You are on page 1of 11

Efficacy and immunogenicity of influenza vaccine in

HIV-infected children: a randomized, double-blind,


placebo controlled trial
Shabir A. Madhia,b,c, Sylvia Dittmerd, Locadiah Kuwandab,c,
Marietjie Ventera, Haseena Cassimd, Erica Lazarusd, Teena Thomasd,
Afaaf Libertyd, Florette Treurnicha, Clare L. Cutlandb,c,
Adriana Weinberge and Avy Violarid
Downloaded from http://journals.lww.com/aidsonline by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 07/22/2021

Background: HIV-infected children are at heightened risk for severe influenza illness;
however, there is no study on the efficacy or effectiveness of influenza vaccine in these
children. We evaluated the safety, immunogenicity, and efficacy of nonadjuvanted,
trivalent inactivated influenza vaccine (TIV) against confirmed seasonal influenza virus
illness in HIV-infected children.
Methods: A double-blind, placebo-controlled trial was undertaken in Johannesburg in
2009. Four hundred and ten children were randomized to two doses of TIV or placebo 1
month apart. Nasopharyngeal aspirates obtained at respiratory illness visits were tested
by influenza-specific reverse transcriptase-PCR (RT-PCR). Vaccine immunogenicity
was evaluated by hemagglutinin inhibition (HAI) assay. Influenza isolates were
sequenced and evaluated in maximum likelihood phylogenetic analysis.
Results: Overall, the median age of participants was 23.8 months and their median
CD4% was 33.5. Ninety-two percent of enrolees were on antiretroviral therapy. Among
children receiving both doses of vaccine/placebo, confirmed seasonal influenza illness
occurred in 13 (all H3N2) of 205 TIV recipients and 17 (15 H3N2 and two influenza B)
of 200 placebo recipients with vaccine efficacy of 17.7% (95% confidence interval
<0–62.4%). The proportion of TIV recipients who seroconverted after second dose
against vaccine strains of H1N1, H3N2, and influenza B were 47.5, 50.0, and 40.0%,
compared to 4.7, 11.6, and 0%, respectively among placebo recipients. There were no
TIV-related serious adverse events. Sequence analysis of wild-type H3N2 strains
indicated drift from the H3N2 vaccine strain.
Conclusion: Poor immunogenicity of TIV, coupled with drift of circulating H3N2
wild-type compared to vaccine strain, may explain the lack of efficacy of TIV in
young HIV-infected children. Alternate TIV vaccine schedules or formulations warrant
evaluation for efficacy in HIV-infected children.
ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins

AIDS 2013, 27:369–379

Keywords: children, efficacy, HIV, immunogenicity, influenza vaccine,


pneumonia, safety

a
National Institute of Communicable Diseases, Centre for Respiratory and Meningitis Diseases, Sandringham, bDepartment of
Science and Technology/National Research Foundation, Vaccine Preventable Diseases, cMedical Research Council: Respiratory
& Meningeal Pathogens Research Unit, dPerinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South
Africa, and eDepartment of Pediatrics, Medicine and Pathology, University of Colorado, Anschutz Medical Campus, Aurora,
Colorado, USA.
Correspondence to Shabir A. Madhi, National Institute for Communicable Diseases, National Health Laboratory Service, 1
Modderfontein Road, Sandringham, 2131 Johannesburg, Gauteng, South Africa.
Tel: +27 113866137; e-mail: shabirm@nicd.ac.za
Received: 16 August 2012; revised: 21 September 2012; accepted: 24 September 2012.

DOI:10.1097/QAD.0b013e32835ab5b2

ISSN 0269-9370 Q 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins 369
Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
370 AIDS 2013, Vol 27 No 3

Background (0.5 ml if 36 months or 0.25 ml if younger) or equal


volume of matching placebo (0.9% sterile saline) 1 month
Antiretroviral therapy (ART)-naive HIV-infected chil- apart as recommended [7]. The study pharmacist
dren have an eight-fold greater risk of hospitalization due decanted 0.25 or 0.5 ml VAXIGRIP or 0.9% saline for
to influenza virus-associated pneumonia and trend toward the TIV and placebo group, respectively into 1 ml
higher case fatality rates (8.0%) compared to HIV- syringes, and dispensed according to the computer-
uninfected children (2.0%) [1,2]. Additionally, HIV generated randomization list. The preparations were
infection is associated with prolonged shedding of macroscopically indistinguishable. Study drug was admi-
influenza virus [3]. The Advisory Committee on nistered intramuscularly by the study doctor or nurse
Immunization Practices (ACIP) recommend annual following randomization.
immunization of HIV-infected children with seasonal
trivalent influenza vaccine (TIV) [4], premised primarily Participant enrollment and randomization
on the safety and immunogenicity of TIV in HIV- Study enrollment was between 18 February and 22 May
infected children [5], without any supporting efficacy or 2009, that is, prior to the anticipated start of the influenza
effectiveness studies. Annual TIV immunization of HIV- season in South Africa [8]. Randomization lists were
infected children in South Africa and other settings with a generated by the study statistician using a computer
high prevalence of HIV is uncommon. Reasons for this random number generator (SAS 9.1; SAS Institute Inc.,
include lack of access to TIV in public immunization Cary, North Carolina, USA) in random permuted group
programs, limited data on the burden of influenza illness blocks of 10 with five in each study arm. Participants and
in such countries, and absence of efficacy or effectiveness study personnel, excluding the statistician and pharma-
studies in HIV-infected children. cist, were blinded to treatment assignment. A subset of
participants stratified by age, equal numbers below and
The primary objectives of our study were to evaluate the above 36 months of age, was coenrolled in the
efficacy and immunogenicity of seasonal TIV in HIV- immunogenicity cohort. Study nurses and study doctors
infected children. The study was registered under who were blinded to the study vaccine allocation were
ClinicalTrials.gov, number NCT00883012. involved in participant enrollment, vaccination, and
follow-up.

Assessment of vaccine efficacy


Methods Surveillance for influenza-like illness (ILI) was initiated
from enrollment and continued until the end of the
Study design and participants seasonal influenza season based on National Institute for
HIV-infected children aged 6–59 months attending the Communicable Diseases (NICD) surveillance. The child
Perinatal HIV Research Unit Clinic in Johannesburg, follow-up time for seasonal influenza was calculated to
South Africa were approached for study participation. have begun on 27 April and ended on 2 August 2009
Study eligibility criteria included CD4% at least 15 or [8,9]. Weekly short message text (SMS) reminders or
receiving ART for at least 3 months prior to enrollment; telephonic contact was made with the child’s caregiver to
parent/guardian contactable for weekly visits/calls enquire about ILI symptoms. ILI was defined as an acute
throughout the study; and participant available to attend history (<7 days) of at least two of the following signs and
illness visits at the clinic. Study exclusion criteria included symptoms: fever more than 38.5oC; myalgia or irrit-
contraindication to influenza vaccine; presence of ability; sore throat or pharyngitis; rhinorrhea and/or
underlying chronic lung disease requiring treatment in cough of less than 14 days of duration. Caregivers were
the past 6 months; contraindication to intramuscular trained to use digital thermometers for recording axillary
injections; known existing grade 3 or grade 4 laboratory temperature and requested to bring the child to the clinic
or clinical toxicity as per Division of Acquired within 48 h if there were at least two ILI criteria
Imunonediciency Syndrome (Division of AIDS) toxicity symptoms or other intercurrent illness.
tables [6]; previous history of receiving influenza vaccine;
and systemic steroid treatment for more than 21 days in Nasopharyngeal aspirates (NPAs) were obtained as
the past month. described [1], when the child presented with ILI
symptoms or any respiratory tract infection. NPA samples
Study vaccine were immersed into viral transport medium and
Single-dose 0.5-ml vials of TIV (VAXIGRIP; Sanofi- transported on wet ice to the NICD for testing within
Aventis; Lyon, France), per WHO recommended strain 12 h. Samples were tested for influenza A and influenza B
selection for the southern hemisphere for 2009, were by shell vial culture and a qualitative one-step reverse
procured commercially. Each dose contained 15 mg each transcriptase-PCR (RT-PCR) assay. Influenza A isolates
of split influenza A/Brisbane/59/2007(H1N1), A/Uru- were subtyped for seasonal-H1N1 and H3N2. (CDC
guay/716/2007(H3N2), and B/Florida/4/2006. Chil- RT-PCR protocol for detection and characterization of
dren were randomized to receive two doses of TIV influenza, CDC ref# I-007–05). Influenza identification

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Influenza vaccine in HIV-positive children Madhi et al. 371

either by shell vial culture and/or real-time RT-PCR was respiratory illness. Vaccine efficacy was calculated using
considered a confirmed illness. the formula of incidence rates as 1–IL/IP, (IL ¼ the case
incidence rate in TIV-vaccinated children; IP ¼ the case
Assessment of vaccine safety and incidence rate in placebo recipients). The 95% CIs were
immunogenicity constructed and differences between the intervention
Blood samples, obtained prior to the first and 1 month arms tested at a equal to 0.05 significance level. A per-
following the second study vaccine dose, were collected protocol analysis included children who received both
for immunogenicity testing in a subset. Samples were doses of correct study intervention per randomization
centrifuged and serum archived at 70oC until shipping and with outcomes occurring at least 14 days following
on dry ice to the Molecular and Virology Clinical the second dose. Only the first episode of an outcome
Laboratories at the University of Colorado (Denver, following receipt of study vaccine was included in
USA) for hemagglutinin antibody inhibition assay (HAI). the analysis.

Adverse events were actively solicited telephonically for Immunogenicity analysis included calculation of geo-
3 days postvaccination and parents could also contact metric means of the titers (GMTs) prior to the first dose
study staff to report adverse events. Severe adverse events and 21–35 days after the second dose of study vaccine.
(SAEs) were recorded throughout the study. CD4þ cell Treatment groups were compared with respect to the fold
counts and HIV PCR viral load (Roche Amplicor rise by a two-sided, two-sample t-test and the corre-
version 1.5; Roche Diagnostics GmbH, Mannheim, sponding 95% confidence intervals for the ratio, all using
Germany) within 3 months of randomization were logarithmic transformation. HAI titers of at least 1:40 if
abstracted. CD4þ lymphocyte counts and percentage baseline titers were less than 1:10, or an at least four-fold
were also measured 1 month after the second dose of TIV increase in those with baseline titers at least 1:10 were
by the panleukogating method [10]. considered as evidence of seroconversion. The primary
immunogenicity endpoint was seroconversion rates to
Molecular characterization of the hemagglutinin each of the three vaccine strains. The proportion of
gene children with HAI at least 1:40 after second dose of
The HA1 region of the hemagglutinin gene was amplified vaccination was a secondary endpoint, and an exploratory
by nested reverse transcription PCR assay using the Titan analysis was undertaken to compare the proportion
One Tube RT-PCR System (Roche Diagnostics GmbH) between groups achieving HAI threshold titers at least
and PFx Platinum DNA polymerase (Invitrogen 1:110. Differences in proportion between study groups
Corporation, Carlsbad, California, USA) reagents. were compared with Kruskal–Wallis test. All statistical
PCR products were directly sequenced using the BigDye analyses were done using SAS software V9.1.
V3.1 Terminator reagents (Applied Biosystems, Foster
City, California, USA). Sequence alignments were done Ethical considerations
in BioEdit Version 7.0.8.0 and evolutionary analysis and The study was approved by the Human Research Ethics
neighbor-joining phylogenetic trees analysis were done in Committee of the University of the Witwatersrand and
MEGA 4 using the Maximum likelihood options with conducted in accordance with Good Clinical Practice
the Tamura-Nei model and 1000 bootstrap iterations as guidelines. Signed, written informed consent was
well as P-distance analysis [11]. obtained from caregiver/parent of study participants.
The study was registered at ClinicalTrial.gov number:
Study sample size and statistical analysis NCT NCT00883012.
On the basis of an estimated attack rate of 20% for
confirmed influenza illness in the placebo group and a
power of 80% with a a of 0.05 to detect at least a 50%
reduction among vaccinees, the required sample size Results
calculated was 420. The sample size was adjusted to 550
for possible withdrawals and/or noncompliance with Of the 410 children enrolled, 203 (98.5%) TIV recipients
study protocol. A sample size of 100 was calculated for the and 200 (98.0%) placebo recipients received both study
nested immunogenicity study to demonstrate serocon- vaccine doses (Fig. 1), at a median interval of 28 days
version rates of at least 40% among TIV recipients and less between doses. Overall, 76% of children were aged
than 5% in placebo recipients. Enrollment into the 6–35 months. The median age, CD4þ lymphocyte
immunogenicity subgroup was stratified to include count, HIV viral load, and ART regimens of the overall
50 children aged 6–35 months (younger age group) efficacy and immunogenicity subset cohorts are shown in
and 50 aged 36–60 months (older age group). Table 1. Baseline demographic and other clinical
parameters were similar between study arms in the
The primary efficacy endpoint was reduction of overall efficacy and immunogenicity subset cohorts
confirmed seasonal influenza illness. Secondary outcome (Table 1). Among TIV recipients, there were no
measures included differences for ILI and any acute significant changes in median CD4% or absolute

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
372 AIDS 2013, Vol 27 No 3

Total enrolled
N = 410

Allocated to TIV group (N = 206) Allocated to placebo (N = 204)


Received TIV (N = 206) Randomized Received placebo (N = 204)
(203 received both doses) (200 received both doses)

Immunogenicity cohort; N = 44 Immunogenicity cohort; N = 52

Loss to follow-up/relocated = 4 Loss to follow-up/relocated=0


Withdrawal: 1 Withdrawal: 0
No post-vaccine sample taken; n = 2 No post-vaccine sample taken; n = 4
Post-immuno after 21–35 days, n = 2 Post-immuno after 21–35 days, n = 5
Evaluable = 40 Evaluable = 43

Loss to follow-up/relocated = 8 Loss to follow-up/relocated = 6


Withdrawal = 2 Withdrawal = 1
Died=1; not HIV-infected = 1 Died = 1
Included in ITT analysis = 206 Included in ITT analysis = 204
Included in per protocol analysis = 203 Included in per protocol analysis = 200

Influenza-like illness: N = 66 Influenza-like illness: N = 78


Seasonal influenza Seasonal influenza
Confirmed influenza: N = 19 Confirmed influenza: N = 21

Fig. 1. Disposition of patients randomized and included in the analysis. ITT, intent-to-treat; TIV, trivalent influenza vaccine.

CD4þ cell counts postvaccination compared to pre- significantly lower for A/Uruguay(H3N2) compared to
vaccine levels in younger children (P ¼ 0.88 and 0.11; older age group (Table 2).
respectively). Also, no change in median CD4þ cell count
was observed in the older age group (P ¼ 0.62), but The proportion of TIV recipients with postvaccination
postvaccination, CD4þ% was higher (P ¼ 0037; Table 1). HAI at least 1:40 was lower in the younger compared to
older age group for A/Uruguay(H3N2) (52.2 vs. 94.1%;
Immunogenicity of trivalent influenza vaccine P ¼ 0.005) and B/Florida (43.5 vs. 88.2%; P ¼ 0.004;
The immunogenicity per-protocol analysis was limited to Table 3). Similarly, fewer younger TIV recipients (31.0%)
83 (86.5%) of 96 children (Fig. 1). Postvaccination, had HAI at least 1:110 to A/Uruguay(H3N2) compared
seroconversion was observed in 47.5, 50.0, and 40.0% of to older TIV recipients (52.9%; P ¼ 0.013; Table 2). The
TIV recipients for A/Brisbane(H1N1), A/Uruguay- proportion of TIV recipients with HAI at least 1:215 was
(H3N2), and B/Florida strains, respectively less than 30% to A/Brisbane(H1N1), less than 24% to
(P < 0.0001 for all strains compared to placebo recipients; A/Uruguay(H3N2), and less than 12% to B/Florida in
Table 2). Among TIV recipients, less than 40% of younger both age groups (Table 2). Postvaccination, GMTs were
children seroconverted to any vaccine strain, and this was lower in the younger compared to older age group of TIV

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Table 1. Demographic and clinical characteristics for the study cohort evaluating the efficacy of trivalent influenza vaccine included in the intent-to-treat analysis.

Measure Efficacy cohort Immunogenicity cohort subseta

TIV N ¼ 206 Placebo N ¼ 204 TIV N ¼ 40 Placebo N ¼ 43 TIV 6–35 months N ¼ 23 TIV 36–59 months N ¼ 17 P

Median age; months 23.1 (6.0–59.9) 24 (6.0–59.8) 34.0 (11.3–59.1) 41.9 (8.9–56.8) 21.4 (11.3–36.0) 47.1 (36.6–59.1) Not applicable
(range)
6 to <36 months 163 (79.1)b 148 (72.6) 23 (57.5) 20 (46.5) Not applicable
36–59 months 43 (20.9) 56 (27.4) 17 (42.5) 23 (53.5)
Women; n (%) 122 (59.2) 109 (53.4) 28 (70.0) 25 (58.1) 17 (73.9) 11 (64.7) 0.53
Ever breastfed, n (%) 34 (16.5) 41 (20.1) 5 (12.5) 9 (20.9) 1 (4.4) 4 (23.5) 0.14
Median weight kg 10.9 (5.4–23.9) 10.9 (5.8–19.5) 11.9 (6.9–18.9) 12.4 (7.5–19.2) 10.8 (6.8–18.5) 14.5 (10.9–18.9) <0.0001
(range)
Median CD4þ % 33.5 (16.3–59.7) 33.4 (15.2–54) 33.1 (16.7–55.9) 34.7 (17.4–50.5) 37.6 (16.7–55.9) 31.2 (22.5–43) 0.02
(range)
Median CD4þ cell 1770.0 (464–4390) 1648.0 (491–4615) 1614 (1165–2301) 1592 (1156–2849) 1793 (1531–2366) 1218 (1080–2093) 0.08
count (IR)
HIV <400 copies 117/153 (76.5) 113/148 (76.4) 15/20 (75.0) 23/25 (92.0) 7/9 (77.8) 8/11 (72.7) 0.99
per ml (%)
On ART (%) 190 (92.2) 187 (91.7) 36 (90.0) 41 (95.3) 22 (95.7) 14 (82.4) 0.29
D4T-3TC- Kaletrac 128 127 20 29 12 8
ZDV-3TC- Kaletra 50 48 11 7 9 2
Other 12 12 5 5 1 4
Postvaccination
Median CD4% Limited to immunogenicity subset 35.5 (18–55.9) 34.0 (15.4–52) 35.9 (18–55.9) 35.4 (22.8–48.2) 0.50
(range)
Median CD4þ cell 1841 (1377–2406) 1452.5 (1083–2059) 2040 (1677–2632) 1377 (1041–1687) 0.011
count (IR)

3TC, lamivudine; ART, antiretroviral therapy; D4T, stavudine; IR, interquartile range; TIV, trivalent influenza vaccine; ZDV, zidovudine.
a
Children also coenrolled into the nested immunogenicity study.
b
Values in parentheses indicate percentage, unless otherwise indicated in row.
c
Kaletra, lopinavir/ritonavir.
Influenza vaccine in HIV-positive children Madhi et al.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
373
374
AIDS

Table 2. Hemagglutinin inhibition assay titers preintervention and 1 month following two doses of trivalent inactivated influenza or placebo in HIV-infected children.

TIV Placebo TIV, 6–35 months TIV, 36–59 months


2013, Vol 27 No 3

Measure N ¼ 40 N ¼ 43 P N ¼ 23 N ¼ 17 Pa

A/Brisbane (H1N1)
Prevaccine HAI40; N (%) 25 (62.5) 30 (69.8) 0.48 13 (56.5) 12 (70.6) 0.36
Seroconversionb; N (%) 19 (47.5) 2 (4.7) <0.0001 9 (39.1) 10 (58.8) 0.22
Postvaccination HAI40; N (%) 34 (85.0) 34 (79.1) 0.48 20 (87.0) 14 (82.4) 0.99
Postvaccination HAI110; N (%) 18 (45.0) 3 (7.0) <0.0001 9 (39.1) 9 (52.9) 0.39
Postvaccination HAI215; N (%) 10 (25.0) 0 (0) <0.0001 5 (21.7) 5 (29.4) 0.58
Prevaccine GMT (95% CI)c 33.1 (26.7–40.9) 38.7 (32.8–45.8) 0.24 29.6 (23.4–37.5) 38.4 (25.5–57.7) 0.23
Postvaccine GMT (95% CI) 91.9 (64.9–130.2) 44.8 (37.4–53.6) 0.0003 77.6 (49.2–122.5) 115.5 (64.7–206.0) 0.26
d
Mean GMT fold increase (95% CI) 2.8 (1.9–4.1) 1.2 (1.0–1.3) <0.0001 2.6 (1.7–4.0) 3.0 (1.4–6.3) 0.73
A/Uraguay (H3N2)
Prevaccine HAI40; N (%) 7 (17.5) 8 (18.6) 0.90 1 (4.4) 6 (35.3) 0.29
Seroconversion (%) 20 (50.0) 5 (11.6) <0.0001 8 (34.8) 12 (70.6) 0.025
Postvaccination HAI40; N (%) 28 (70.0) 14 (32.6) 0.001 12 (52.2) 16 (94.1) 0.005
Postvaccination HAI110; N (%) 12 (30.0) 7 (16.3) 0.14 3 (13.0) 9 (52.9) 0.013
Postvaccination HAI215; N (%) 5 (12.5) 1 (2.33) 0.10 1 (4.4) 4 (23.5) 0.14
Prevaccine GMT (95% CI) 16.8 (12.2–23.1) 16.0 (12.4–20.5) 0.79 12.3 (9.5–16.1) 25.5 (13.4–48.5) 0.020
Postvaccine GMT (95% CI) 57.6 (39.9–82.9) 25.5 (18.4–35.3) 0.001 33.4 (22.2–50.1) 120.3 (72.6–199.3) 0.0002
Mean GMT fold increase (95% CI) 3.4 (2.3–5.1) 1.6 (1.2–2.1) 0.001 2.7 (1.8–4.1) 4.7 (2.1–10.5) 0.17
B/Florida
Prevaccine HAI40; N (%) 5 (12.5) 2 (4.7) 0.25 1 (4.4) 4 (23.5) 0.14
Seroconversion (%) 16 (40.0) 0 <0.0001 8 (34.8) 8 (47.1) 0.43
Postvaccination HAI40; N (%) 25 (62.5) 4 (9.3) <0.0001 10 (43.5) 15 (88.2) 0.007
Postvaccination HAI110; N (%) 7 (17.5) 0 (0) 0.004 4 (17.4) 3 (17.7) 0.99
Postvaccination HAI215; N (%) 2 (5.0) 0 (0) 0.23 0 (0) 2 (11.8) 0.17
Prevaccine GMT (95% CI) 12.1 (8.7–16.8) 9.8 (8.2–11.9) 0.27 7.9 (6.0–10.2) 21.7 (11.7–40.2) 0.001
Postvaccine GMT (95% CI) 39.3 (27.2–56.8) 11.6 (9.6–14.0) <0.0001 27.9 (17.0–45.7) 62.6 (37.3–105.2) 0.025
Mean GMT fold increase (95% CI) 3.2 (2.3–4.6) 1.2 (1.0–1.3) <0.0001 3.5 (2.3–5.4) 2.9 (1.5–5.6) 0.57
a
P value comparing seroconversion rates in trivalent influenza vaccine (TIV) recipients between children 6–36 months of age compared to older children.
b
Seroconversion defined as hemagglutinin inhibition (HAI) titers of 1:40 if baseline titers were <1:10; or an at least four-fold increase in those with baseline titers 1:10.
c
GMT, geometric mean titers of hemagglutinin inhibition antibody. 95% Confidence intervals (95% CI) shown in parenthesis.
d
Measure fold increase in GMT comparing prevaccine to postvaccine HAI titers.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Influenza vaccine in HIV-positive children Madhi et al. 375

23.4 (63.0 to 64.7)

24.7 (64.7 to 66.4)

21.2 (80.3 to 66.3)


Table 3. Efficacy of trivalent inactivated influenza vaccine in preventing any respiratory illness, influenza-like illness, and influenza confirmed illness in HIV-infected children during circulation of
recipients for A/Uruguay(H3N2) and B/Florida

Vaccine efficacy

Not calculated

Not calculated
(Table 2).

(95% CI) Efficacy of trivalent influenza vaccine against


seasonal influenza illness
Investigation for influenza virus occurred in 123 children
who were investigated on 144 illness occasions, including
74 (51.4%) which fulfilled ILI criteria (Table 3). Multiple
N ¼ 200
Placebo

(2.9)
(1.2)
(0.7)
(0.6)

17 (0.6)

15 (0.6)
Per-protocol

2695.4

illness visits spaced at least 7 days apart were undertaken in


78
31
18
17
20 children, including one with three illness visits. The
median age of children was 18.8 months (range 6–54.6)
for ILI and 20.9 months (range 6–54.6) for confirmed
influenza illness. There were 19 episodes of confirmed
N ¼ 203

(2.4)
(0.7)
(0.5)
(0.5)

13 (0.5)

12 (0.4)
2736.4
TIV

influenza illness among TIV recipients and 21 in placebo


66
19
14
14

recipients. The dominant strain of influenza virus


identified was H3N2 (37 of 40 cases). Two TIV
recipients, aged 13.5 and 21.7 months with CD4þ% of
27.6 and 36.8 and HIV viral load of less than 40 and more
(2.7)
(0.9)
(0.6)
(0.6)

30 (0.6)

27 (0.5)
N ¼ 403
5431.8
Total

than 750 000 copies/ml at enrollment, respectively,


50
32
31
144

experienced two episodes of confirmed H3N2 illness


spaced 13 and 38 days apart.

The overall incidence (per 100 child-weeks) was 2.6 (95%


22.6 (36.2 to 56.6)

19.1 (61.0 to 59.9)

15.9 (72.8 to 59.5)


Vaccine efficacy

CI 2.2–3.1) for medically attended respiratory illness,


Not calculated

Not calculated
(95% CI)

including 1.3 (95% CI 1.1–1.7) for ILI and 0.7 (95% CI


0.5–1.0) for confirmed influenza illness (Table 3). The
incidence rates of ILI or confirmed influenza illness did
not differ between study arms in the intent-to-treat or
per-protocol analysis (Table 3). There were three TIV
recipients in the immunogenicity subset with confirmed
H3N2 influenza illness, all of whom received both TIV
Intent-to-treat

N ¼ 204
Placebo

(2.8)
(1.6)
(1.1)
(0.8)

21 (0.8)

19 (0.7)

doses and were on ART. These children were 21.4, 25.0,


2749.8

and 54.6 months of age and had CD4þ% of 24.3, 31.3,


CI, confidence interval; ILI, influenza-like illness; TIV, trivalent influenza vaccine.
78
45
31
21

and 31.4, respectively. Two children less than 36 months


failed to mount an immune response to any vaccine strain,
although postvaccination HAI titers were 1:40 for each
N ¼ 206

(2.4)
(1.1)
(0.9)
(0.7)

17 (0.6)

16 (0.6)
2752.3

vaccine strain in the older child.


Total follow-up time during circulation of seasonal influenza virus.
TIV

66
29
24
19

There were one TIV and two placebo recipients


hospitalized for pneumonia during the influenza season,
none of whom tested positive for influenza virus. One
(2.6)
(1.3)
(1.0)
(0.7)

38 (0.7)

35 (0.6)
N ¼ 410
5502.1

death occurred in a placebo recipient outside of a health


Total

facility during the seasonal influenza period, which was


74
55
40
144

attributed to pneumonia but not tested for influenza


virus.
Seasonal influenza follow-up period

Phylogenetic analysis on 21 randomly selected wild-type


1st episode of confirmed influenza
1st episode of ILI; n (incidence)b

Incidence per 100 child-weeks.

H3N2 viruses from study participants formed two clusters


1st episode of confirmed H3N2
All confirmed influenza illness;

with a mean protein distance of 1.2 and 2.6%,


Illness visits; n (incidence)b
ILI episodes; n (incidence)b

respectively, to the vaccine strain (Supplemental digital


seasonal influenza virus.

illness; n (incidence)b

illness; n (incidence)b

material, http://links.lww.com/QAD/A267). Site-by-


site amino acid analysis of the H3N2 viruses (Fig. 2)
in child-weeksa

n (incidence)b

showed evidence of drift in reference to the


A/Uruguay(H3N2) vaccine strain, as indicated by mis-
matched amino acid changes mapped to the immuno-
Measure

dominant epitopes A (S138A), B (K158N; N189K;


P194L), and D (K173Q; T212A).
b
a

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
376 AIDS 2013, Vol 27 No 3

130 140 150 160 170 180 190 100 210 220

A/Uruguay/716/2007
30324
19226
23857
50993
45538
43766
26582
51757
34187
43717
52343
51042
47112
55427
51255
24860
44648
54226
54565
45606
43075
A/Perth/16/2009

Epitope A Epitope B Epitope D Epitope B Epitope D

Fig. 2. Multiple sequence alignment showing amino acid (aa) mismatches in the immunodominant epitope positions (aa120 to
aa225) in reference to the A/Uruguay/716/2007 (H3N2) vaccine strain. X denotes a heterozygous aa position namely 124(S/N).

Safety of trivalent influenza vaccine Immunodominant epitopes A and B induce high


There were no grade 3 or above solicited adverse events efficiency neutralizing antibodies. Whereas single epitope
following either the first or second dose of study vaccine. changes for A or B are associated with minor antigenic
The only solicited adverse events documented among drift, mutations resulting in two or more changed
TIV recipients within 3 days of either dose of vaccine epitopes as in our study are considered as major antigenic
were pain at site of injection (n ¼ 1; 2.3%), fever at least drift which necessitates selection of a new vaccine strain
37.5oC (n ¼ 2; 4.2%), induration at injection site (n ¼ 1; [14]. HAI analysis of the wild-type H3N2 strains was not
2.3%), and weakness (n ¼ 2; 4.5%). possible due to the phenotypic characteristic of recent H3
strains which do not hemagglutinate red blood cells from
various species [15]. A reference serum to A/Uru-
guay(H3N2)(2007), however, was negative in HAI assays
Discussion against A/Perth/16/2009, suggesting a similar result
could be expected for wild-type strains with the same
To our knowledge, this is the first randomized-controlled mutations at receptor-binding site positions 138 and 194
study which evaluated the efficacy of TIV in HIV- [13].
infected children. In our study of children mainly 6–35
months of age, who are at heightened risk of severe The lack of efficacy may also be attributed to the poor
influenza illness independent of HIV infection [12], TIV immunogenicity of TIV, particularly in younger HIV-
vaccination did not prevent confirmed seasonal influenza infected children. Although we did not enroll a control
illness or ILI. The lack of efficacy was in part corroborated group of HIV-uninfected children, the seroconversion
by the poor immunogenicity of TIVagainst the dominant rates in the younger age group were lower compared to
wild-type circulating strain, particularly in the younger the 77.8% (H3N2) to 86.1% (Influenza/B) among
age group (32.0% seroconversion). healthy Canadian children aged 6–35 months [16], in
whom an identical TIV formulation and dosing
Possible reasons for the lack of efficacy in our study concentration was evaluated. The seroconversion rates
include those which are inherent to any study evaluating in the Canadian study were also generally higher
TIV vaccine efficacy. These include the unpredictability compared to our study’s older age group. The impaired
of the severity of the influenza virus which emerges and immune responses to TIV in our study may be due to
possible vaccine strain mismatch to the wild-type strain. HIV-induced impaired B-lymphocyte function, required
Genotypic analysis of H3N2 isolates from our study for immune responses to TIV, which persist even
revealed only a 1.2% change in mean protein distance following immune reconstitution with ART [17–19].
compared to A/Uruguay-like virus. These amino acid Impaired memory B-cell response to TIV in HIV-
changes, however, affected key positions in three infected individuals has been attributed to deficiencies in
hemagglutinin epitopes (A, B, and D). The changes in serum and B-cell activating factor and APRIL (a
epitopes A (S138A) and B (P194L) corresponded to proliferating-inducing ligand) and alteration in their
positions that form part of the receptor-binding site [13]. receptors [19].

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Influenza vaccine in HIV-positive children Madhi et al. 377

Traditionally HAI titers at least 1:40 have been used as a efficacy between HIV-infected adults and children may
putative measure of relative protection against influenza include that the adults were possibly more likely to have
illness in adults [20,21]. The relevance of this threshold in established underlying cell-mediated immunity to influ-
children is unclear [22,23]. Additionally, a HAI titer of enza virus because of more frequent exposure to wild-
1:40 is associated with only 50% protection against type virus over their life time. This would have possibly
influenza illness in adults [20,23], despite the likely enhanced their protection against influenza illness
presence of underlying cell-mediated immunity to because of the role of cell-mediated immunity in clearing
influenza induced by previous exposure to influenza influenza infection as well as conferring some protection
antigens from vaccination or from circulating wild-type against influenza strains which are heterologous to the
influenza-virus. This underlying cell-mediated immunity vaccine strain [24].
is associated with cross-protection against different
influenza strains [24], and boosting thereof by vaccination Recent studies have evaluated alternate influenza vaccine
may contribute towards enhancing the efficacy of TIV in formulations, such as virosomal adjuvant vaccines [33,34],
adults, which may also in-part mitigate the effect of any MF59-adjuvant and ASO3-adjuvant influenza vaccine,
mismatch between the vaccine and circulating wild-type and LAIV [35–40], for safety and immunogenicity in HIV-
influenza strains. Although cell-mediated immune infected children. Palma et al. [40] reported immunogeni-
responses are induced in young children immunized city of MF59-adjuvanted monovalent H1N1 influenza
with live-attenuated influenza vaccine (LAIV), such vaccine in HIV-infected children following a single dose of
responses are less evident with TIV in the absence of vaccine, in whom seroconversion rate was 60%, but less
previous exposure to related vaccine strains [24]. In than that in healthy controls (82%). In healthy children
addition, young children are more likely to be influenza- aged 6–72 months, the MF59-adjuvanted influenza
naive and consequently may require higher HAI titers, vaccine was 81% more efficacious than unadjuvanted
although vaccine strain mismatch may be more relevant in TIV. This included 79% efficacy against confirmed
them compared to that in adults. Black et al. [23] modeled influenza illness in children aged 6–35 months and 92%
that in children, HAI titers of at least 1:110 and at least efficacy in children aged 36–72 months, which was
1:215 were predictive of 50 and 70% protection, approximately double the efficacy observed for non-
respectively. In our study, an exploratory analysis adjuvanted TIV [41].
identified only 13% of younger and 52.9% of older
TIV recipients achieved HAI titers at least 110 to Limitations of our study include being a single-center
A/Uruguay (H3N2), and less than 30% in both age study because of it primarily been investigator-funded.
groups had titers at least 1:215 for any vaccine strain. The Furthermore, the evaluable sample size (n ¼ 403) only
one vaccine failure due to H3N2 involved a 54.6-month- had 40% power to detect a 50% reduction in confirmed
old child in our study, who had postvaccination H3N2- influenza based on the 10% attack rate observed among
HAI titer of 1:40. placebo recipients. The sample size was, however,
sufficient to detect at least 73% vaccine efficacy at 80%
Most other immunogenicity studies on influenza vaccine power based on the observed attack rate. Although the
in HIV-infected children have involved older HIV- study did not complete enrollment of the targeted 550
infected children and adolescents [25–30]. Some of these participants prior to the peak of the influenza season, this
studies preceded management of HIV-infected children was offset by only 4.1% (n ¼ 17) of enrollees, rather than
with combination, triple ART [25,26,28], and more the projected 20%, being lost to follow-up or withdrawn.
recently have focused on children/adolescents on ART A further limitation of our study is that it was conducted
who were immunologically reconstituted and virologi- over a single influenza season. The study is, therefore,
cally suppressed [29–31]. Among HIV-infected children unable to definitively conclude the relative roles of poor
on ART, immune responses to TIV correlated with TIV immunogenicity in younger children or antigenic
immunological status prior to vaccination, with moder- drift of the circulating wild-type H3N2 away from the
ate-to-severe immunocompromised children benefiting vaccine strain as being responsible for the lack of efficacy
from a second dose of vaccine [29]. In addition, the observed against confirmed influenza illness. Also, very
magnitude of immune response and proportion of few (<18%) of the children in our study were breastfed at
children with presumed seroprotective levels were lower any stage. It is possible that predominantly breastfed
than in healthy controls for at least two vaccine strains children may have mounted better immune responses, as
[30]. breastfeeding contributes to boosting of the infant
adaptive and innate immune system [42].
The lack of efficacy in our study was surprising,
considering the 75% efficacy reported in the same setting The results from our study do not support current
among HIV-infected adults despite only modest ser- recommendations for immunization of young HIV-
oconversion rates to TIV (47.4–60.8%) [32]. In addition infected children with nonadjuvanted TIV, even if on
to the slightly lower seroconversion rates to TIV in this ART. We propose that further multicentered controls
study (40–50%), other reasons for the difference in TIV studies, which include a placebo arm and which are

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
378 AIDS 2013, Vol 27 No 3

powered to investigate efficacy against confirmed 6. 2004. DAIDS Toxicity Tables. http://rcc.tech-es.com/Docu-
ment/ safetyandpharmacovigilance/Toxicity Tables_Pediatri-
influenza illness and not only immunogenicity, are c_Over3MonthsAge_v03.pdf. [Accessed 26 June 2012].
required. Such studies should consider evaluating higher 7. Sanofi. PRODUCT MONOGRAPH VAXIGRIP: inactivated in-
strength doses of TIV in young children, as well as the fluenza vaccine trivalent types A and B (Split Virion) http://
wwwnovaccinecom/pdffiles/vaxigrip_package_insertpdf. [Ac-
possible roles of LAIV and adjuvanted TIV in protecting cessed 21 May 2012].
HIV-infected children against influenza illness. 8. National Institute for Communicable Diseases. Consolidated
Influenza Surveillance Weekly Reports. http://www.nicd.ac.za.
[Accessed 26 June 2012].
9. Van Kerkhove MD, Mounts AW. 2009 versus 2010 comparison
of influenza activity in southern hemisphere temperate coun-
tries. Influenza Other Respir Viruses 2011; 5:375–379.
Acknowledgements 10. Glencross D, Scott LE, Jani IV, Barnett D, Janossy G. CD45-
assisted PanLeucogating for accurate, cost-effective dual-
The authors would like to thank all the children and platform CD4R T-cell enumeration. Cytometry 2002; 50:69–
77.
their parents for their participation in this study. In 11. Tamura K, Dudley J, Nei M, Kumar S. MEGA4: Molecular
addition, the support of the nursing, pharmacy, and Evolutionary Genetics Analysis (MEGA) software version
general staff at Perinatal HIV Research Unit and data 4.0. Mol Biol Evol 2007; 24:1596–1599.
12. Neuzil KM, Mellen BG, Wright PF, Mitchel EFJ, Griffin MR. The
clerks at the Respiratory and Meningeal Pathogens effect of influenza on hospitalizations, outpatient visits, and
Research is acknowledged. courses of antibiotics in children. N Engl J Med 2000; 342:225–
231.
13. Ndifon W, Wingreen NS, Levin SA. Differential neutralization
S.A.M. and A.V. contributed to the study conceptualiz- efficiency of hemagglutinin epitopes, antibody interference,
ation and design. First draft of the article was cowritten by and the design of influenza vaccines. Proc Natl Acad Sci U S A
S.A.M. and A.V. S.D., H.C., E.L., and T.T. were involved 2009; 106:8701–8706.
14. Huang JW, Yang JM. Changed epitopes drive the antigenic drift
in participant follow-up and management. M.V. and F.T. for influenza A (H3N2) viruses. BMC Bioinformatics 2011; 12
were responsible for laboratory testing of influenza virus. (Suppl 1):S31.
A.W. was responsible for doing the hemagglutinin 15. Medeiros R, Escriou N, Naffakh N, Manuguerra JC, van der Werf
S. Hemagglutinin residues of recent human A(H3N2) influenza
inhibition tests. C.L.C. was involved in protocol devel- viruses that contribute to the inability to agglutinate chicken
opment and study logistics. M.V. and F.T. made input on erythrocytes. Virology 2001; 289:74–85.
the article sections on genotypic characteristic of the wild- 16. Langley JM, Scheifele DW, Quach C, Vanderkooi OG, Ward B,
McNeil S, et al. Safety and immunogenicity of 2010-2011
type influenza virus, and A.W. on the immunogenicity H1N12009-containing trivalent inactivated influenza vaccine
data. All the authors critically reviewed the article and in children 12-59 months of age previously given AS03-adju-
contributed to the final draft. vanted H1N12009 pandemic vaccine: a PHAC/CIHR Influenza
Research Network (PCIRN) study. Vaccine 2012; 30:3389–
3394.
The study was funded in part through an unrestricted 17. Cagigi A, Nilsson A, Pensieroso S, Chiodi F. Dysfunctional
grant from Secure the Future Fund. The rest of the study, B-cell responses during HIV-1 infection: implication for influ-
enza vaccination and highly active antiretroviral therapy.
including vaccine procurement, was investigator-funded. Lancet Infect Dis 2010; 10:499–503.
Secure the Future did not have any input into the study 18. Cagigi A, Palma P, Nilsson A, Di Cesare S, Pensieroso S,
design, data analysis, or write-up of the article. Kakoulidou M, et al. The impact of active HIV-1 replication
on the physiological age-related decline of immature-transi-
tional B-cells in HIV-1 infected children. AIDS 2010; 24:2075–
Conflicts of interest 2080.
19. Pallikkuth S, Kanthikeel SP, Silva SY, Fischl M, Pahwa R, Pahwa
There are no conflicts of interest. S. Innate immune defects correlate with failure of antibody
responses to H1N1/09 vaccine in HIV-infected patients.
J Allergy Clin Immunol 2011; 128:1279–1285.
20. Hobson D, Curry RL, Beare AS, Ward-Gardner A. The role of
serum haemagglutination-inhibiting antibody in protection
References against challenge infection with influenza A2 and B virus.
J Hyg (Lond) 1972; 70:767–777.
1. Madhi SA, Schoub B, Simmank K, Blackburn N, Klugman KP. 21. Coudeville L, Bailleux F, Riche B, Megas F, Andre P, Ecochard
Increased burden of respiratory viral associated severe lower R. Relationship between haemagglutination-inhibiting anti-
respiratory tract infections in children infected with human body titres and clinical protection against influenza: develop-
immunodeficiency virus type-1. J Pediatr 2000; 137:78–84. ment and application of a bayesian random-effects model.
2. Madhi SA, Ramasamy N, Bessellar TG, Saloojee H, Klugman BMC Med Res Methodol 2010; 10:18.
KP. Lower respiratory tract infections associated with influen- 22. Fox JP, Cooney MK, Hall CE, Foy HM. Influenzavirus infections
za A and B viruses in an area with a high prevalence of pediatric in Seattle families, 1975-1979. II. Pattern of infection in
human immunodeficiency type 1 infection. Pediatr Infect Dis J invaded households and relation of age and prior antibody
2002; 21:291–297. to occurrence of infection and related illness. Am J Epidemiol
3. Evans KD, Kline MW. Prolonged influenza A infection respon- 1982; 116:228–242.
sive to rimantadine therapy in a human immunodeficiency 23. Black S, Nicolay U, Vesikari T, Knuf M, Del Giudice G, Della
virus-infected child. Pediatr Infect Dis J 1995; 14:332–334. Cioppa G, et al. Hemagglutination inhibition antibody titers as
4. Fiore AE, Shay DK, Broder K, Iskander JK, Uyeki TM, Mootrey G, a correlate of protection for inactivated influenza vaccines in
et al. Prevention and control of influenza: recommendations of children. Pediat Infect Dis J 2011; 30:1081–1085.
the Advisory Committee on Immunization Practices (ACIP). 24. Forrest BD, Pride MW, Dunning AJ, Capeding MR, Chotpitaya-
MMWR Recomm Rep 2008; 57 (RR-7):1–60. sunondh T, Tam JS, et al. Correlation of cellular immune
5. Prevention and control of influenza: recommendations of the responses with protection against culture-confirmed influenza
Advisory Committee on Immunization Practices (ACIP). Morb virus in young children. Clin Vaccine Immunol 2008; 15:1042–
Mortal Wkly Rep 1997; 46:1–25. 1053.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Influenza vaccine in HIV-positive children Madhi et al. 379

25. Lyall EG, Charlett A, Watkins P, Zambon M. Response to 34. Tanzi E, Esposito S, Bojanin J, Amendola A, Trabattoni D,
influenza virus vaccination in vertical HIV infection. Arch Pariani E, et al. Immunogenicity and effect of a virosomal
Dis Child 1997; 76:215–218. influenza vaccine on viral replication and T-cell activation
26. Jackson CR, Vavro CL, Valentine ME, Pennington KN, Lanier ER, in HIV-infected children receiving highly active antiretroviral
Katz SL, et al. Effect of influenza immunization on immunologic therapy. J Med Virol 2006; 78:440–445.
and virologic characteristics of pediatric patients infected with 35. Vigano A, Giacomet V, Pariani E, Giani E, Manfredini V,
human immunodeficiency virus. Pediatr Infect Dis J 1997; Bedogni G, et al. Long-term immunogenicity after one and
16:200–204. two doses of a monovalent MF59-adjuvanted A/H1N1 influ-
27. Ramilo O, Hicks PJ, Borvak J, Gross LM, Zhong D, Squires JE, enza virus vaccine coadministered with the seasonal 2009-
et al. T cell activation and human immunodeficiency virus 2010 nonadjuvanted influenza virus vaccine in HIV-infected
replication after influenza immunization of infected children. children, adolescents, and young adults in a randomized con-
Pediatr Infect Dis J 1996; 15:197–203. trolled trial. Clin Vaccine Immunol 2011; 18:1503–1509.
28. Chadwick EG, Chang G, Decker MD, Yogev R, Dimichele D, 36. Phongsamart W, Sirisanthana V, Wittawatmongkol O, Malee-
Edwards KM. Serologic response to standard inactivated influ- satharn A, Sudjaritruk T, Chearskul P, et al. Immunogenicity and
enza vaccine in human immunodeficiency virus-infected chil- safety of monovalent influenza A (H1N1) 2009 in HIV-infected
dren. Pediatr Infect Dis J 1994; 13:206–211. Thai children. Vaccine 2011; 29:8705–8711.
29. Kosalaraksa P, Srirompotong U, Newman RW, Lumbiganon P, 37. Weinberg A, Song LY, Fenton T, Nachman SA, Read JS, Patter-
Wood JM. Serological response to trivalent inactive influenza son-Bartlett J, et al. T cell responses of HIV-infected children
vaccine in HIV-infected children with different immunologic after administration of inactivated or live attenuated influenza
status. Vaccine 2011; 29:3055–3060. vaccines. AIDS Res Hum Retroviruses 2010; 26:51–59.
30. Machado AA, Machado CM, Boas LS, Lopes MC, Gouvea Ade F, 38. Levin MJ, Song LY, Fenton T, Nachman S, Patterson J, Walker R,
Succi RC, et al. Immunogenicity of an inactivated influenza et al. Shedding of live vaccine virus, comparative safety, and
vaccine and postvaccination influenza surveillance in HIV- influenza-specific antibody responses after administration of
infected and noninfected children and adolescents. AIDS Res live attenuated and inactivated trivalent influenza vaccines to
Hum Retroviruses 2011; 27:999–1003. HIV-infected children. Vaccine 2008; 26:4210–4217.
31. Montoya CJ, Toro MF, Aguirre C, Bustamante A, Hernandez M, 39. Weinberg A, Song LY, Walker R, Allende M, Fenton T, Patter-
Arango LP, et al. Abnormal humoral immune response to son-Bartlett J, et al. Antiinfluenza serum and mucosal antibody
influenza vaccination in pediatric type-1 human immunodefi- responses after administration of live attenuated or inactivated
ciency virus infected patients receiving highly active anti- influenza vaccines to HIV-infected children. J Acquir Immune
retroviral therapy. Mem Inst Oswaldo Cruz 2007; 102:501– Defic Syndr 2010; 55:189–196.
508. 40. Palma P, Romiti ML, Bernardi S, Pontrelli G, Mora N, Santilli V,
32. Madhi SA, Maskew M, Koen A, Kuwanda L, Besselaar TG, et al. Safety and immunogenicity of a monovalent MF59(R)-
Naidoo D, et al. Trivalent inactivated influenza vaccine adjuvanted A/H1N1 vaccine in HIV-infected children and
in African adults infected with human immunodeficient young adults. Biologicals 2012; 40:134–139.
virus: double blind, randomized clinical trial of efficacy, 41. Vesikari T, Knuf M, Wutzler P, Karvonen A, Kieninger-Baum D,
immunogenicity, and safety. Clin Infect Dis 2011; 52:128– Schmitt HJ, et al. Oil-in-water emulsion adjuvant with influ-
137. enza vaccine in young children. N Engl J Med 2011; 365:1406–
33. Vigano A, Zuccotti GV, Pacei M, Erba P, Castelletti E, Giacomet 1416.
V, et al. Humoral and cellular response to influenza vaccine in 42. M’Rabet L, Vos AP, Boehm G, Garssen J. Breast-feeding and its
HIV-infected children with full viroimmunologic response to role in early development of the immune system in infants:
antiretroviral therapy. J Acquir Immune Defic Syndr 2008; consequences for health later in life. J Nutr 2008; 138:1782S–
48:289–296. 1790S.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like