Skip to main content
Human Vaccines & Immunotherapeutics logoLink to Human Vaccines & Immunotherapeutics
. 2020 Sep 29;17(5):1329–1341. doi: 10.1080/21645515.2020.1814094

Subcutaneous vaccine administration – an outmoded practice

Ian F Cook 1,
PMCID: PMC8086591  PMID: 32991241

ABSTRACT

Subcutaneous vaccine (SC) administration is an outmoded practice which complicates vaccine administration recommendations. Local adverse events following immunization (AEFIs) are a recognized determinant of vaccine hesitancy/refusal which can lead to an increased prevalence of vaccine-preventable disease.

This extensive narrative review provides high-grade evidence that intramuscular (IM) administration of all vaccine types [adjuvanted, live virus and non-adjuvanted (inactivated whole cell, split cell and subunit)] significantly reduces the likelihood of local adverse events. This, combined with moderate grade evidence that IM injection generates significantly greater immune response compared with SC injection, allows a strong recommendation to be made for the IM injection of all vaccines except BCG and Rotavirus.

This will simplify vaccination practice, minimize the inadvertent misadministration of vaccines and potentially improve public trust in vaccination.

KEYWORDS: Literature review, outmoded practice, vaccine administration, subcutaneous, intramuscular, local reactogenicity, immunogenicity

Introduction

Vaccination has made, and will continue to make, a very significant contribution to world health.1 However, adverse events following immunization (AEFTs), including injection site reactions (ISRs), are a significant driver2,3 of vaccine hesitancy and refusal. The latter has resulted4 in significantly increased risks of pertussis, varicella and pneumococcal infections in non-vaccinated children compared with vaccinated children.

Consequently, the definition and implementation of best vaccination practice (site, route and technique of injection) in terms of AEFIs (reactogenicity) and immune response (immunogenicity) are mandatory.

The current mantra5 for vaccination practice has been to administer adjuvanted vaccines by intramuscular injection, live virus vaccines by subcutaneous injection and non-adjuvanted, inactivated whole cell, split and subunit vaccines by either route. This complicated regimen for vaccine administration is due to the unacceptable reactogenicity6 of subcutaneously administered adjuvanted vaccines.

Evidence-based medicine (EBM) has been championed7 as a way of improving the quality of patient care through a stepwise process of formulating the clinical questions to be answered, collating and appraising relevant data and defining the optimal response.

The purpose of this review is to use EBM to seek to rationalize the route of administration of vaccines given by SC, IM or either routes. The PICO elements8 for this review are P = human vaccine recipients, I = intramuscular route of injection, C = subcutaneous route of injection and O = reactogenicity and immunogenicity of vaccines.

Methods

Searches were made using Pubmed. Google Scholar, Scopus, Embase, Biological Abstracts, Science Citation index, Cochrane Database of Systematic Reviews (CDSR), Cochrane Central Register of Controlled Trials (CENTRAL) and Databases of Abstracts of Reviews of Effects (DARE) using the following search terms and their word variants; “vaccines,” “administration,” “subcutaneous,” “intramuscular,” “adverse reactions” and “immunogenicity.” Manual searches were made from the following journals for the date in parenthesis to January 2020: Acta Paediatrica (1998), Acta Tropica (1980), American Journal of Medicine (1946), American Journal of Public Health (1971), American Journal of Tropical Medicine and Hygiene (1998), Annals of Internal Medicine (1995), Annals of Tropical Pediatrics (1999), Archives of Diseases of Childhood (1926), Bio Drugs (1998), Biologicals (1990), British Medical Journal (1991), Canadian Medical Association Journal (1911), Clinical Infectious Diseases (1999), Clinical and Vaccine Immunology (2006), European Journal of Pediatrics (1997), Infection and Immunity (1970), Journal of Pediatrics and Childhood (1998), Expert Review of Vaccines (2002), Human Vaccines (2005), Human Vaccines & immunotherapeutics (2012), Journal of Pediatrics (1995), Journal of Travel Medicine (1997), Journal of Tropical Pediatrics (1995), Lancet (1990), Medical Journal of Australia (2004), New England Journal of Medicine (1992), Pediatrics (1960), Pediatric Infectious Disease Journal (1995), Pediatrics International (1999), Public Health (1995), Scandinavian Journal of Infectious Disease (1997), Transactions of the Royal Society of Tropical Medicine and Hygiene (1920), Vaccine (1983) and to find additional studies where these were not abstracted.

Bibliographies of all relevant articles were searched for additional studies. All route comparative studies were included for analysis except those involving patients with chronic cutaneous, subcutaneous and muscular disorders and non-English language studies unless the full article was available for translation.

Results

Fifty-eight studies, which satisfied the inclusion criteria, were retrieved by the searches (51 by literature search, 7 by a manual search of appropriate Journals). They were divided into two study design groups, randomized trials and observational studies, as recommended in the GRADE guidelines.9 The former has the potential to provide moderate to high-grade evidence whilst the latter could only give very low to low-grade evidence.

Local reactogenicity data were recorded as warmth, pain, redness and swelling. These and immunogenicity data were collated into vaccine groups; adjuvanted vaccines, live virus vaccines and non-adjuvanted vaccines (inactivated whole cell, split cell and subunit). These are presented as Tables 1–3 respectively.

Table 1.

Adjuvanted vaccines and intramuscular compared with subcutaneous administration – reactogenicity and immunogenicity

Author Study design Patients Intervention Outcome
Wright et al10 Multi-center, randomized, double-blind, phase IV study. Healthy US adults 18–61 y old n = 1564 Anthrax toxoid (AVA) vaccine administered according to 7 different protocols. Reactogenicity
IM < SC odds ratio for warmth, tenderness, erythema, induration, subcutaneous nodules.
Immunogenicity
IM not inferior to SC at 9 weeks post vaccination.
Marano et al11 Multi-center,
randomized, double blind, phase IV study.
Healthy US adults. 18–64 y old
n = 1005
Anthrax toxoid (AVA) vaccine administered according to 7
different protocols.
Reactogenicity
IM < SC odds ratio for warmth, tenderness, erythema, induration, subcutaneous nodules and pain immediately after injection.
Immunogenicity
IM not inferior to SC administration.
Pittman et al12 and
Pittman13
Single-center,
randomized, double-blind study.
Healthy US adults 18–61 y old n = 173 Anthrax toxoid (AVA)
vaccine administered according to 7 different protocols.
Reactogenicity
SC > IM odds ratio and p < .05 for warmth, tenderness, erythema, induration and subcutaneous nodule.
Immunogenicity
IM and SC comparable response12 and no data.13
Campbell et al14 Single-center, randomized, open, phase I study. Healthy US adults 18–40 y old. n = 80 Experimental Anthrax vaccine n = 60
Anthrax toxoid (AVA) vaccine,
n = 20
IM n = 10
SC n = 10
SC > IM, p < .05 for subcutaneous nodules
for AVA
Immunogenicity
Peak antibody, SC and IM comparable.
Pondo et al15 Multi-center, randomized, double-blind, phase IV study. Healthy US adults 18–61 y old n = 1564 Anthrax toxoid (AVA) vaccine administered according to 7 different protocols. Reactogenicity
SC > IM, p < .05 for warmth, tenderness, erythema, induration and subcutaneous nodule.
Immunogenicity
IM not inferior to SC administration.
Edelman et al16 Randomized,
double-blind, Phase II study.
US adults
18–40 y old
n = 144
Clostridium botulinum type F toxoid vaccine.
Data for 116 patients.
Total number of injections n = 419
IM n = 167
SC n = 252
Reactogenicity
SC > IM, p < .5 for subcutaneous nodules at primary injection.
Immunogenicity
Similar immune response in both SC and IM groups.
Carlsson et al17 Multi-center, randomized, open study. Swedish infants,
3 months old. n = 287
D, DT, DT/inactivated polio (IPV) vaccine reconstituted with Haemophilus influenzae type b, Hib-T (Act-Hib)
Data for:
n = 365 (injections.)
IM n = 184
SC n = 181
Reactogenicity
SC > IM, p < .05 for pain, redness and subcutaneous nodules.
Immunogenicity
IM and SC comparable response
Volk et al18 Multi-center,
observational study.
US children and adults. Ages not given.
Adults, n = 1338.
Children, n = 2126
Toxoid antigen
3 or 5 antigen preparations;
3 contained diphtheria, pertussis and scarlet fever;
5 contained the above 3 as well as tetanus and typhoid antigens.
Data for injections, n = 9236
IM n = 6760
SC n = 2376
Reactogenicity
SC > IM, p < .5 for sterile abscess (antigen cysts).
Immunogenicity
No data recorded
Mark et al19 Multi-center, randomized, open study. Healthy Swedish infants, 3 months old. n = 252 Diphtheria/tetanus toxoid (DT) vaccine.
Data for n = 243
IM n = 122
SC n = 121
Reactogenicity
SC > IM, p < .5 for
Redness and swelling.
SC > IM for pain
immediately after injection, but not statistically significant.
Immunogenicity
IM and SC comparable response.
Rothstein et al20 Multi-center, randomized, double-blind study. US infants,
3 months old.
n = 80
Diphtheria/tetanus/
acellular pertussis (DTaP) vaccine.
Data for n = 80
IM n = 40
SC n = 40
Reactogenicity
SC > IM, p < .05 for redness with 1st, 2nd and 3rd dose.
Immunogenicity
IM and SC comparable response.
Diggle & Deeks21 Multi-center,
randomized, single-blind study.
UK infants,
4 months old. n = 119
Diphtheria/tetanus/
whole cell pertussis (DTwP) vaccine plus HibTITER vaccine.
Data for 110
IM n = 53
SC n = 57
Reactogenicity
SC > IM, p < .05 for redness and swelling.
Immunogenicity
No data recorded.
Diggle et al22 Multi-center,
randomized, single-blind study.
UK infants 2, 3 and 4 months old.
n = 564
DTwP/Hib administered concomitantly with meningococcal C vaccine into contralateral thigh.
Data for n = 368
IM n = 189
SC n = 179
Reactogenicity
Significantly less local reactions for IM compared with SC.
Immunogenicity
IM and SC comparable response.
Jackson et al23 Multi-center, open, non-randomized,
post licensure safety study.
US children
4–6 y old.
n = 1315
DTaP vaccine.
Data for n = 1315
IM n = 985
SC n = 430
Reactogenicity
SC > IM, p < .05 for redness, swelling and
pain.
Immunogenicity
No data recorded
Ipp et al24 Multi-center, open, non-randomized study. US children
18 months old. n = 205
DTwP- Polio vaccine.
Route comparative study.
Data for n = 131
IM n = 67
SC n = 64
Reactogenicity
SC > IM, p < .05 for any reaction, any swelling.
Immunogenicity
No data provided.
Holt & Bousfield25 Multi-center, open, non-randomized
study.
English children, age data not clearly defined.
n = 895
Diphtheria toxoid vaccine (PTAP)
Data for n = 895
IM n = 556
SC n = 339
Reactogenicity
No data supplied.
Immunogenicity
IM gave significantly greater Schick conversion rate than SC injection.
Ragni et al26 Multi-center
open, randomized, phase IV study.
US patients
2–18 y old with hemophilia compared with non-hemophilic siblings.
n = 86
Inactivated,
adjuvanted Hepatitis A (HAV) virus vaccine.
Data for n = 86
IM n = 41 non- hemophilic siblings.
SC n = 45, patients with hemophilia.
M > F, p = ≤0.05 hemophilia patients compared with non-hemophilic siblings.
Reactogenicity
SC > IM,
but p > .05 for swelling.
Immunogenicity
IM > SC. GMT, anti-HAV
At 1 month:
233mIU/ml, 185mIU/ml
At 8 months:
1022mIU/ml, 584mIU/ml
Frosner et al27 Two single- center, open, randomized pilot studies.
One compared IM with SC administration.
Healthy Swiss adults, 18–45 y old.
n = 115
Virosomal, adjuvanted hepatitis A vaccine.
Data for n = 115
IM n = 71
SC n = 44
Reactogenicity
SC > IM, but p > .05 for local reaction, pain and tenderness after primary vaccination.
Immunogenicity
Seroconversion:
IM 95.8% vs SC 93.2%.
Fisch et al28 Two-center,
open, randomized study
French adults 19–59.6 y old. n = 147 Inactivated, adjuvanted Hepatitis A (HAV) vaccine
Given by IM or SC by needle injection:
Data for n = 99
IM n = 50
SC n = 49
Reactogenicity
SC > IM, p < .05 for local reaction.
Immunogenicity
IM and SC comparable response.
Parent du Chatelet et al29 Multi-center,
randomized study
French adults,
18–60 y old. n = 138
Inactivated, adjuvanted Hepatitis A vaccine. Given by IM or SC needle injection. Data for n = 92
IM n = 46
SC n = 46
Reactogenicity
SC > IM, p < .05 for redness.
Immunogenicity
IM and SC comparable response.
Ogawa et al30 Retrospective study. Healthy Japanese University students, age 19–30 y old.
n = 1135
Inactivated, adjuvanted Hepatitis B vaccine.
Data for n = 620
IM n = 247
SC n = 373
Reactogenicity
No data supplied.
Immunogenicity
Significantly better seroconversion IM vs SC
At 2 months:
IM 84.6%, SC 62.7%
At 5 months:
IM 93.5%, SC 77.0%
Probst et al31 Single center, randomized study. Swiss hemodialysis adult patients, aged 47–50 ± 14 y old.
n = 81
Adjuvanted, recombinant Hepatitis B vaccine.
Data for n = 54
IM n = 27 Deltoid muscle.
SC n = 27 Volar aspect of forearm.
Reactogenicity
No data supplied.
Immunogenicity
Seroconversion:
IM 76%, SC 69%
GMT, HBsAb
IM 443 mIU/ml
SC 79 mIU/ml
Yamamoto et al32 Single center,
open, randomized, phase I study.
Healthy Japanese adults ≥ 18 y old.
n = 124
Adjuvanted, recombinant Hepatitis B vaccine.
Data for n = 124
IM n = 62
SC n = 62
Reactogenicity
No data supplied.
Immunogenicity
Seroconversion:
IM 98%, SC 97%
GMT, HBsAb, IM > SC,
IM 791mIU/ml
SC 168mIU/ml
Suzuki et al33 Single center, phase I,
multicenter, phase II and III, open, non- randomized
studies.
Japanese patients, children ≥ 10 y old and adults.
n = 2137
Yeast derived, adjuvanted, recombinant, pre S and S containing Hepatitis B vaccine.
Data for injections n = 4723
IM n = 2693
SC n = 2030
Reactogenicity
SC > IM, p < .05 for pain, redness, swelling and warmth.
Immunogenicity
At 7 months:
IM > SC
GMT, HBsAb:
IM 1396mIU/ml
SC 748mIU/ml
Anti-pre S2:
IM1185mIU/ml
SC 566mIU/ml
Kishino et al34 Multicenter,
randomized study.
Healthy Japanese adults. Age 20–35 y old.
n = 383
Recombinant, inactivated adjuvanted Hepatitis B vaccine.
Data for n = 383
IM n = 94
SC n = 279
Reactogenicity
SC > IM, p < .5 for pain, redness, swelling and pruritis.
Immunogenicity
Seroconversion:
IM 98.7%, SC 91.6%
GMT, HBsAb
IM 1064mIU/ml
SC 231.5mIU/ml
De Lalla et al35 Single center,
open, randomized study.
Healthy Italian adults, age range 26.3–28 y old. n = 151 Adjuvanted, recombinant Hepatitis B vaccine.
Data for n = 151
IM n = 75,
SC n = 76
Reactogenicity
SC > IM, p > .05
Immunogenicity
Seroconversion:
IM 88%
SC 75%
Carpenter et al36 Retrospective study. US children with bleeding disorders,
n = 207
Testing for HbsAb was done at:
SC 53 ± 20 months,
IM 60 ± 20 months after vaccinations,
p = .02 for time after vaccination.
Adjuvanted Hepatitis B vaccine
Data for n = 206
IM n = 114
SC n = 92
Reactogenicity
SC > IM, p > .05 for intramuscular hematoma
Immunogenicity
IM and SC comparable response.
Vink et al37 Single center, open-label, randomized, Phase III study. Japanese adults, mean age 61.9 y old.
n = 60,
Herpes zoster recombinant, adjuvanted, subunit vaccine (HZ/su) containing VZV.
Data for n = 58
IM n = 29
SC n = 29
Reactogenicity
SC > IM, p < .05 for redness and swelling.
Immunogenicity
Seroconversion rates:
IM and SC 100%
Anti-gE antibody Geometric mean concentration:
IM 45521mIU/ml
SC 44126mIU/ml
Ikeno et al38 Single center, randomized, phase I study. Japanese males, 20–40 y old.
n = 120
Inactivated, adjuvanted, monovalent, whole viruses A/H5N1 influenza vaccine.
Data for n = 120
3 different doses:
(1.7 µg, 5 µg, 15 µg)
IM n = 20 each dose,
SC n = 20 each dose.
2 doses 21 d apart.
Reactogenicity
SC > IM, p < .05 for redness and swelling in 1st and 2nd dose.
Immunogenicity
Seroconversion:
After 1st dose:
1.7 µg IM 10%, SC 0%,
5 µg IM 35%, SC 10%
15 µg IM 65%, SC 42%
After 2nd dose:
1.7 µg IM 20%, SC 20%,
5 µg IM 50%, SC 20%
15 µg IM 75%, SC 68%
Hopf et al39 Single center,
open, randomized, study.
Healthy Austrian adults, 18–60 y old.
n = 116
Adjuvanted, Inactivated tick-borne encephalitis (TBE) virus vaccine.
Data for 116
IM n = 58,
SC n = 58
Reactogenicity
SC > IM, p < .05 for pain, redness and swelling.
Immunogenicity
IM and SC comparable response

Seroconversion hepatitis B vaccine – HbsAb ≥ 10mIU/ml

Seroconversion hepatitis A vaccine – anti-HAV level ≥ 20mIU/ml

Seroconversion influenza vaccine – percentage with >4 fold increase in post-vaccination hemagglutinin inhibition (HI) titer.

Table 2.

Live virus vaccines and intramuscular compared with subcutaneous administration – reactogenicity and immunogenicity

Author Study design Patients Intervention Outcome
Bernstein et al40 Single-center,
double-blind, randomized, placebo controlled
phase I study.
Healthy US adults, 18–45 y old
n = 40
Cytomegalovirus vaccine.
IM n = 16
SC n = 16
Placebo n = 8
Low dose n = 16,
IM n = 8
SC n = 8
High dose n = 16
IM n = 8
SC n = 8
Reactogenicity
Redness and swelling only seen in those who received active vaccine by SC administration.
Immunogenicity
Similar antibody response IM and SC groups.
Diez-Domingo et al41 Multi-center,
randomized,
open-label
study.
Healthy German and Spanish adults ≥ 50 y old
n = 354
Live attenuated herpes zoster vaccine.
Data for 352
IM n = 175
SC n = 177
Reactogenicity
SC > IM, p < .05 for
injection site reaction (0–21 d)
Immunogenicity
Similar antibody titers IM and SC groups.
Koblin42 Multi-center, randomized,
open-label
study.
US and Peru adults
18–50 y old n = 90
HIV DNA prime and booster with rAd5 vaccine.
Data for n = 40
IM n = 20
SC n = 20
Reactogenicity
SC > IM, p < .05 for redness/induration and pain.
Immunogenicity
Similar antibody titers in both IM and SC groups
Peters et al43 Double-blind, randomized, placebo-controlled, dose-escalation study. UK and Kenya Adults.
18–59 y old
n = 70 Nairobi,
n = 45 London.
n = 115
PTHr HIVA DNA and recombinant MVA HIVA vaccines.
Data for n = 68
IM n = 35
SC n = 33
Reactogenicity
SC > IM, p < .05 for
Moderate/severe local reactions
Immunogenicity
Similar antibody titers IM and SC groups.
Enama et al44 Single-center
randomized, open, phase I study.
US adults
18–50 y old.
n = 60
HIV, DNA and comparator rAd5 HIV vaccines
Data for DNA primes,
IM n = 10
SC n = 10
Data for rAd5 prime,
IM n = 10
SC n = 10
Reactogenicity
HIV DNA
SC > IM, p < .05 for swelling.
rAd5
SC > IM, p < .05 for swelling, redness and injection site reaction, subcutaneous nodules. (SC 2, IM 0).
Immunogenicity
Similar antibody titers IM and SC groups.
Lafeber et al45 Single-center
randomized, open study.
Dutch children 14 months old
n = 67
MMR vaccine
Data for n = 67
IM n = 33
SC n = 34
Reactogenicity
SC > IM, p > .05 for
pain immediately after injection.
Immunogenicity
Response to vaccine antigens not significantly different.
Kuter et al46 Post-licensure analysis of 33 studies. Infants/children
11–18 months old.
n = 752
MMRII – rHA vaccine and Varivax® (Varicella vaccine).
No data for numbers given by IM or SC administration.
Reactogenicity
SC > IM, p < .05 for injection site reactions.
Immunogenicity
Seropositivity rates after IM and SC administration were comparable.
Knuf et al47 Multi-center,
randomized study.
German infants/children 11–21 months old.
n = 328
MMR vaccine.
Data for n = 318
IM n = 161
SC n = 157
Reactogenicity
SC > IM, p > .05
Immunogenicity
IM and SC comparable antibody responses for all antigens.
Gillet et al48 Multi-center,
randomized,
Open-label study.
French infants/children 12–18 months old.
n = 752
Measles, mumps, rubella vaccine.
Data for n = 712
IM n = 349
SC n = 363
Reactogenicity
MMR
SC > IM, p < .05 for any injection site reaction and redness.
Varicella SC > IM, p < .05 for any injection site reaction and redness.
Immunogenicity
comparable immune response SC and IM.
Haas et al49 Multi-center
randomized, open-label, Phase III study.
Healthy French infants/children 12–18 months old.
n = 405
Measles/Mumps/
Rubella/Varicella
vaccine.
Data for n = 405
IM n = 202
SC n = 203
Reactogenicity
SC > IM, p < .05 for injection site reaction.
Immunogenicity
GMTs comparable for SC and IM groups.
Pittman et al50 Single-center, randomized, open-label,
phase I study.
Healthy US adults, at least
18 y old.
n = 43
Rift Valley Fever vaccine (MP-12)
IM n = 6 (103.4 pfu)
SC n = 10(104.7pfu)
IM n = 27(104.4pfu)
Reactogenicity
SC and IM comparable for tenderness.
Immunogenicity
IM and SC comparable
Wilck et al51 Multi-center, randomized, open, dose escalation study. US adults
18–34 y old.
n = 72
Live attenuated Vaccinia vaccine;
Modified Vaccine Ankara (MVA)
107 or 108 TCID50
Data for n = 40
IM n = 20
SC n = 20
Reactogenicity
SC > IM, p < .05 for
severe local erythema and induration (31–70 mm)
Immunogenicity
IM and SC comparable antibody responses.
Vollmar et al52 Single-center, randomized,
double-blind, phase I study.
Healthy German males 20–55 y old
n = 86
Live attenuated Vaccinia vaccine MVA-BN
108 TCID50
Data for n = 36
IM n = 18
SC n = 18
Reactogenicity
SC > IM, p < .05 for
Redness and swelling
Immunogenicity
IM and SC comparable antibody responses.
Frey et al53 Single-center,
randomized,
partially-blinded,
phase I study.
Healthy US adults aged 18–32 y old
n = 90
Live attenuated Vaccinia vaccine
MVA-BN
108 TCID50
Data for n = 30
IM n = 15
SC n = 15
for each group.
Reactogenicity
1st dose
SC > IM, p < .05 for
redness and induration
Immunogenicity
IM and SC comparable antibody responses.
Seaman et al54 Single-center, randomized, double-blind, placebo controlled study. Healthy US adults 18–34 y old.
n = 36
Live attenuated MVA Vaccinia vaccine.
107 TCID50
challenge with Vaccinia vaccine Dryvax®
Data for n = 12
IM n = 5
SC n = 7
Reactogenicity
No data supplied
Immunogenicity
SC < IM, p > .05
Dennehy et al55 Two-center,
randomized, study.
US infants and children,
12 months –
10 y old
n = 132
Varicella vaccine.
Data for n = 132
IM n = 67
SC n = 65
Reactogenicity
SC > IM, p < .05 for
injection site reaction
Immunogenicity
GMTs comparable for IM and SC administration.
Fox et al56 Non- randomized study. Brazilian male military personnel.
15–40 y old.
n = 552
Yellow Fever Vaccine
17D-NY104, dose escalation, route comparative studies. Minimum immunizing dose assessed as a 50% lethal dose of a mouse lot.
Reactogenicity
No data supplied
Immunogenicity
Minimum Immunizing Dose (mid)
IM 1.6
SC 2.5

pfu – plaque forming units

TCID50 – Median tissue culture infectious dose

mid – minimum immunizing dose

Table 3.

Non-adjuvanted (whole cell, split cell and subunit) vaccines and intramuscular compared with subcutaneous administration – reactogenicity and immunogenicity

Author Study design Patients Intervention Outcome
Leung et al57 Non-randomized study.
Every 2nd child given SC injection.
Canadian children 15 months to 5 y.
n = 498
Inactivated, whole cell Haemophilus influenzae type b polysaccharide vaccine.
Data for n = 398
IM n = 194
SC n = 194
Reactogenicity
IM > SC, p < .05 for crying
Immunogenicity
No data supplied
Cook et al58 Single-center, randomized, observer-blind study. Australian adults ≥65 y old, 55 y old if had physician diagnosed chronic disease.
n = 720
Split-virus influenza vaccine.
Data for n = 709
IM n = 356
SC n = 353
Reactogenicity
SC > IM, p < .05 for
redness, swelling and tenderness.
Immunogenicity
Seroconversion:
H3N2 IM 80.5%, SC 71.1%, p = .0045.
H1N1 IM 37.2%,
SC 26.9%, p = .0043.
B, IM 57.0%,
SC 51.0%, p = .1948.
Ruben & Jackson59 Multi-center, randomized study. US Adults 18–25 y old with small number of older subjects. Four subunit influenza vaccines,
A2/Aichi and B/Mass.
No number given for IM and SC injection.
Reactogenicity
SC > IM – 2 fold for local pain.
SC > IM – 8 fold for erythema and induration.
Immunogenicity
Fold increase in titer (post:pre vaccination) A2/Aichi:
IM 20.5, SC 6.8
A2/Aichi vs B/Mass
IM 20.5 and 8.0 respectively.
Sanchez et al60 Two-center, randomized, phase I/II, double-blind study. Japanese adults ≥65 y old.
n = 120
High dose, split virus influenza vaccine.
Data for n = 110
IM n = 55
SC n = 55
Reactogenicity
SC > IM for injection site pain, erythema, swelling and induration. p < .05
Immunogenicity
Fold increase:
H3N2 IM 16.93, SC8.31
H1N1 IM 16.0, SC 9.25
B Yamagata IM 7.51, SC 4.68
B Victoria IM 10.69, SC 6.92
Delafuente et al61 Multi-center, randomized, single-blind study Elderly males, mean age 68 y old, range 61–81 y.
On warfarin anticoagulant.
n = 26
Split virus influenza vaccine, 1991–1992.
Data for n = 26
IM n = 13, SC n = 13
Reactogenicity
No difference in adverse events between IM and SC administration.
Immunogenicity
Comparable immune response in IM and SC.
Ballester-Torrens et al62 Single-center, randomized, single-blind, phase IV study adults n = 59 Split-virus influenza vaccine.
Data for n = 59
IM n = 30
SC n = 29
Reactogenicity
SC > IM, p < .05 for local reaction and pain.
Immunogenicity
No data provided
Casajuana et al63 Multi-center, randomized, single-blind study. Spanish adults older than 18 y on oral anticoagulants
n = 229
Split virus influenza vaccine.
Data for n = 207
IM n = 92
SC n = 115
Reactogenicity
SC > IM, p < .05 for erythema
Immunogenicity
No data supplied
Laurichesse et al64 Single-center, double-blind, randomized, placebo-controlled study. French adults 18–40 y old. n = 84 Inactivated, whole cell Leptospira interrogans (Serogroup icterohaemorrhag-iae) vaccine.
Data for n = 60
IM n = 30
SC n = 30
Reactogenicity
SC > IM, p < .05 for
local reaction at 14 d.
Immunogenicity
Similar antibody response for IM and SC routes.
Ruben et al65 Single-center,
randomized study.
US adults.
Mean age:
IM 21.9 y old
SC 20.6 y old
n = 141
Inactivated, whole cell meningococcal vaccine (A,C,Y,W-135).
Data for n = 132
IM n = 66
SC n = 66
Reactogenicity
SC > IM, p < .05 for
erythema.
Immunogenicity
IM and SC comparable response.
Scheifele et al66 Single-center, non-randomized study. Canadian
children 4–6 y old
n = 101
Inactivated, whole cell meningococcal polysaccharide vaccine (A, C, Y, W-135)
First 53 given SC immunization, subsequent 48 given IM immunization.
Reactogenicity
SC > IM, p < .05 for any redness or swelling.
Immunogenicity
No data provided
Cook et al67 Single-blind, randomized, prospective trial Australian adults ≥65 y old, 55 y old if had physician diagnosed chronic disease.
n = 254
Inactivated, whole cell pneumococcal 23 valent vaccine
Data for n = 254.
IM n = 127
SC n = 127
Reactogenicity
SC > IM, odds ratio 3.2
95% CI (1.13–1.93)
Immunogenicity
Comparable antibody response IM and SC route.

Seroconversion influenza vaccine – percentage with > 4 fold increase in post-vaccination hemagglutinin inhibition(HI) titer.

Fold increase influenza vaccine – Ratio of post- to pre-vaccination titer.

Thirty studies10–39 comparing intramuscular with subcutaneous administration of adjuvanted vaccines are presented in alphabetical order in Table 1 (6 anthrax10–15, 1 botulinum toxoid,16 9 diphtheria and tetanus toxoid containing vaccines,17–25 4 hepatitis,26–29 7 hepatitis, 30–36 1 herpes zoster,37 1 influenza38 and 1 tick-borne encephalitis39). These studies could be subdivided into two groups; one with 21 randomized trials and the other with 7 observational studies and 2 randomized trials with unacceptable biases.

The 21 randomized trials being; 6 anthrax10−15, 1 botulinum toxoid,16 5 diphtheria toxoid containing vaccines,17,19-22 3 hepatitis, 27–29 3 hepatitis, 32,34,35 with 1 each of herpes zoster,37 influenza38 and tick-borne encephalitis39 vaccines. There were 7 observational studies. These were 4 diphtheria/tetanus toxoid containing vaccines18,23-25 and 3 hepatitis B vaccines.30,33,36

Two studies were excluded from the randomized trial group due to unacceptable biases. In the study, Ragni et al.26 with hepatitis A vaccine, patients with hemophilia were given SC injection and compared with non-hemophilic siblings given IM injection. Whilst in the study by Probst et al.31 with hepatitis B vaccine IM injection was given into the deltoid muscle and SC injection was given into the volar surface of the forearm.

Five studies20–24 with diphtheria/tetanus toxoid were included where the vaccines were given with a 16 mm compared with 25 mm long needle as the former was considered to give SC injection and the latter to give IM injection.

In the 21 randomized trials, local reactogenicity data were provided in 20 studies. In 18 studies,10–17,19–22,28,29,34,37-39 SC injection gave significantly greater rates of reaction than IM injection. In two other studies,27,35 SC gave greater rates of reaction than IM injection but this did not reach statistical significance. Subcutaneous nodules were significantly more frequent for SC compared with IM injection for anthrax vaccine10−15, botulinum toxoid vaccine16 and a combination diphtheria toxoid vaccine.17 In an observational study18 with diphtheria toxoid containing vaccines, sterile abscess formation was significantly greater for SC compared with IM injection.

Pain immediately after injection (assessed with a pain analogue scale) was reported11 to be significantly less for a 4 IM regimen of anthrax vaccine compared with a 4 SC regimen. Mark et al.19 reported a similar trend but this did not reach statistical significance.

Immunogenicity data were recorded in 19 of the randomized trials.10–12,14–17,19,20,22,27-29,32,34,35,37-39 Immunogenicity was greater for IM compared with SC injection in six studies27,32,34,35,37,38 being significantly greater in the studies by Kishino et al.34 (hepatitis B vaccine) and Ikeno et al.38 (first dose of an influenza vaccine). In the remaining 13 studies10–12,14–17,19,20,22,28,29,39, the immune response was comparable for IM and SC injection.

Seventeen studies comparing IM with SC administration of live virus vaccines are presented in alphabetical order in Table 2 (1 cytomegalovirus,40 1 herpes zoster,41 3 human Immunodeficiency virus,42–44 5 measles-mumps-rubella,45–49 1 Rift Valley fever,50 4 vaccinia,51–54 1 varicella55 and 1 yellow fever56). Fifteen of the 17 studies were randomized trials.40–45,47-55

In 13 studies40-44,47–53,55 out of the15 studies where reactogenicity data were provided, SC injection gave significantly greater rates of local reaction than IM injection. In the study by Lafeber et al.,45 pain immediately after injection was greater with SC compared with IM injection but this did not reach statistical significance. Two subcutaneous nodules were observed following SC injection of one HIV vaccine44 but not with IM injection.

IM and SC immunogenicity data were comparable in 15 randomized trials.40–45,47-55 Immunogenicity was greater for IM compared with SC injection in one study.54 In this study by Seaman et al.54 immunogenicity was greater for IM compared with SC injection but this did not reach statistical significance.

Eleven studies comparing IM with SC administration of non-adjuvanted, inactivated (whole cell, split cell and subunit) vaccines are presented in alphabetical order in Table 3 (1 Hemophilus influenzae type b,57 6 influenza,58–63 1 leptospirosis,64 2 meningococcal,65,66 1 pneumococcal67).

Nine of the 11 studies were randomized trials.58–65,67 In 858–60,62-65,67 of the 9 studies where reactogenicity data were provided, SC injection was associated with significantly greater rates of reaction than IM injection. In seven of the nine randomized trials where immunogenicity data were provided,58–61,64,65,67 IM gave comparable results with SC injection in four studies.61,64,65,67 In three studies,58–60 antibody response was significantly greater for IM compared with SC injection for influenza A.

Discussion

This extensive narrative review provided high-grade evidence9 that intramuscular (IM) injection significantly reduced the likelihood of local reactogenicity compared with subcutaneous (SC) injection. High-grade evidence was drawn from studies with all vaccine types (adjuvanted n = 18, live virus n = 13, non-adjuvanted inactivated (whole cell, split and subunit) n = 8).

The greater rates of reactogenicity were also seen for vaccines recommended5 to be given by SC injection (quadrivalent meningococcal polysaccharide (4vMenPV), varicella (VV), measles-mumps-rubella/varicella (MMR/V), herpes zoster vaccine) and vaccines recommended to be given by either IM or SC route (influenza and 23-valent pneumococcal (23vPPV)).

Direct route comparative studies have not been reported for inactivated polio (IPV), Japanese encephalitis (Imojev®), Q fever and rabies vaccine. Studies with IPV17,68 given IM or SC with other antigens have shown comparable immunogenicity for IPV. Consequently, the recommendation for IPV alone to be given by SC injection is inconsistent with these data.

Older rabies vaccines were derived from animal neural tissue and given by subcutaneous injection.69 Currently recommended70 rabies vaccines are derived from cell cultures and are given by IM injection. The latter are more immunogenic and associated with less severe adverse reactions than the older rabies vaccines.

Subcutaneous nodules are uncommonly reported in this review and almost entirely with adjuvanted vaccines (anthrax10−15, botulinum toxoid16 and diphtheria combination vaccine17). A single report44 of the transient formation of two nodules was made with an HIV vaccine. Subcutaneous nodules have been considered71 to be benign, self-limiting AEFIs but this is clearly not the case as demonstrated by Bernstein et al.72 who reported 11.4% of nodules persisting at 180 d post anthrax vaccination. These nodules may persist73 for years and are often associated with pruritis and superficial dermatological features such as eczema, lichenification and hyperpigmentation.

Route of administration and use of aluminum salt adjuvants are recognized71 determinants of their formation. However, the role of aluminum hydroxide sensitivity in the pathogenesis of these nodules is controversial with some authors demonstrating this phenomenon74 whilst others75 claiming that nodule formation reflects SC rather than IM injection of aluminum adjuvanted vaccines. Sterile abscess formation was also significantly greater with SC than IM injection for an adjuvanted diphtheria toxoid vaccine in an observational study.18

Pain immediately after injection might be expected76 to be greater with IM compared with SC injection as the former has a dense supply of nociceptive nerve endings with the subcutaneous space being relatively devoid of pain receptors. Pain assessed (using standardized pain assessment scales) was significantly greater with SC than IM with anthrax vaccine11 in this review. The same trend was seen with MMR45 and DT toxoid vaccines19 using the same methodology but did not reach statistical significance.

This review provided moderate grade evidence that IM injection significantly improved the immunogenicity of vaccines compared with SC injection. This grade of evidence was drawn from better antibody response/seroconversion data with adjuvanted vaccines n = 6, live virus vaccines n = 1 and non-adjuvanted, inactivated (whole cell, split and subunit) vaccines n = 3 for IM compared with SC injection. In this review, no study with SC injection was observed to be more immunogenic than IM injection. The extent and availability of the immunogenicity data were influenced by trial design factors (e.g. set to demonstrate non-inferiority between routes of administration and Phase I studies)

Phase I studies77 are safety and tolerance studies with one of their objectives to identify preferred routes of administration. In the randomized trials of this review, 33% had less than 100 patients (3/21 adjuvanted vaccines,14,20,37 9/15 live virus vaccines40,42,44,45,50-54 and 3/9 non-adjuvanted, inactivated (whole cell, split cell and subunit vaccines)).61,62,64

The combination of high-grade reactogenicity evidence with the moderate grade immunogenicity evidence allows a strong recommendation78 that all vaccines, except BCG (intradermal) and rotavirus (oral), should be given by IM injection. This will simplify vaccination practice and prevent the inadvertent misadministration of vaccines (meningococcal conjugate vaccine79 and recombinant zoster vaccine80). It may potentially reduce vaccine hesitancy/refusal2,3 due to a lower rate of ISRs with IM compared with SC injection.

The use of evidence-based medicine in vaccinology should replace highly idiosyncratic and divergent practices that are outmoded by promoting accountability based on best scientific principles.

References

  • 1.Greenwood B. The contribution of vaccination to global health: past, present and future. Phil Trans R Soc Lond Biol Sci. 2014;369(1645):20130433. doi: 10.1098/rstb.2013.0433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gowda C, Dempsey AF.. The rise (and fall?) of parental vaccine hesitancy. Hum Vaccin Immunther. 2013;9(8):1755–62. doi: 10.4161/hv.25085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Salmon DA, Moulton H, Omer SB, DeHart MP, Stokley S, Hansey NA. Factors associated with refusal of childhood vaccines among parents of school-aged children: A case-control study. Arch Pediatr Adolesc Med. 2005;159(5):470–76. doi: 10.1001/archpedi.159.5.470. [DOI] [PubMed] [Google Scholar]
  • 4.Siddiqui M, Salmon D, Omer SB. Epidemiology of vaccine hesitancy in the United States. Hum Vaccin Immunother. 2013;9(12):2643–48. doi: 10.4161/hv.27243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Australian Immunisation Handbook . Canberra. Australia. 10th ed. Australian Government Department of Health. Vaccination procedures, Administration of vaccines, Route of administration; 2016. update. immunisationhandbook.health.gov.au/vaccination-procedures/administration-of-vaccines. [Google Scholar]
  • 6.Volk VK. Safety and effectiveness of multiple antigenic preparations in a group of free-living children. Am J Pub Health. 1949;39:1299–313. doi: 10.2105/AJPH.39.10.1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bauchner H. Evidence-based medicine: A new science or an epidemiologic fad. Pediatrics. 1999;103(5):1029–31. doi: 10.1542/peds.103.5.1029. [DOI] [PubMed] [Google Scholar]
  • 8.Brockmeier AJ, Meizhi J, Przybyla P, Ananiadou S. Improving reference prioritization with PICO recognition. BMC Med Inform Decis Mak. 2019;19(1):256. doi: 10.1186/s12911-019-0992-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Balshem H, Helfand M, Schunemann HJ, Oxman AD, Kunz R. GRADE guidelines: 3. Rating the quality of evidence. J Clin Epidemiol. 2011;64(4):401–06. doi: 10.1016/jclinepi.2010.07.015. [DOI] [PubMed] [Google Scholar]
  • 10.Wright JG, Plikaytis BD, Rose CE, Parker SD, Babcock J, Keitel W, El Sahly H, Poland GA, Jacobson RM, Keyserling HL. Effect of reduced dose schedules and intramuscular injection of anthrax vaccine adsorbed on immunological response and safety profile: A randomized trial. Vaccine. 2014;32(8):1019–28. doi: 10.1016/j.vaccine.2013.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Marano N, Plikaytis BD, Martin SW, Rose C, Semenova VA. Effects of a reduced dose schedule and intramuscular administration of anthrax vaccine adsorbed on immunogenicity and safety at 7 months. JAMA. 2008;300(13):1532–43. doi: 10.1001/jama.300.13.1532. [DOI] [PubMed] [Google Scholar]
  • 12.Pittman PR, Kim-Ahn G, Pifat DY, Coonan K, Gibbs P, Little S, Pace-Templeton JG, Myers R, Parker GW, Friedlander AM. Anthrax vaccine: immunogenicity and safety of a dose-reduction, route-change comparison study in humans. Vaccine. 2002;20(9–10):1412–20. doi: 10.1016/s0264-410x(01)00462-5. [DOI] [PubMed] [Google Scholar]
  • 13.Pittman PR. Aluminum-containing vaccines associated adverse events: role of administration and gender. Vaccine. 2002;20(Suppl 3):S48–50. doi: 10.1016/s0264-410x(02)00172-x. [DOI] [PubMed] [Google Scholar]
  • 14.Campbell JD, Clement KH, Wasserman SA, Donegan S, Chrisley L, Kotloff KL. Safety, reactogenicity and immunogenicity of a recombinant protective antigen anthrax vaccine given to healthy adults. Hum Vaccin. 2007;3(5):205–11. doi: 10.4161/hv.3.5.4459. [DOI] [PubMed] [Google Scholar]
  • 15.Pondo T, Rose CE, Martin SW, Kietel WA, Keyserling HC, Babcock J, Parker S, Jacobson RM, Poland GA, McNeil MM. Evaluation of sex, race, body mass index and pre-vaccination serum progesterone levels and post-vaccination serum anti-anthrax protective immunoglobulin G on injection site adverse events following anthrax vaccine adsorbed (AVA) in the CDC-AVA human clinical trial. Vaccine. 2014;32(28):3548–54. doi: 10.1016/j.vaccine.2014.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Edelman R, Wasserman SA, Bodison SA, Perry JG, O’Donnoghue M, De Tolla LJ. Phase II safety and immunogenicity study of type F botulinum toxoid in adult volunteers. Vaccine. 2003;21(27–30):4335–47. doi: 10.1016/s0264-410x(03)00460-2. [DOI] [PubMed] [Google Scholar]
  • 17.Carlsson R-M, Claesson BA, Kayhty H, Selstam U, Iwarson S. Studies on a Hib-tetanus toxoid conjugate vaccine: effects of co-administered tetanus toxoid vaccine, of administration route and of combined administration with an inactivated polio vaccine. Vaccine. 2000;18(5–6):468–78. doi: 10.1016/x0264-410x(99)00238-8. [DOI] [PubMed] [Google Scholar]
  • 18.Volk VK, Top FH, Bunney WE. Significance of “cysts” following injections of antigens. Am J Public Health. 1954;44:1314–25. doi: 10.2105/AJPH.44.10.1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mark A, Carlsson R-M, Granstrom M. Subcutaneous versus intramuscular injection for booster DT vaccination of adolescents. Vaccine. 1999;17(15–16):2067–72. doi: 10.1016/s0264-410x(98)00410-1. [DOI] [PubMed] [Google Scholar]
  • 20.Rothstein EP, Kamiya H, Nii R, Matsuda T, Bernstein HH, Long SS, Hosbach PH, Meschievitz CK. Comparison of diphtheria-tetanus-two component acellular pertussis vaccines in United States and Japanese infants at 2, 4 and 6 months of age. Pediatrics. 1996;97:236–42. [PubMed] [Google Scholar]
  • 21.Diggle L, Deeks JJ. Effect of needle length on incidence of local reactions to routine immunization in infants aged 4 months: randomized, controlled trial. BMJ. 2000;321:931–33. doi: 10.1136/bmj.321.7266.931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Diggle L, Deeks JJ, Pollard AJ. Effect of needle size on immunogenicity and reactogenicity of vaccines in infants: randomized, controlled trial. BMJ. 2006;333:571–74. doi: 10.1136/bmj.38906.704549.7C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Jackson LA, Starkovich P, Dunstan M, Yu O, Nelson J, Dunn J, Rees T, Zavitkovsky A, Maus D, Froeschle JE. Prospective assessment of the effect of needle length and injection site on the risk of local reaction to the fifth diphtheria-tetanus-acellular pertussis vaccination. Pediatrics. 2008;121(3):e646–52. doi: 10.1542/peds.2007-1653. [DOI] [PubMed] [Google Scholar]
  • 24.Ipp MM, Gold R, Goldbach M, Maresky DC, Saunders N, Greenberg S, Davy T. Adverse reactions to diphtheria, tetanus, pertussis-polio vaccination at 18 months of age: effect of injection site and needle length. Pediatrics. 1989;83:679–82. [PubMed] [Google Scholar]
  • 25.Holt LB. Bousfield G. P.T.A.P.: the present position. BMJ. 1949;1(4607):695–99. doi: 10.1136/bmj.1.4607.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ragni MV, Lusher JM, Koerper MA, Manco-Johnson M, Krause DS. Safety and immunogenicity of subcutaneous hepatitis A vaccine in children with haemophilia. Haemophilia. 2000;6:98–103. doi: 10.1046/j.1365-2516,2000.00386x. [DOI] [PubMed] [Google Scholar]
  • 27.Frosner G, Steffen R, Herzog C. Virosomal hepatitis A vaccine: comparing intradermal and subcutaneous with intramuscular administration. J Travel Med. 2009;16(6):413–19. doi: 10.1111/j.1708-8305.2009.00351.x. [DOI] [PubMed] [Google Scholar]
  • 28.Fisch A, Cadilhac P, Vidor E, Prazuck T, Dublanchet A, Lafaix C. Immunogenicity and safety of new inactivated hepatitis A vaccine: a clinical trial with comparison of administration route. Vaccine. 1996;14(12):1132–36. doi: 10.1016/0264-410x(96)00044-8. [DOI] [PubMed] [Google Scholar]
  • 29.Parent Du Chatelet I, Lang J, Schlumberger M, Vidor E, Soula G, Genet A, Standaert SM, Saliou P. Clinical immunogenicity and tolerance studies of liquid vaccines delivered by jet-injection and a new single-use cartridge (Imule®): comparison with standard syringe injection, Imule Investigators Group. Vaccine. 1997;15(4):449–58. doi: 10.1016/s0264-410x(96)00173-9. [DOI] [PubMed] [Google Scholar]
  • 30.Ogawa M, Akine D, Sasahara T. Comparison of hepatitis B vaccine efficacy in Japanese students: a retrospective study. Environ Health Prev Med. 2019;24:80. doi: 10.1186/s12199-019-0837-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Propst T, Propst A, Lhotta K, Vogel W, Konig P. Reinformed intradermal hepatitis B vaccination in hemodialysis patients is superior in antibody response to intramuscular or subcutaneous vaccination. Am J Kidney Dis. 1998;32(6):1041–45. doi: 10.1016/s0272-6386(98)70081-2. [DOI] [PubMed] [Google Scholar]
  • 32.Yamamoto S, Kuroki T, Kurai K, Iino S. Comparison of results of phase I studies with recombinant and plasma-derived hepatitis B vaccines and controlled study comparing intramuscular and subcutaneous injection of recombinant hepatitis B vaccine. J Infect. 1986;13(SupplA):53–60. doi: 10.1016/s0163-4453(86)92698-8. [DOI] [PubMed] [Google Scholar]
  • 33.Suzuki H, Iino S, Shiraki K, Akahane Y, Okamoto H, Domoto K, Mishiro S. Safety and efficacy of a recombinant yeast-derived pre-S2 + S containing hepatitis B vaccine (TGP-943): phase 1, 2 and 3 clinical testing. Vaccine. 1994;12(12):1090–96. doi: 10.1016/0264-410X(94)90178-3. [DOI] [PubMed] [Google Scholar]
  • 34.Kishino H, Takahashi K, Sawata M, Tanaka Y. Immunogenicity, safety and tolerability of a recombinant hepatitis B vaccine manufactured by a modified process in healthy young Japanese adults. Hum Vaccin Immunother. 2018;14(7):1773–78. doi: 10.1080/21645515.2018.1452578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.de Lalla F, Rinaldi K, Santoro D, Pravettoni G. Immune response to hepatitis B vaccine given at different injection sites and by different routes: a controlled randomized study. Eur J Epidemiol. 1988;4(2):256–58. doi: 10.1007/bf00144763. [DOI] [PubMed] [Google Scholar]
  • 36.Carpenter SL, Soucie JM, Presley RJ, Ragni MV, Wicklund BM, Silvey M, Davidson H. The hemophilia treatment center network investigators. Hepatitis B vaccination is effective by subcutaneous route in children with bleeding disorders: a universal data collection database analysis. Haemophilia. 2015;21(1):e30–43. doi: 10.1111/hae.12569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vink P, Shiramoto M, Ogawa M, Eda M, Douha M, Heineman T, Lal H. Safety and immunogenicity of a Herpes Zoster subunit vaccine in Japanese population aged ≥ 50 years when administered subcutaneously vs intramuscularly. Hum Vaccin Immunother. 2017;13(3):574–78. doi: 10.1080/21645515.2016.1232787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ikeno D, Kimachi K, Kino Y, Harada S, Yoshida K, Tochihara S, Itamura S, Odagiri T, Tashiro M, Okada K. Immunogenicity of an inactivated adjuvanted whole-virion influenza A (H5N1, NIBRG-14) vaccine administered by intramuscular or subcutaneous injection. Microbiol Immunol. 2010;54:81–88. doi: 10.1111/j.1348-0421.2009.0091.x. [DOI] [PubMed] [Google Scholar]
  • 39.Hopf S, Garner-Spitzer E, Hofer M, Kundi M, Wiedermann U. Comparable immune responsiveness but increased reactogenicity after subcutaneous versus intramuscular administration of tick-borne encephalitis (TBE) vaccine. Vaccine. 2016;34(17):2027–34. doi: 10.1016/j.vaccine.2015.12.057. [DOI] [PubMed] [Google Scholar]
  • 40.Bernstein DJ, Reap EA, Katen K, Watson A, Smith K, Norberg P, Olmsted RA, Hoeper A, Morris J, Negri S. Randomized, double-blind, phase I trial of an alphavirus replicon vaccine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine. 2010;28(2):484–92. doi: 10.1016/j.vaccine.2009.09.135. [DOI] [PubMed] [Google Scholar]
  • 41.Diez-Domingo J, Weinke T, Garcia de Lomas J, Meyer CU, Bertrand I, Eymin C, Thomas S, Sadorge C. Comparison of intramuscular and subcutaneous administration of a herpes zoster live-attenuated vaccine in adults aged ≥ 50 years: A randomized non-inferiority clinical trial. Vaccine. 2015;33(6):789–95. doi: 10.1016/j.vaccine.2014.12.024. [DOI] [PubMed] [Google Scholar]
  • 42.Koblin BA, Casapia M, Morgan C, Qin L, Wang ZM, Defawe OD, Baden L, Goepfert P, Tomaras GD, Montefiori DC. Safety and immunogenicity of an HIV adenoviral vector boost after DNA plasmid vaccine prime by route of administration: a randomized clinical trial. PloS ONE. 2011;6(9):e24517. doi: 10.1371/journal.pone.0024517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Peters BS, Jaoko W, Vardas E, Panayotakopoulos G, Fast P, Schmidt C, Gilmour J, Bogoshi M, Omosa-Manyonyi G, Dally L. Studies of a prophylactic HIV-I vaccine candidate based on modified vaccinia virus Ankara (MVA) with and without DNA priming: effects of dosage and route on safety and immunogenicity. Vaccine. 2007;25(11):2120–27. doi: 10.1016/j.vaccine.2006.11.016. [DOI] [PubMed] [Google Scholar]
  • 44.Enama ME, Ledgerwood JE, Novik L, Nason MC, Gordon IJ, Holman L, Bailer RT, Roederer M, Koup RA, Mascola JR. Phase I randomized clinical trial of VRC DNA and rAd5 HIV-I Vaccine delivery by intramuscular (IM), subcutaneous (SC) and intradermal (ID)administration (VRCOII). PloS ONE. 2014;9(3):e91366. doi: 10.1371/journal.pone.0091366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lafeber AF, van der Klis FRM, Marzec AHJO, Labadie J, van Ommen R, Strieder TG, Berbers GAM. MMR vaccine in 14 months old children, intramuscular versus subcutaneous administration RIVM Report 00002 001.
  • 46.Kuter BJ, Brown M, Wiedmann RT, Harzel J, Musey L. Safety and immunogenicity of M-M-RII (combination measles-mumps-rubella vaccine) in clinical trials of healthy children conducted between 1988 and 2009. Pediatr Infect Dis J. 2016;35(9):1011–20. doi: 10.1097/INF,000000000001241. [DOI] [PubMed] [Google Scholar]
  • 47.Knuf M, Zepp F, Meyer CU, Habermehl P, Maurer L, Burow H-M, Behre U, Janssens M, Willems P, Bisanz H. Safety, immunogenicity and immediate pain of intramuscular versus subcutaneous administration of a measles-mumps-rubella -varicella vaccine to children aged 11-21 months. Eur J Pediatr. 2010;169:925–33. doi: 10.1007/s00431-010-1142-6. [DOI] [PubMed] [Google Scholar]
  • 48.Gillet Y, Habermehl P, Thomas S, Eymin C, Fiquet A. Immunogenicity and safety of concomitant administration of a measles, mumps and rubella vaccine (M-M-Rvax Pro®) and varicella vaccine (VARIVAX®) by intramuscular or subcutaneous routes at separate injection sites: a randomized clinical trial. BMC Med. 2009;7(16). doi: 10.1186/1741-7015-7-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Haas H, Richard P, Eymin C, Fiquet A, Kuter B, Soubeyrand B. Immunogenicity and safety of intramuscular versus subcutaneous administration of a combined measles, mumps, rubella and varicella vaccine to children 12 to 18 months of age. Hum Vaccin Immunother. 2019;15(4):778–85. doi: 10.1080/21645515.2018.1549452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pittman PR, McClain D, Quinn X, Coonan KM, Mangiafico J, Makuch RS, Morrill J, Peters CJ. Safety and immunogenicity of a mutagenized, live attenuated Rift Valley fever vaccine, MP-12, in a Phase I dose escalation and route comparison study in humans. Vaccine. 2016;34(4):424–29. doi: 10.1016/j.vaccine.1015.12.030. [DOI] [PubMed] [Google Scholar]
  • 51.Wilck MB, Seaman MS, Baden LR, Walsh SR, Grandpre LE, Devoy C, Giri A, Kleinjan J, Noble L, Stevenson K. Safety and immunogenicity of modified vaccinia Ankara (ACAM 3000): effect of dose and route of administration. J Infect Dis. 2010;201(9):1361–70. doi: 10.1086/651561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vollmar J, Arndtz N, Eckl KM, Thomsen T, Petzold B, Mateo L, Schlereth B, Handley A, King L, Hülsemann V. Safety and immunogenicity of IMVAMUNE, a promising candidate as a third generation smallpox vaccine. Vaccine. 2006;24(12):2065–70. doi: 10.1016/j.vaccine.2005.11.022. [DOI] [PubMed] [Google Scholar]
  • 53.Frey SE, Newman FK, Kennedy JS, Sobek V, Ennis FA, Hill H, Yan LK, Chaplin P, Wollmar J, Chaitman BR, et al. Clinical and immunologic responses to multiple doses of IMVAMUNE (Modified Vaccinia Ankara) followed by Dryvax® challenge. Vaccine. 2007;25(51):8562–73. doi: 10.1016/j.vaccine.2007.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Seaman MS, Wilck MB, Baden LR, Walsh SR, Grandpre LE, Devoy C, Giri A, Noble L, Kleinjan J, Stevenson K. Effect of vaccination with modified Vaccinia Ankara (ACAM3000) on subsequent challenge with Dryvax. J Infect Dis. 2010;201(9):1353–60. doi: 10.1086/651560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dennehy PH, Reisinger KS, Blatter MM, Veloudis PA. Immunogenicity of subcutaneous versus intramuscular Oka/Merck Varicella vaccination in healthy children. Pediatrics. 1991;88:604–07. [PubMed] [Google Scholar]
  • 56.Fox JP, Kossobudzki SL, Fonseca da Cunha J. Field studies on the immune response to 17D yellow fever virus: relation to virus sub-strain, dose and route of inoculation. Am J Epidem. 1943;38:113–38. doi: 10.1093/oxfordjournals.aje.a118875. [DOI] [Google Scholar]
  • 57.Leung AK, Chiu AS, Siu TO. Subcutaneous versus intramuscular administration of haemophilus influenzae type b vaccine. J R Soc Health. 1989;109(2):71–73. doi: 10.1177/146642408910900213. [DOI] [PubMed] [Google Scholar]
  • 58.Cook IF, Barr I, Hartel G, Pond D, Hampson AW. Reactogenicity and immunogenicity of an inactivated influenza vaccine administered by intramuscular or subcutaneous injection in elderly adults. Vaccine. 2006;24(13):2395–402. doi: 10.1016/j.vaccine.2005.11.057. [DOI] [PubMed] [Google Scholar]
  • 59.Ruben FL, Jackson GG. A new subunit influenza vaccine: acceptability compared with standard vaccines and effect of dose on antigenicity. J Infect Dis. 1972;125(6):656–64. doi: 10.1093/infdis/125.6.656. [DOI] [PubMed] [Google Scholar]
  • 60.Sanchez L, Matsuoka O, Inoue S, Inoue T, Meng Y, Nakama T, Kato K, Pandey A, Chang LJ. Immunogenicity and safety of high dose quadrivalent influenza vaccine in Japanese adults ≥ 65 years of age: a randomized controlled clinical trial. Hum Vaccin Immunother. 2020;16(4):858–66. doi: 10.1080/21645515.2019.1677437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Delafuente JC, Davis JA, Meuleman JR, Jones RA. Influenza vaccination and warfarin anticoagulation: A comparison of subcutaneous and intramuscular routes of administration in elderly men. Pharmacotherapy. 1998;18:631–36. [PubMed] [Google Scholar]
  • 62.Ballester-Torrens MP, Acosta MA, Perez MTM, Perez BI, Brunet CJ, Doval GL, Garre MP. Intramuscular route for the administration of the anti flu vaccine in patients receiving oral anticoagulation therapy. Med Clin (Barc). 2005;124:291–94. doi: 10.1157/13072321. [DOI] [PubMed] [Google Scholar]
  • 63.Casajuana J, Iglesias B, Fabregas M, Fina F, Valles J-A, Aragones R, Benitez M, Zabaleta E. Safety of intramuscular influenza vaccine in patients receiving anticoagulation therapy: a single-blinded, multi-centre randomized controlled trial. BMC Blood Disord. 2008;8:1. doi: 10.1186/1471-2326-8-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Laurichesse H, Gourdon F, Smits HL, Abdoe TH, Estavoyer JM, Rebika H, Pouliquen P, Catalina P, Dubray C, Beytout J. Safety and immunogenicity of subcutaneous or intramuscular administration of a monovalent inactivated vaccine against Leptospira interrogans serogroup Icterohaemorrhagiae in healthy volunteers. Clin Microbiol Infect. 2007;13(4):395–401. doi: 10.1111/j.1469-0691.2007.01662.x. [DOI] [PubMed] [Google Scholar]
  • 65.Ruben FL, Froeschle JE, Meschievitz C, Chen K, George J, Reeves-Hoche MK, Pietrobon P, Bybel M, Livingood WC, Woodhouse L. Choosing a route of administration for quadrivalent meningococcal polysaccharide vaccine: intramuscular versus subcutaneous. Clin Infect Dis. 2001;32:170–72. doi: 10.1086/317553. [DOI] [PubMed] [Google Scholar]
  • 66.Scheifele DW, Bjornson G, Boraston S. Local adverse effects of meningococcal vaccine. Can Med Assoc J. 1994;150:14–15. [PMC free article] [PubMed] [Google Scholar]
  • 67.Cook IF, Pond D, Hartel G. Comparative reactogenicity and immunogenicity of 23 valent pneumococcal vaccine administered by intramuscular or subcutaneous injection in elderly adults. Vaccine. 2007;25(25):4767–74. doi: 10.1016/j.vaccine.2007.04.017. [DOI] [PubMed] [Google Scholar]
  • 68.Marshall H, Nolan T, Robertson D, Richmond P, Lambert S, Jacquet M, Schuerman L. A comparison of booster immunization with a combination DTPa-IPV vaccine or DTPa plus IPV separate injections when co-administered with MMR at age 4-6 years. Vaccine. 2006;24(35–36):6120–28. doi: 10.1016/j.vaccine.2006.05.017. [DOI] [PubMed] [Google Scholar]
  • 69.Rupprecht CE, Hanlon CA, Hemachudha T. Rabies re-examined. Lancet Infect Dis. 2002;2(6):327–43. doi: 10.1016/s1473-3099(02)00287-6. [DOI] [PubMed] [Google Scholar]
  • 70.Rabies vaccines: WHO position paper . Weekly epidemiological record No.32, 2010; 85: 309–20. http:/www.who.int/wer [Google Scholar]
  • 71.Silcock R, Crawford NW, Perrett KP. Subcutaneous nodules: an important adverse event following immunization. Expert Rev Vaccines. 2019;18(4):405–10. doi: 10.1080/14760584.2019.1586540. [DOI] [PubMed] [Google Scholar]
  • 72.Bernstein DI, Jackson L, Patel SM, El Sahly HM, Spearman P, Rouphael N, Rudge TL, Hill H, Goll JB. Immunogenicity and safety of four different dosing regimens of anthrax vaccine adsorbed for post-exposure prophylaxis for anthrax in adults. Vaccine. 2014;32(47):6284–93. doi: 10.1016/j.vaccine.2014.08.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Bergfors E, Bjorkelund C, Trollfors B. Nineteen cases of persistent pruritic nodules and contact allergy to aluminium after injection of commonly used aluminium-adsorbed vaccines. Eur J Pediatr. 2005;164(11):691–97. doi: 10.1007/s00431-005-1704-1. [DOI] [PubMed] [Google Scholar]
  • 74.Bergfors E, Trollfors B, Inerot A. Unexpectedly high incidence of persistent itching nodules and delayed hypersensitivity to aluminium in children after the use of adsorbed vaccines from a single manufacturer. Vaccine. 2003;22(1):64–69. doi: 10.1016/S0264-410X(03)00531-0. [DOI] [PubMed] [Google Scholar]
  • 75.Thierry-Carstensen B. Stellfeld. Itching nodules and hypersensitivity to aluminium after the use of adsorbed vaccines from SSI. Vaccine. 2004;22(15–16):1845. doi: 10.1016/j.vaccine.2003.11.048. [DOI] [PubMed] [Google Scholar]
  • 76.Young B, Heath J. Nervous tissue, wheater’s functional histology. 4th ed. New York (NY): Churchill Livingstone; 2003. p. 140–41. [Google Scholar]
  • 77.Australian Government Department of Health, Therapeutic Goods Administration, Australian Clinical Trials Handbook, Clinical trial phases and stages, 12th October 2018.. www.tga.gov.au/book-page/clinical-trial-phases-and-stages
  • 78.Andrews JC, Schunemann HJ, Oxman AD, Pottie D, Meerpohl JJ, Coello PA, Rind D, Montori VM, Brito JP, Norris S. GRADE guidelines: 15. Going from evidence to recommendation – determinants of a recommendation’s direction and strength. J Clin Epidemiol. 2013;66(7):726–35. doi: 10.1016/j.jclinepi.2013.02.003. [DOI] [PubMed] [Google Scholar]
  • 79.Inadvertent misadministration of meningococcal conjugate vaccine – United States. June-August 2005 . MMWR Morb Mortal Weekly Report 2006; 55: 1016–17. [PubMed] [Google Scholar]
  • 80.Notes from the Field: Vaccine administration errors involving recombinant zoster vaccine – United States 2017-2018 . MMWR. Morb Mortal Weekly Report. 2018;67(20):385–86. doi: 10.15585/mmwr.mm6720a4. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Human Vaccines & Immunotherapeutics are provided here courtesy of Taylor & Francis

RESOURCES