0%), respectively, had an eightfold increase. At baseline, 6 (0.3%)
was reported by one recipient in the HBV-ISS group (Wegener’s of 1799 participants in the HBV-ISS group and 3 (0.5%) of 602
granulomatosis 171 days post second dose) and one recipient in participants in the HBV-Eng group had positive anti-ds-DNA
the HBV-Eng group (systemic vasculitis 127 days after the second results. At week 28, 10 (0.6%) and 3 (0.5%) participants, respectively,
dose). Four participants discontinued participation as a result of an had positive anti-ds-DNA results. From baseline, 9 (0.5%) of HBV-
adverse event. A 35-year-old woman discontinued after developing ISS participants and 3 (0.5%) of HBV-Eng participants went from a
Guillain–Barré Syndrome 111 days after the second dose of HBV- negative to positive result, and 5 (0.3%) and 3 (0.5%), respectively,
ISS and 5 days after receipt of influenza vaccine. A 25-year-old male went from positive to negative.
S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563 2561

Fig. 3. Geometric mean antibody concentrations after each dose of HBV-ISS (closed
Fig. 2. Proportion of participants seroprotected (anti-HBsAg ≥ 10 mIU/mL) after circles; n = 1548–1557) or HBV-Eng (closed squares; n = 531–533). Error bars repre-
each dose of HBV-ISS (closed circles; n = 1548–1557) or HBV-Eng (closed squares; sent 95% confidence intervals.
n = 531–533). Error bars represent 95% confidence intervals.

superiority was demonstrated for the HBV-ISS group compared to


3.3. Antibody response the HBV-Eng group at week 8 (50.2% vs. 9.0%), week 12 (63.7% vs.
6.6%), week 24 (83.5% vs. 9.8%), and week 28 (82.2% vs. 65.5%). Anti-
Four weeks after the first vaccination, 23.6% (95% CI 21.5–25.8) body levels ≥100 mIU/mL were superior in the HBV-ISS group for
of HBV-ISS recipients and 3.95% (2.30–5.61) of HBV-Eng recipi- both the younger and older cohorts (data not shown).
ents had seroprotective antibody levels of ≥10 mIU/mL (Fig. 2).
At 8 weeks (4 weeks post-dose 2), 88.5% (86.9–90.1) of HBV-ISS 4. Discussion
recipients and 26.4% (22.6–30.1) of HBV-Eng recipients had sero-
protective levels. At 24 weeks (20 weeks post-dose 2 and pre-dose This study was designed to test the primary hypothesis that the
3 for the HBV-Eng group), 98.3% (97.6–98.9) of HBV-ISS participants SPR 8 weeks after two doses of the HBV-ISS vaccine would be nonin-
and 32.4% (28.4–36.4) of HBV-Eng participants had seroprotective ferior to the SPR 4 weeks after three doses of the licensed HBV-Eng.
levels. At week 28 (24 weeks post-dose 2 and 4 weeks post-saline The established noninferiority criterion was met in the study at the
placebo for the HBV-ISS recipients and 4 weeks post-dose 3 for the primary time point of maximal response of the HBV-ISS as well
HBV-Eng recipients), 97.9% (97.2–98.7) of the HBV-ISS recipients as at all other times when antibody levels were measured. More-
and 81.1% (77.7–84.4) of the HBV-Eng recipients had seroprotec- over, the SPR of HBV-ISS was statistically superior at all time points
tive levels. For the primary immunogenicity outcome, the SPR at compared to the licensed HBV-Eng. The magnitude of the antibody
week 12 (8 weeks postvaccination) in the HBV-ISS group (95.1%; response to HBV-ISS, as measured by GMC, was also noninferior to
94.0–96.1) was noninferior and superior to that of the HBV-Eng HBV-Eng at all time periods assessed. Statistical superiority of the
group at week 28 (4 weeks post-dose 3) (difference of HBV-ISS GMC after HBV-ISS compared to HBV-Eng was also demonstrated at
minus HBV-Eng = 13.9%; 10.6–17.6). Noninferiority and superior- all time periods except for week 28 (24 weeks post-dose 2 of HBV-
ity of SPR were also demonstrated at all other serology time points. ISS and 4 weeks post-dose 3 of HBV-Eng). Of note, the superiority
Higher SPRs were observed in younger participants (aged 18–39 of the immune response to HBV-ISS was observed for both younger
years) and older participants (aged 40–55 years) injected with (<40 years of age) and older (≥40 years of age) participants. Both
HBV-ISS at each time point compared to participants injected with vaccines were well tolerated by participants. Mild injection-site
HBV-Eng (Table 3). Higher SPRs were achieved by younger recipi- adverse events were more commonly reported by HBV-ISS recip-
ents compared to older recipients of both vaccines. ients than HBV-Eng recipients but systemic adverse events were
The magnitude of the immune response was also greater after reported with equal frequency after either vaccine.
the first and second doses of HBV-ISS compared to HBV-Eng (Fig. 3). The findings of this study support the results of previous stud-
In HBV-Eng recipients, a substantial rise in GMC did not occur until ies with the HBV-ISS vaccine. Seroprotection rates as high as 87.5%
after dose 3, when the GMC rose from 7.19 (6.31–8.20) mIU/mL after the first dose and 100% after the second dose given 2 months
to 348.2 (265.9–455.9) mIU/mL. In contrast, in the HBV-ISS group, apart were found in a phase 1 dose-ranging study [37]. In a phase
the GMC rose from 5.5 (5.13–5.88) mIU/mL 4 weeks post-dose 2 comparison of two doses of HBV-ISS given at weeks 0 and 8 with
1 to 81.5 (75.1–88.5) mIU/mL 4 weeks post-dose 2, to 136.9 three doses of a licensed hepatitis B vaccine given at weeks 0, 8,
(127.5–146.9) mIU/mL 8 weeks post-dose 2. The GMC continued to and 24 [38], 79% of HBV-ISS and 12% of HBV-Eng recipients had a
rise in this group and at week 24 (20 weeks post-dose 2) was 342.5 protective antibody response 4 weeks post dose 1. By 1 week post
(320.2–366.5) mIU/mL. Four weeks later, the GMC in the HBV-ISS dose 2, 100% of HBV-ISS and 18% of HBV-Eng recipients had pro-
group (24 weeks post-dose 2) was 320.0 (298.2–343.3) mIU/mL, tective levels. Rates of seroprotection after HBV-Eng in the current
similar to the GMC in the HBV-Eng group 4 weeks post-dose 3. study and the previous phase 2 study are similar to those reported
GMCs of participants in the younger age stratum were higher in other studies of HBV-Eng [14].
than those in the older age group for both HBV-ISS and HBV-Eng The rapid induction of protective antibody levels after a two-
(Table 3). For all time points, the younger recipients of the HBV- dose schedule at 0 and 4 weeks has a number of advantages,
ISS vaccine had GMCs approximately twofold higher than the older particularly where rapid protection is required or where follow-up
participants. Lower responses for the older participants were also for subsequent doses cannot be guaranteed. Three-dose hepatitis B
observed after the first two doses of HBV-Eng although the results immunization schedules spread over 6 months are problematic for
were less dramatic given the low GMCs of both age strata. How- adolescents who are less frequent users of the health care system
ever, post-dose 3 of HBV-Eng, younger participants achieved a GMC [14]; a licensed two-dose adolescent schedule decreases the num-
threefold higher than the older cohort. ber of injections but still requires that the doses be separated by 6
The magnitude of the immune response was also examined by months. For individuals with high-risk behaviors, rapid induction
comparing the proportion of participants who exceeded a thresh- of immunity diminishes the period of time the individual remains
old antibody level of ≥100 mIU/mL at each time point. Statistical at risk for acquisition of hepatitis B [18]. Two-dose programs
2562 S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563

Table 3
Summary of seroprotection and geometric mean concentrations (GMC) by week and age stratification.

Age stratification Week Seroprotection GMC

HBV-ISS % (95% CI) HBV-Eng % (95% CI) HBV-ISS % (95% CI) HBV-Eng % (95% CI)

18–39 0
4 30.5 (27.0, 34.0) 5.80 (2.74, 8.87) 6.93 (6.18, 7.78) 3.16 (2.81, 3.55)
8 94.8 (93.1, 96.5) 33.0 (26.9, 39.2) 125.1 (112.1, 139.5) 8.43 (6.63, 10.7)
12 98.7 (97.8, 99.5) 30.1 (24.1, 36.1) 212.0 (193.7, 232.1) 7.24 (5.81, 9.03)
24 99.6 (99.1, 100.0) 39.7 (33.3, 46.1) 512.0 (470.2, 557.6) 9.63 (7.66, 12.1)
28 99.3 (98.6, 99.9) 89.4 (85.4, 93.4) 475.7 (434.4, 520.9) 655.9 (443.3, 970.4)

40–55 0
4 18.5 (15.9, 21.0) 2.61 (0.82, 4.39) 4.60 (4.25, 4.99) 2.77 (2.59, 2.96)
8 83.7 (81.2, 86.1) 21.5 (16.9, 26.1) 58.6 (52.3, 65.7) 5.29 (4.51, 6.20)
12 92.3 (90.5, 94.0) 16.9 (12.7, 21.1) 97.8 (88.6, 108.0) 4.46 (3.91, 5.09)
24 97.3 (96.2, 98.3) 27.0 (22.1, 32.0) 251.3 (228.5, 276.4) 5.81 (5.00, 6.76)
28 96.9 (95.8, 98.1) 74.9 (70.1, 79.8) 236.1 (213.9, 260.5) 218.4 (152.2, 313.6)

would be less expensive to deliver than those requiring three doses may be useful to demonstrate further the full range of potential
in terms of vaccine cost and human resources. Additionally, the benefits.
improved immunogenicity in the older cohort raises the potential
for improved seroprotection amongst other hypo-responders and
nonresponders [21,22]. Finally, noncompletion of travel-related Acknowledgments
vaccination series is common because of insufficient planning prior
to travel; rapid protection after a two-dose course completed in 4 At the Canadian Center for Vaccinology, we thank Petra Rykers,
weeks would facilitate protection prior to travel. Donna MacKinnon-Cameron, Christina Wang, and Bruce Smith for
A number of other vaccines and schedules with improved undertaking the independent analysis of the data and Paula Ogg
immunogenicity have also recently become available. Three-dose for preparing the tables and figures for publication. We thank the
schedules of licensed vaccine given at 0, 1, and 2 months have been nurses and research assistants at all of the study sites for their
advocated to achieve more rapid responses for travelers and health careful attention to detail.
care workers with a fourth dose given at 1 year but result in lower
GMC [4]. A hepatitis B vaccine with a novel adjuvant (AS04) com-
prising aluminum phosphate and 3-O-desacyl-4# -monophosphoryl References
lipid A (MPA) also provides more rapid and greater levels of protec-
tion against HBV after a four-dose regimen than the licensed alum [1] Lee WM. Hepatitis B virus infection. N Engl J Med 1997;337:1733–45.
[2] McMahon BJ, Alward WL, Hall DB, Heyward WL, Bender TR, Francis DP,
adjuvanted vaccine in renal failure and dialysis patients but is not et al. Acute hepatitis B virus infection: relation of age to the clinical expres-
licensed for use in healthy adults [39,40]. sion of disease and subsequent development of the carrier state. J Infect Dis
One limitation of the current study was our inability to enroll 1985;151:599–603.
[3] Kane MA. Global status of hepatitis B immunisation. Lancet 1996;348:696.
sufficient adolescent participants. Although the intention was to
[4] Zanetti AR, Van Damme P, Shouval D. The global impact of vaccination against
enroll adolescents and adults at study sites in Canada, the existence hepatitis B: a historical overview. Vaccine 2008;26:6266–73.
of universal, school-based, adolescent HBV vaccination programs [5] Custer B, Sullivan SD, Hazlet TK, Iloeje U, Veenstra DL, Kowdley KV. Global
with high coverage rates in Canada for several years prior to the epidemiology of hepatitis B virus. J Clin Gastroenterol 2004;38:S158–68.
[6] Goldstein ST, Zhou F, Hadler SC, Bell BP, Mast EE, Margolis HS. A mathematical
study left very few eligible adolescents. Thus, only 13 adolescents model to estimate global hepatitis B disease burden and vaccination impact.
were enrolled in the study and the analysis was undertaken with Int J Epidemiol 2005;34:1329–39.
participants from 18 to 55 years of age. This limits the general- [7] World Health Organization. Hepatitis B vaccines. Wkly Epidemiol Rec
2009;84:405–20.
izability of the study to adolescents, and additional studies with [8] Szmuness W, Stevens CE, Harley EJ, Zang EA, Oleszko WR, William DC, et al.
pre-adolescents and adolescents will likely be required. Given the Hepatitis B vaccine: demonstration of efficacy in a controlled clinical trial in a
improved immunogenicity to all hepatitis B vaccines including high-risk population in the United States. N Engl J Med 1980;303:833–41.
[9] McAleer WJ, Buynack EB, Maigetter RZ, Wampler DE, Miller WJ, Hilleman MR.
HBV-ISS in younger individuals, it is likely that adolescents will Human hepatitis B vaccine from recombinant yeast. Nature 1984;307:178–80.
have a robust immune response to HBV-ISS. However, as HBV-ISS [10] Chen DS. Hepatitis B vaccination: the key towards elimination and eradication
contains a novel adjuvant, studies in this age cohort to demonstrate of hepatitis B. J Hepatol 2009;50:805–16.
[11] André FE. Summary of safety and efficacy data on a yeast-derived hepatitis B
safety, tolerability, and duration of protection are needed. Although vaccine. Am J Med 1989;87(Suppl. 3A):14S–20S.
hepatitis B vaccines are used widely in infants and younger chil- [12] Mahoney FJ, Kane M. Hepatitis B vaccine. In: Plotkin SA, Orenstein WA, editors.
dren, studies with HBV-ISS in these age cohorts will likely await Vaccines. Philadelphia: Saunders; 1999. p. 158–82.
[13] Heineman TC, Clements-Mann ML, Poland GA, Jacobson RM, Izu AE, Sakamoto
the results of longer term postlicensure safety studies in adults.
D, et al. A randomized, controlled study in adults of the immunogenicity of a
In summary, HBV-ISS was welltolerated and immunogenic in novel hepatitis B vaccine containing MF59 adjuvant. Vaccine 1999;17:2769–78.
healthy adults from 18 to 55 years of age and demonstrated [14] Cassidy WM, Watson B, Ioli VA, Williams K, Bird S, West DJ. A randomized
superior immunogenicity and earlier onset of protection with a trial of alternative two- and three-dose hepatitis B vaccination regimens in
adolescents: antibody responses, safety, and immunologic memory. Pediatrics
two-dose schedule over 4 weeks compared to three doses of a 2001;107:626–31.
licensed, alum-adjuvanted hepatitis B vaccine. Comparison of the [15] Ambrosch F, Wiedermann G, Kundi M, Leroux-Roels G, Desombere I, Garcon N,
antibody responses 1 year after immunization and beyond with et al. A hepatitis B vaccine formulated with a novel adjuvant system. Vaccine
2000;18:2095–101.
the currently licensed alum-adjuvanted vaccines will help to fur- [16] André F, Safary A. Summary of clinical findings on Engerix-B, a genetically
ther characterize the long-term benefits of this novel hepatitis engineered yeast-derived hepatitis B vaccine. Post Grad Med J 1987;63:169–77.
B vaccine. Additional studies in adolescents, immunocompro- [17] Advisory Committee on Immunization Practices. Hepatitis B virus: a compre-
hensive strategy for eliminating transmission in the United States through
mised hosts, dialysis and renal failure patients, hyporesponders universal childhood vaccination: recommendations of the Immunization Prac-
and nonresponders, and eventually young infants and children tices Advisory Committee (ACIP). Morbid Mortal Wkly Rep 1991;40:1–19.
S.A. Halperin et al. / Vaccine 30 (2012) 2556–2563 2563

[18] Centers for Disease Control and Prevention. Hepatitis B vaccination among [30] Krieg AM, Matson S, Fisher E. Oligodeoxynucleotide modifications determine
high-risk adolescents and adults—San Diego, California, 1998–2001. Morb Mor- the magnitude of B cell stimulation by CpG motifs. Antisense Nucleic Acid Drug
tal Wkly Rep 2002;51:618–21. Dev 1996;6:133–9.
[19] Denis F, Mounier M, Hessel L, Michel JP, Gualde N, Dubois F, et al. Hepatitis B [31] Krieg AM, Yi AK, Matson S, Waldschmidt TJ, Bishop GA, Teasdale R, et al. CpG
vaccination in the elderly. J Infect Dis 1984;149:1019. motifs in bacterial DNA trigger direct B-cell activation. Nature 1995;374:546–9.
[20] McLean AA, Hilleman MR, McAleer WJ, Buynak EB. Summary of worldwide [32] Liang H, Nishioka Y, Reich CF, Pisetsky DS, Lipsky PE. Activation of human B cells
experience with H-B-Vax® (B, MSD). J Infect 1983;7:95–104. by phosphorothioate oligodeoxynucleotides. J Clin Invest 1996;98:1119–29.
[21] Weber DJ, Rutala WA, Samsa GP, Santimaw JE, Lemon SM. Obesity as a predic- [33] Hartmann G, Weiner GJ, Krieg AM. CpG DNA: a potent signal for growth,
tor of poor antibody response to hepatitis B plasma vaccine. J Am Med Assoc activation, and maturation of human dendritic cells. Proc Natl Acad Sci USA
1985;254:3187–9. 1999;96:9305–10.
[22] Kubba AK, Taylor P, Graneek B, Strobel S. Non-responders to hepatitis B vacci- [34] Martin-Orozco E, Kobayashi H, Van Uden J, Nguyen MD, Kornbluth RS, Raz
nation: a review. Commun Dis Public Health 2003;6:106–12. E. Enhancement of antigen-presenting cell surface molecules involved in
[23] Pisetsky DS. Immune activation by bacterial DNA: a new genetic code. Immu- cognate interactions by immunostimulatory DNA sequences. Int Immunol
nity 1996;5:303–10. 1999;11:1111–8.
[24] Bird AP. CpG-rich islands and the function of DNA methylation. Nature [35] Roman M, Martin-Orozco E, Goodman JS, Nguyen MD, Sato Y, Ronaghy A, et al.
1986;321:209–13. Immunostimulatory DNA sequences function as T helper-1-promoting adju-
[25] Yamamoto S, Yamamoto T, Katakoa T, Kuramoto E, Yano O, Takunaga T. vants. Nat Med 1997;3:849–54.
Unique palindromic sequences in synthetic oligonucleotides are required [36] Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to
to induce IFN and augment IFN-mediated natural killer activity. J Immunol work. Immunity 2010;33:492–503.
1992;148:4072–6. [37] Halperin SA, Van Nest G, Smith B, Abtahi S, Whiley H, Eiden JJ. A phase 1 study of
[26] Yamamoto S, Yamamoto T, Shimada S, Kuramoto E, Yano O, Kataoka T, et al. the safety and immunogenicity or recombinant hepatitis B surface antigen co-
DNA from bacteria, but not from vertebrates, induces interferons, activates administered with an immunostimulatory phosphorothioate oligonucleotide
natural killer cells and inhibits tumor growth. Microbiol Immunol 1992;36: adjuvant. Vaccine 2003;21:2461–7.
983–7. [38] Halperin SA, Dobson S, McNeil S, Langley JM, Smith B, McCall-Sani R, et al.
[27] Sonehara K, Saito H, Kuramoto E, Yamamoto S, Yamamoto T, Toku- Comparison of the safety and immunogenicity of hepatitis B virus sur-
naga T. Hexamer palindromic oligonucleotides with 5# -CG-3# motif(s) face antigen co-administered with an immunostimulatory phosphorothioate
induce production of interferon. J Interferon Cytokine Res 1996;16: oligonucleotide and a licensed hepatitis B vaccine in healthy young adults.
799–803. Vaccine 2006;24:20–6.
[28] Halpern MD, Kurlander RJ, Pisetsky DS. Bacterial DNA induces murine [39] Tong NK, Beran J, Kee SA, Miguel JL, Sánchez C, Bayas JM, et al. Immunogenic-
interferon-gamma production by stimulation of interleukin-12 and tumor ity and safety of an adjuvanted hepatitis B vaccine in pre-hemodialysis and
necrosis factor-alpha. Cell Immunol 1996;167:72–8. hemodialysis patients. Kidney Int 2005;68:2298–303.
[29] Klinman DM, Yi AK, Beaucage SL, Conover J, Krieg AM. CpG motifs present in [40] Beran J. Safety and immunogenicity of a new hepatitis B vaccine for the pro-
bacteria DNA rapidly induce lymphocytes to secrete interleukin 6, interleukin tection of patients with renal insufficiency including pre-haemodialysis and
12, and interferon gamma. Proc Natl Acad Sci USA 1996;93:2879–83. haemodialysis patients. Expert Opin Biol Ther 2008;8:235–47.

Anda mungkin juga menyukai