Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 22 August 2022

A systematic literature review to clarify the concept of vaccine hesitancy

  • Daphne Bussink-Voorend   ORCID: orcid.org/0000-0002-9873-1404 1 ,
  • Jeannine L. A. Hautvast 1 ,
  • Lisa Vandeberg   ORCID: orcid.org/0000-0002-7229-2378 2 ,
  • Olga Visser 1 &
  • Marlies E. J. L. Hulscher   ORCID: orcid.org/0000-0002-2160-4810 3  

Nature Human Behaviour volume  6 ,  pages 1634–1648 ( 2022 ) Cite this article

19k Accesses

77 Citations

61 Altmetric

Metrics details

  • Human behaviour
  • Infectious diseases
  • Preventive medicine

Vaccine hesitancy (VH) is considered a top-10 global health threat. The concept of VH has been described and applied inconsistently. This systematic review aims to clarify VH by analysing how it is operationalized. We searched PubMed, Embase and PsycINFO databases on 14 January 2022. We selected 422 studies containing operationalizations of VH for inclusion. One limitation is that studies of lower quality were not excluded. Our qualitative analysis reveals that VH is conceptualized as involving (1) cognitions or affect, (2) behaviour and (3) decision making. A wide variety of methods have been used to measure VH. Our findings indicate the varied and confusing use of the term VH, leading to an impracticable concept. We propose that VH should be defined as a state of indecisiveness regarding a vaccination decision.

Similar content being viewed by others

literature review on immunization

A scoping review of global COVID-19 vaccine hesitancy among pregnant persons

literature review on immunization

An effective COVID-19 vaccine hesitancy intervention focused on the relative risks of vaccination and infection

literature review on immunization

Revisiting COVID-19 vaccine hesitancy around the world using data from 23 countries in 2021

In 2019, vaccine hesitancy (VH) was named by the World Health Organization (WHO) as one of the top-10 threats to global health, following a five-fold global increase in measles, a disease that can be prevented by vaccination 1 , 2 . The largest increase was reported in the WHO regions covering Europe and the Americas 2 . The impact of these measles outbreaks is substantial, with rises in morbidity, mortality and costs 3 , 4 , 5 . The increasing incidence of measles and other vaccine-preventable diseases has been attributed to a failure to reach adequate immunization coverage rates 2 , 6 . In the European region, VH has been identified as the main barrier to vaccination coverage 7 , 8 . This is in contrast to other regions, such as sub-Saharan Africa, where immunization coverage rates are challenged by a combination of barriers, including access and availability 9 .

In the past decade, VH has become a key topic of research in various fields, following rises in vaccine-preventable diseases, the introduction of new vaccines, the spread of misinformation and lagging vaccination coverage 10 . Moreover, the COVID-19 pandemic has drawn further attention to the role of VH in limiting the uptake of vaccines and failure to achieve collective immunity 11 , 12 , 13 . This has led to the proliferation of scientific literature on VH in the public health, biomedical and social science research fields 10 .

In 2012, the WHO established a strategic advisory group of experts (SAGE) working group with the mandate of defining VH and suggesting how to monitor and address it. The working group proposed a broad definition, describing a VH continuum from acceptance to refusal of vaccines or as a delay in acceptance or refusal despite the availability of the vaccines. The working group described VH as “A complex behavioural phenomenon specific to vaccines, context, time, and place and influenced by factors of complacency, convenience, and confidence” 14 . This broad definition emphasizes variability by describing that VH may vary between types of vaccines and different contexts, may change over time or between different geographical locations and is influenced by various determinants.

The concept of VH has been described and applied in various ways. When definitions are broad and lack clarity, this can lead to the emergence of different concepts with overlapping domains, with various concepts being used interchangeably by some and recognized as distinct entities by others 15 . Additionally, lack of conceptual clarity can lead to inadequate operationalization and cause confusion among researchers 15 . This is problematic because when studies use similar terminology with a different meaning, their results are incomparable across subgroups, locations or contexts. A clear conceptualization is needed to develop meaningful measures allowing comparison of results 16 .

A lack of conceptual clarity is observed in the literature on VH, where VH is variously conceptualized as a psychological state and as different types of vaccination behaviour 17 , 18 . In addition, the terms ‘vaccine confidence’, ‘low uptake’ and ‘low intention to vaccinate’ are often equated with VH 19 , 20 . Confusion among researchers is then illustrated by inconsistencies in the applied definitions 21 , 22 . It has even been argued that VH is a catch-all category, aggregating many different concepts rather than being one measurable construct; and this is impeding progress in the research field 23 .

A good concept definition consists of characteristics, attributes or features that are unique to that concept and distinguish it from other closely related concepts 15 . Given the importance of VH for predicting and influencing individual vaccination decisions, it is important to explore the uses of VH and propose an optimal operationalization, distinguishing VH from other closely related concepts. Such clarification could enable a universally adopted definition and aid further research in this area.

The purpose of this systematic review was to provide an overview of how VH is operationalized in the literature in terms of conceptualizations, subpopulations and measurements. Following an assessment of the various conceptualizations, we differentiated the common themes, related concepts, research fields and vaccine types. The scope and structure of this systematic review is visualized in Fig. 1 . On the basis of an interpretation of these findings, we suggest a way forward by proposing a renewed definition for VH.

figure 1

Aiming to give an overview of VH, we recognize three types of operationalizations: conceptualizations (blue), identification of subpopulations (orange) and measurements (green). Conceptualizations of VH are analysed at three levels: (1) common themes, (2) closely related concepts and (3) potential variation in conceptualization between research field and vaccine type. Each type of operationalization and its levels are discussed in separate sections.

Study selection and characteristics

The search strategy yielded 7,427 publications. After screening the titles and abstracts, 919 publications were selected for full-text screening. A total of 420 publications met the inclusion criteria. Seven additional studies were found through citation searching, two of which met the inclusion criteria, adding up to a total of 422 studies. Some studies met the criteria of more than one category, with 36 studies categorized under VH conceptualizations, 63 under VH subpopulations and 373 under VH measurements. The search process is summarized in the PRISMA flow chart (Fig. 2 ) 24 . The characteristics of included studies are described in more detail in Supplementary Table 1 .

figure 2

Visualization of the process involving identification of records from databases, screening of records, assessing reports for eligibility, inclusion of eligible studies and exclusion of non-eligible reports with reasons for exclusion. The number of records or reports in each step of the process is shown in brackets.

The included studies cover a wide geographical distribution. The limited majority (54%) originated in high-income countries (HIC), mainly the United States, Canada, Italy, Australia and France. A smaller group (43%) originated in low- and middle-income countries (LMICs), primarily China, India and Turkey. The remaining studies (3%) originated in a combination of HIC and LMICs. The majority (60%) were published in 2021 and 2022.

The included studies approach VH in relation to various vaccine types: 51% pertaining to COVID-19, 29% to childhood, 4% to human papillomavirus, 4% to influenza and 2% to miscellaneous vaccines. Additionally, 11% of the studies concern vaccines in general. Various research fields are represented, including public health (43%), biomedical science (30%), paediatrics (15%) and social sciences (12%). Mixed methods appraisal tool (MMAT) scores were calculated for 88% of the included studies, while the others could not be assessed due to their study types. The majority (68%) scored 3 or higher, indicating that 60% of the quality criteria were met.

Vaccine hesitancy conceptualization

From the 36 studies on VH conceptualization, we extracted and analysed 304 excerpts. Supplementary Table 2 shows the extracted text excerpts for each study. Our thematic analysis revealed that 93 excerpts describe an overall characterization of VH. The majority of these (69%) describe the nature of VH as heterogenous 14 , 21 , 23 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , complex 14 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 29 , 33 , 35 , 38 , 39 , 40 , 41 , 42 , 43 or varied, depending on the type of vaccine and the context 14 , 18 , 20 , 21 , 23 , 27 , 28 , 30 , 33 , 35 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 .

VH is conceptualized in 208 excerpts. The thematic analysis revealed three predominant conceptualizations in 165 (79%) excerpts: cognitions or affect, behaviour and decision making. These three conceptualizations overlap in the majority of the studies and excerpts. Illustrative excerpts of each conceptualization are presented in Table 1 . The remaining 45 (22%) excerpts represent a fragmented group of conceptualizations, without emerging themes.

Vaccine hesitancy conceptualized as cognitions or affect

From all 36 studies 14 , 17 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 53 , 98 excerpts were extracted as conceptualizing VH in terms of cognitions or affect, including questioning, emotions or beliefs regarding vaccination. For this conceptualization, we rank-ordered the most frequently used descriptions of VH, including having or expressing concerns 21 , 25 , 26 , 27 , 29 , 30 , 34 , 35 , 36 , 40 , 42 , 43 , 46 , 51 , 53 , doubts 21 , 28 , 29 , 36 , 43 or questions 21 , 26 , 47 and being reluctant 23 , 27 , 29 , 32 , 36 , 38 , 45 , 49 , 53 , 54 or unsure 14 , 21 , 27 , 29 , 34 . Many authors describe VH as pertaining to beliefs 34 , 49 , attitudes 21 , 26 , 37 , 43 , 51 or both 23 , 29 , 30 , 55 . Furthermore, vaccine-hesitant individuals are described as ambivalent to vaccination or perceiving ambiguity in vaccine-related risks 21 , 36 , 50 , 53 .

Vaccine hesitancy conceptualized as behaviour

From 35 studies 14 , 17 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 94 excerpts were extracted as conceptualizing VH as a behaviour. The majority of the excerpts describe VH in terms of various behaviours 14 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 27 , 29 , 31 , 32 , 34 , 35 , 37 , 38 , 39 , 40 , 41 , 44 , 45 , 51 , as illustrated by the following example: “VH refers to a ‘delay’ in acceptance or ‘refusal’ of vaccines” 14 . Other excerpts describe VH as a range or continuum between the extreme ends of accepting all vaccines and refusing all vaccines 21 , 22 , 27 , 28 , 29 , 30 , 31 , 33 , 36 , 38 , 43 . In a minority of the excerpts, VH is described as a specific type of vaccination behaviour, including vaccinating as recommended (despite reluctance, concerns or feeling unsure) 26 , 46 , 47 , 49 , refusing vaccines 28 or delaying vaccines and choosing an alternative schedule 50 . Some studies explicitly state that VH should not be described as a vaccination behaviour 17 , 18 , 36 , 40 . Within articles, there were inconsistencies in the behavioural descriptions of VH 18 , 22 , 26 , 27 , 28 , 29 , 31 , 38 , 41 .

Vaccine hesitancy conceptualized as decision making

From 19 studies 18 , 21 , 23 , 26 , 27 , 30 , 31 , 32 , 36 , 37 , 38 , 40 , 42 , 44 , 45 , 50 , 52 , 53 , 30 excerpts were extracted as conceptualizing VH in terms of vaccine decision-making. Some authors adopt the term VH when describing individuals who are undecided, indecisive or under consideration, and not yet having made a final vaccine decision 21 , 23 , 26 , 31 , 32 , 45 , 50 . Vaccine-hesitant individuals are described as being in various states of indecision 23 , 31 , 32 , 37 or as seeking more information to make ‘the right decision’ about vaccination 21 , 53 . Moreover, some authors describe VH as an approach to 38 or a transient stage in the process of vaccine decision-making itself 21 , 23 , 37 .

Vaccine hesitancy and related concepts

VH is often described in relation to other concepts. We extracted 142 excerpts from 31 studies describing closely related concepts 14 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 27 , 29 , 30 , 32 , 33 , 34 , 35 , 36 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 50 , 51 , 52 , 53 . The three most common concepts are confidence or trust, complacency and convenience. Together, these are referred to as ‘the 3 Cs’ 14 and described in 69 of 142 (49%) excerpts. Most often, the 3 Cs are described as having a causal relationship with VH and as representing determinants 14 , 18 , 20 , 29 , 33 , 35 , 38 , 41 , 48 , 56 .

From 25 studies, 46 excerpts were extracted as describing confidence 14 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 27 , 29 , 30 , 33 , 34 , 35 , 36 , 38 , 39 , 41 , 42 , 43 , 44 , 46 , 47 , 48 , 52 . ‘Confidence’ is defined as the trust that people have in the immunizations, the healthcare system itself, and the process leading to decisions on licensing or recommended schedules 14 , 27 , 35 . Few studies describe the (lack of) trust or confidence as a component of VH 23 , 34 , 52 .

From 22 studies 14 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 29 , 30 , 33 , 35 , 38 , 39 , 40 , 41 , 43 , 44 , 47 , 48 , 50 , 52 , 41 excerpts were extracted on the theme of complacency. ‘Complacency’ is the individual evaluation of the risks and benefits of vaccines and of the need to vaccinate 14 , 18 , 20 , 35 . The concept of complacency in relation to VH is described as the tendency to perceive the risks of vaccination as unknown or disproportionally high and the risks of the vaccine-preventable disease as low 44 , 50 . Vaccine-hesitant individuals are more committed to assessing vaccine risks and seeking ways to minimize them 23 , 40 , 47 , 50 .

From 15 studies 14 , 18 , 20 , 21 , 22 , 25 , 29 , 33 , 35 , 38 , 39 , 41 , 42 , 43 , 48 , 27 excerpts were extracted as describing the theme of convenience. ‘Convenience’ concerns not only physical availability and geographical accessibility of vaccines, but also the user-friendliness of and ability to understand immunization services 14 , 18 , 35 , 42 . In our analysis, we found that many authors refer to convenience by describing VH as the delaying or refusal of vaccines ‘despite availability’ 14 , 18 , 21 , 22 , 23 , 25 , 26 , 29 , 33 , 35 , 38 , 39 , 41 . This description acknowledges that availability of vaccines is related to vaccine uptake, while VH itself is not influenced by availability issues. However, one study adopts inconvenience and difficulty to access vaccines as dimensions of VH 42 .

Variations between research fields and vaccine types

We identified the respective research field and vaccine type of each study in the qualitative analysis to explore related differences in descriptions of VH. We identified 19 public health studies 18 , 21 , 23 , 25 , 26 , 27 , 28 , 29 , 32 , 33 , 36 , 37 , 38 , 41 , 45 , 47 , 50 , 51 , 53 , 6 paediatric studies 14 , 31 , 34 , 35 , 39 , 48 , 8 social science studies 17 , 20 , 22 , 42 , 44 , 46 , 49 , 52 and 3 biomedical studies 30 , 40 , 43 . The primary difference observed was that conceptualizations of VH in terms of decision making emerged predominantly in the public health 18 , 21 , 23 , 32 , 38 , 50 , 54 and social science fields 42 , 44 , 52 . In studies conceptualizing VH in terms of cognitions or affect, the terms ‘beliefs’ and ‘concerns’ were used in all research fields, while ‘reluctance’, ‘doubts’ and ‘questions’ were used almost exclusively in the public health field. The conceptualization of VH as a behaviour occurred in all research fields.

VH was discussed in relation to vaccination in general 14 , 17 , 18 , 22 , 23 , 27 , 28 , 29 , 32 , 33 , 35 , 36 , 38 , 41 , 42 , 43 , 46 , 48 , 49 or specifically with regard to childhood vaccines 21 , 25 , 26 , 30 , 31 , 34 , 37 , 39 , 40 , 47 , 50 , 51 , 53 , in 19 and 13 of the studies, respectively. The remaining 4 studies discussed VH in relation to COVID-19 44 , 45 , 52 and influenza 20 . Our analysis compared the studies on general vaccination and childhood vaccines but found no major differences in their respective conceptualizations.

Vaccine hesitancy subpopulations

Of the 422 included studies, 63 identified various VH subpopulations. We extracted text excerpts describing the classifications of these subpopulations and the authors’ rationales for the distinctions. The analysis identified themes aligned with the three VH conceptualization categories. Fourteen studies grouped VH subpopulations on the basis of criteria from the conceptualization as cognitions or affect 21 , 23 , 57 , 58 , 59 , 60 , 61 , 62 , 63 , 64 , 65 , 66 , 67 , 68 and 3 studies grouped VH on the basis of the conceptualization of decision making 69 , 70 , 71 . VH subpopulations grouped solely on the basis of criteria from the behaviour conceptualization were not found. However, 19 studies grouped hesitant individuals on the basis of criteria from the conceptualizations of both cognitions or affect, and behaviour 26 , 47 , 72 , 73 , 74 , 75 , 76 , 77 , 78 , 79 , 80 , 81 , 82 , 83 , 84 , 85 , 86 , 87 , 88 . The remaining 27 studies did not identify subpopulations in terms of the three conceptualizations. Twelve studies identified subpopulations on the basis of degree of VH 51 , 89 , 90 , 91 , 92 , 93 , 94 , 95 , 96 , 97 , 98 , 99 . Although degree of VH does not directly contribute to understanding of the VH concept, the instruments used to quantify it and determine cut-off values for the subpopulations contain valuable information about the operationalizations. These instruments are discussed in the following section. In addition, a group of 10 studies distinguished a VH subpopulation by asking about willingness to be vaccinated but used different criteria to do so 100 , 101 , 102 , 103 , 104 , 105 , 106 , 107 , 108 , 109 . This method was mainly found in studies on COVID-19 vaccination, published in 2021. This demonstrates the emergence of a conceptual VH category that was not identified from the conceptual studies. The final 5 studies grouped subpopulations according to miscellaneous criteria 45 , 49 , 110 , 111 , 112 . An overview is provided Supplementary Table 3 .

Measurements of vaccine hesitancy

Of the 422 studies included, 373 report a measurement of VH in individuals. An overview is provided in Supplementary Table 4 , grouping the studies according to the instruments used. The most common, albeit highly heterogenous, method used in 210 (56%) studies is a brief VH assessment comprising 1–3 questions 64 , 65 , 66 , 68 , 71 , 74 , 75 , 84 , 85 , 88 , 90 , 96 , 97 , 98 , 100 , 102 , 103 , 105 , 106 , 107 , 108 , 109 , 111 , 113 , 114 , 115 , 116 , 117 , 118 , 119 , 120 , 121 , 122 , 123 , 124 , 125 , 126 , 127 , 128 , 129 , 130 , 131 , 132 , 133 , 134 , 135 , 136 , 137 , 138 , 139 , 140 , 141 , 142 , 143 , 144 , 145 , 146 , 147 , 148 , 149 , 150 , 151 , 152 , 153 , 154 , 155 , 156 , 157 , 158 , 159 , 160 , 161 , 162 , 163 , 164 , 165 , 166 , 167 , 168 , 169 , 170 , 171 , 172 , 173 , 174 , 175 , 176 , 177 , 178 , 179 , 180 , 181 , 182 , 183 , 184 , 185 , 186 , 187 , 188 , 189 , 190 , 191 , 192 , 193 , 194 , 195 , 196 , 197 , 198 , 199 , 200 , 201 , 202 , 203 , 204 , 205 , 206 , 207 , 208 , 209 , 210 , 211 , 212 , 213 , 214 , 215 , 216 , 217 , 218 , 219 , 220 , 221 , 222 , 223 , 224 , 225 , 226 , 227 , 228 , 229 , 230 , 231 , 232 , 233 , 234 , 235 , 236 , 237 , 238 , 239 , 240 , 241 , 242 , 243 , 244 , 245 , 246 , 247 , 248 , 249 , 250 , 251 , 252 , 253 , 254 , 255 , 256 , 257 , 258 , 259 , 260 , 261 , 262 , 263 , 264 , 265 , 266 , 267 , 268 , 269 , 270 , 271 , 272 , 273 , 274 , 275 , 276 , 277 , 278 , 279 , 280 , 281 , 282 , 283 , 284 , 285 , 286 , 287 , 288 , 289 , 290 , 291 , 292 , 293 , 294 , 295 , 296 , 297 , 298 . The questions, as well as the criteria or cut-off points used to define hesitancy, vary widely between the studies. The majority of questions used in this method cover operationalizations of VH that did not emerge from our conceptual analysis, including intention and willingness. A group of 124 studies assess VH by asking about vaccination intention. For example, one measurement asks “What would you do if a COVID-19 vaccine were available?”. Respondents answering either “I would eventually get a vaccine, but wait a while first”, “I would not get a vaccine” or “I’m not sure” are all classified as hesitant 169 . A group of 35 studies assess VH by asking about willingness, exemplified by the question: “Are you willing to receive the COVID-19 vaccination?”. Respondents answering “yes, but I choose to delay timing of injection” are considered hesitant 100 . Furthermore, 23 studies assess VH by an explicit verbatim assessment of experienced hesitancy levels. This is exemplified by the question: “Overall, how hesitant about childhood vaccines would you consider yourself to be?”. Respondents answering “not too hesitant”, “not sure”, “somewhat hesitant” or “very hesitant” are considered hesitant 136 . Finally, a minority of 14 studies assess VH with questions covering conceptualizations that did emerge from our conceptual analysis; for example, by asking about previous vaccination behaviour: “Have you ever hesitated, delayed, or refused getting a vaccination for your child or yourself due to reasons other than allergies and sickness?”. Respondents answering “yes” to this question are considered hesitant 122 . The remaining 14 studies use miscellaneous questions to assess VH. Notably, the intention and willingness measures to assess VH are found mainly in studies published in 2021 on COVID-19 vaccination, while the other methods have been used throughout the covered period and in the context of different vaccines.

The second most common method, applied by 132 (35%) studies, is the use of a validated instrument. The most common instrument, used in 70 studies, is the parent attitudes about childhood vaccines (PACV) survey, introduced by Opel et al. 34 . The PACV consists of 15 questions about immunization behaviour, beliefs about vaccine safety and efficacy, attitudes toward vaccine mandates and exemptions, and trust 299 , thereby operationalizing VH as both cognitions or affect, and behaviour. Trust (or confidence) is also included in this instrument. In our conceptual analysis, confidence emerged as a distinct concept, albeit closely related to VH. Clear cut-off points for hesitancy were formulated and applied in the vast majority of the studies using this instrument (shown in Supplementary Table 4 ). The PACV is variously used in its original form 34 , 91 , 299 , 300 , 301 , 302 , 303 , 304 , 305 , 306 , 307 , 308 , 309 , 310 , 311 , 312 , 313 , 314 , 315 , 316 , 317 , 318 , 319 , 320 , 321 , 322 , 323 , 324 , 325 , 326 , 327 , 328 , 329 , 330 , 331 , 332 , 333 , 334 , 335 , 336 , 337 , 338 , or in adapted 339 , 340 , 341 , 342 , 343 , 344 , 345 , 346 , 347 , 348 , 349 , 350 , 351 , 352 , 353 , 354 , 355 or shorter versions 51 , 62 , 89 , 93 , 95 , 356 , 357 , 358 , 359 , 360 , 361 .

Other studies use a variety of validated and broadly used instruments. The SAGE instrument is applied in 13 of the studies 41 , 362 , 363 , 364 , 365 , 366 , 367 , 368 , 369 , 370 , 371 , 372 , 373 , with questions reflecting the different conceptualizations (cognitions or affect, behaviour and decision making) and related concepts including convenience, complacency and confidence 41 . The vaccine hesitancy scale (VHS), used in 39 studies 83 , 99 , 374 , 375 , 376 , 377 , 378 , 379 , 380 , 381 , 382 , 383 , 384 , 385 , 386 , 387 , 388 , 389 , 390 , 391 , 392 , 393 , 394 , 395 , 396 , 397 , 398 , 399 , 400 , 401 , 402 , 403 , 404 , 405 , 406 , 407 , 408 , 409 , 410 , was derived from a subscale of the SAGE instrument, narrowed to conceptualize VH as cognitions or affect and include the related concept of confidence 69 . The studies using the SAGE instrument and VHS use varying outcomes or cut-off values (or no outcomes or cut-off values at all) to define hesitancy (shown in Supplementary Table 4 ). The Oxford COVID-19 vaccine hesitancy scale was recently designed exclusively for the assessment of VH for COVID-19 vaccination and subsequently applied in 5 studies 44 , 411 , 412 , 413 , 414 . Other instruments described in the context of VH but intended to assess other concepts include the 5C scale 22 of psychological antecedents of vaccine behaviour, the vaccine acceptance scale (which covers the domains cognitions and affects, confidence and legitimacy of government vaccine mandates 46 ) and the multidimensional vaccine hesitancy scale covering perceptions regarding vaccines in general 42 . Instruments assessing confidence have also been applied to assess hesitancy 415 .

The remaining 31 (8%) studies use a variety of unique, self-developed methods to measure hesitancy. These are classified as ‘miscellaneous’ 25 , 50 , 52 , 69 , 73 , 92 , 94 , 416 , 417 , 418 , 419 , 420 , 421 , 422 , 423 , 424 , 425 , 426 , 427 , 428 , 429 , 430 , 431 , 432 , 433 , 434 , 435 , 436 , 437 , 438 , 439 . Examples include measurement of VH based on vaccination rates from medical records 418 and statistical procedures used to group participants according to their patterned responses to a questionnaire 92 , 439 .

Our systematic review reveals that VH is conceptualized in the literature as involving cognitions or affect, behaviour and decision making, representing three distinct but interacting entities. Closely related concepts include confidence or trust, perceptions of the need to vaccinate and of risk (complacency), and convenience. VH subpopulations are grouped according to a variety of criteria, with the majority originating in the three identified conceptualizations. Studies measuring VH have used a wide variety of instruments. The most commonly applied instruments include a brief assessment comprising 1–3 variable questions and the PACV for childhood vaccines. The instruments operationalize hesitancy using one or more of the three identified conceptualizations, but also introduce novel conceptualizations including intention and willingness. When synergizing the findings on different VH operationalizations, we found psychological and behavioural operationalizations, with the psychological operationalizations being cognitions or affect, and decision making.

Our findings illustrate the challenge of operationalizing VH, with studies adopting different conceptualizations, subpopulations and measurements. Dubé et al. acknowledged this challenge of operationalizing the VH concept due to its heterogeneity and the diversity in attitudes and behaviours 29 . Furthermore, our findings align with a recent study demonstrating the many interpretations of VH used across Europe 440 . These inconsistencies in terminology are even evidenced in the Merriam-Webster dictionary, where ‘hesitancy’ is defined as a quality or state of being that involves indecision or reluctance 441 , aligning with VH conceptualized as decision making and cognitions or affect, while ‘vaccine hesitancy’ is defined as the reluctance or refusal to vaccinate 442 , thereby also including a conceptualization of behaviour.

In the introduction, we describe interchangeable use of various terms with VH 19 , 20 . In our review, we also found numerous examples, including ‘confidence’ 443 , ‘low intention’ 444 and ‘unwillingness’ 270 . We identify these concepts as related but not synonymous to VH. For instance, some authors note that confidence or trust are used interchangeably in relation to VH 19 , 22 , suggesting equivalent meanings. Others describe an inverse relationship, meaning that lower levels of confidence are associated with higher levels of VH 19 , 33 , 54 , 56 , 445 . In line with this, VH is described as originating from a lack of confidence 446 and as a possible indicator of declining confidence 56 .

Additionally, in our analysis of subgroups and measurements, we found that VH is frequently operationalized in terms of willingness and intention, which we did not find in our conceptual analysis of VH. Willingness and intention to vaccinate, similar to the ‘vaccine confidence’ concept, are inversely related concepts that are unequivocally linked to VH but are and should not be treated as synonymous. Using these terms interchangeably is not only inappropriate but also contributes to confusion and unclarity of the VH concept. This clarity is needed because unclear concepts give rise to differences in our understanding of its determinants, correlates and consequences, hindering efforts to study and address VH 15 , 23 , 440 . Furthermore, at an operational level, there may be a mismatch between a concept and its measures 15 . This is demonstrated in our review by the highly variable methods we found to measure VH, leading to incomparable results. Particularly during 2021, there has been a plethora of studies reporting VH measurements that, due to divergent definitions and methods, have been of questionable value. As a way forward, we base our reasoning for a renewed definition of VH on the three main identified conceptual categories—behaviour, cognitions or affect, and decision making—as these have proven most promising by their repeated representation in conceptual, subgroup and measurement studies

We argue that conceptualizing VH as vaccination behaviour is untenable, as mere behaviour is insufficiently discriminating between hesitant and non-hesitant individuals. For instance, people may accept vaccines with or without hesitation or reject vaccines with or without hesitation. As concepts are ideally defined by a unique set of features that distinguishes them from other closely related concepts 15 , vaccination behaviour alone is not sufficient to define VH. Furthermore, vaccination behaviour is generally used as the indicator of (non-)acceptance of vaccination. Thus, to use this also to define another concept would create confusion. Authors have commented on the blurred distinction between VH and refusal of vaccines 25 , 39 and criticized behavioural operationalization for its failure to capture VH 17 , 18 , 23 , 25 , 40 . Although we agree that certain types of vaccination behaviour may be manifestations of VH, we argue that including behaviour in the definition and operationalization of VH is neither necessary nor sufficient.

Our analysis shows that VH is furthermore defined by two closely linked conceptualizations that we identify as psychological—cognitions or affect, and decision making. Larson et al. exemplify this stance, arguing that VH is by nature a state of indecision and reluctance 32 . We propose to reject types of vaccination behaviour as a viable conceptualization of VH; this logically results in the proposition that VH should be considered a psychological construct. This is in line with authors who have argued that VH is a psychological state rather than a behaviour 18 , 22 , 26 , 32 , 40 , inspiring our current investigation of what exactly this vaccine-hesitant state entails. In the conceptualization cognitions or affect, VH is mainly described as ‘doubts’, ‘concerns’ and ‘reluctance’ regarding vaccination. Following our analysis, we interpreted these descriptions as different ways of how VH may be affected, experienced or expressed at an individual level, representing a layer surrounding the central element of VH. We therefore interpret cognitions and affect to go hand-in-hand, but not to be at the core of hesitancy. Moreover, we conclude that cognitions or affect are insufficiently distinctive to define VH.

This interpretation does not mean that the identified cognitions or affect are irrelevant to VH. On the contrary, they may prove crucial in shaping VH. However, to arrive at a clear definition of VH, cognitions and affects should be treated as clearly defined entities as well. Only by unravelling and distinguishing them can the exact nature of their relationship with VH be clarified in further research.

In the conceptualization decision making, VH was described as being ‘undecided’, ‘indecisive’, ‘in consideration’ or ‘not yet making a vaccine decision’. All these descriptions include an element of indecision, and this provides a unique and distinctive feature for VH. Additionally, we found that this conceptualization is predominantly discussed in studies in the public health field. This is rather logical, as one would expect this field of research to take a more pragmatic approach, examining the presence of VH at a stage where people have been offered a vaccine or to anticipate public sentiments around willingness to accept a vaccine when it is offered. This probably triggers a decision-making process where VH can emerge and manifest. On the basis of these findings, we argue that VH is a psychological state of being undecided, indecisive or not yet making a decision regarding vaccination.

The study selection was conducted independently by different members of our research team. However, one possible limitation is that we did not attempt to exclude studies of lower quality, as we wanted to maintain a robust selection of studies to enable a broad overview of the relevant literature. Our MMAT assessment, however, indicates that the majority of the studies are of medium quality. A second limitation is that a considerable number of the included conceptual studies (17 of the 36) 14 , 18 , 20 , 21 , 22 , 23 , 25 , 26 , 29 , 35 , 38 , 39 , 40 , 41 , 42 , 43 , 44 quoted the VH definition introduced by the SAGE working group, which may have led to an amplification of the SAGE definition. This may indicate that this definition is well recognized, but potentially overshadows less recognized conceptual definitions of VH. We chose to include all quoted definitions and found that many studies used more than one. We did not look further into conflicting definitions within the articles, but doing so could yield interesting insights.

In conclusion, we propose a definition of VH as a psychological state of indecisiveness that people may experience when making a decision regarding vaccination. We acknowledge that experiencing concerns, doubts or reluctance regarding vaccination may play a vital role in shaping VH. However, we argue that these factors have the highest potential to advance scientific knowledge when treated as relevant constructs integral to shaping VH, rather than treating them as synonymous to VH. Operationalizing VH by measuring or distinguishing subpopulations should ideally be directed at this state of indecision. To avoid confusion, it is important to separate VH from vaccination behaviour, which is already a well-defined concept. This proposal of a renewed definition of a concept that has been used for a decade could be perceived as ‘putting old wine in new bottles’. However, we feel that due to the large amount of highly varied literature, and given the importance of VH research in predicting, explaining and influencing immunization behaviour, it is necessary to take a snapshot of the status quo. The conclusion of this review is that VH is, for now, an impracticable concept, due to the confusing use of multiple, varied operationalizations. To aid further research, the VH concept must be clearly conceptualized and adapted from its broad and inclusive form to a pragmatic and refined alternative. Working on such an alternative, the field should first reach consensus on the definition and then measure VH accordingly. This approach allows for a much-needed comparison between studies to improve our understanding of VH determinants, correlates and consequences on an individual and societal level. Our way forward is to simplify and clarify the operationalization of VH by returning to its root core of indecisiveness.

This systematic review was registered on 11 November 2020 in the PROSPERO database (CRD42020211046). The record and study protocol are available at https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=211046 .

Relevant publications were searched using the PubMed, Embase and PsycINFO databases to ensure coverage of all relevant research areas in the medical, public health and social science fields. The CINAHL database was also considered, but a pilot search revealed that its unique contributions were limited.

An experienced research librarian used the following keywords to develop a search strategy (Supplementary Methods ): ‘vaccination’, ‘immunization’, ‘vaccination refusal’, ‘vaccination avoidance’, ‘vaccination hesitation’, ‘vaccine hesitancy’, ‘vaccine uptake’, ‘vaccination behaviour’, ‘vaccination attitude’, ‘vaccine confidence’, ‘vaccine acceptance’ and ‘vaccine barriers’. The limitations included a publication date of between 2010 and the date of the search (14 January 2022). Conference abstracts were excluded from the search of the Embase database.

Eligibility criteria

The included studies were all published in peer-reviewed journals and written in English. All study types were eligible, except editorials and commentaries, as we sought to include original studies. Studies on animal vaccines were excluded.

The purpose of this review was to clarify the VH concept by analysing how it is operationalized. We recognized operationalizations at two main levels: conceptual and empirical. This resulted in three main groups: (1) studies describing or defining the VH concept and studies applying the concept by (2) identifying VH subpopulations and (3) measuring VH in individuals. This approach allowed comparison between conceptual and empirical operationalizations of VH.

Study selection

In the first selection round, two members of the research team used RAYYAN software to independently assess the titles and abstracts. Studies were selected when the title or abstract contained the term ‘vaccine hesitancy’. Studies were also selected if the title or abstract indicated that the full text contained further information on VH conceptualization, subpopulations or measurements. Papers without an abstract were selected for full-text screening. After double-screening, the results were de-blinded to allow the researchers to discuss their conflicting judgements until consensus was reached.

In the second selection round, the full texts were screened. The first 30% of studies were double-screened to establish a uniform method. Studies were screened on whether they met the criteria for one or more of the three categories (conceptualization, subpopulations and measurements). The category of ‘conceptualization’ included studies that describe, discuss or explore the VH concept or propose a novel VH measurement instrument. Studies falling into only the second category (subpopulations) were excluded if they merely distinguished between hesitant and non-hesitant groups, since a dichotomous grouping does not contribute to understanding of VH. The references from the included full-text articles were screened to find additional studies matching the selection criteria.

We deviated posthoc from our preregistered study protocol by adjusting the study selection criteria as follows. Initially, we also included studies containing the term ‘vaccine confidence’ (that is, with no mention or operationalization of vaccine hesitancy) as indicated in our study protocol. During the process, we realized that this deviated from our primary aim to clarify the VH concept by differentiating its related concepts. Therefore, we adapted the protocol and excluded 16 studies that were exclusively on vaccine confidence from our analysis

Data collection

The study characteristics were extracted from each of the full-text articles. Data were extracted by one researcher and verified by a second member of the research team. The variables included the first author, year of publication, research field of the first author, type of study, type of participants, number of participants, type of vaccination and country in which the study was conducted (with corresponding economic status) 447 . For the studies that do not include data collection, the country of origin was determined using the affiliation of the first author.

From the studies on VH conceptualization, text excerpts that define or describe VH or describe the relationship of VH to other concepts were extracted. These excerpts were further analysed in the qualitative analysis. From studies that describe different VH subpopulations, information about the categorization of these various subgroups was extracted, including the rationale for the distinguished subpopulations. From studies that describe VH measurements, the instrument(s) and criteria used to define VH were extracted.

Synthesis of results

The text excerpts extracted from the studies conceptualizing VH were thematically coded using ATLAS.ti software. Three research team members developed a coding book of themes and subthemes after independent coding of 30% of the studies. Thereafter, one researcher continued the coding process for the remaining studies. Any emerging new codes were discussed with the other research team members. The results were analysed qualitatively, and the predominant themes were identified by the three team members. When possible, results were grouped by research field and vaccine type to allow for comparison.

The data extracted from the studies describing VH subpopulations were summarized in a table and grouped according to the common themes identified. The data extracted from the studies describing a VH measurement were summarized in a table and grouped according to the instrument or method used. Where multiple measurement instruments are used in one study, the tool used to determine hesitancy was selected as the main instrument.

Quality assessment

The quality of each study was assessed using the MMAT 448 . This tool contains appraisal guidelines for different study types, covering the majority of the included studies. An overall score was calculated (1–5) on the basis of additional communication about the MMAT 2018 version, with higher scores indicating higher quality levels 449 . The first 20% of studies were assessed independently by two members of the research team to ensure consistency. Thereafter, one member of the research team continued the assessment.

Reporting summary

Further information on research design is available in the Nature Research Reporting Summary linked to this article.

Data availability

All data generated or analysed during this study are included in this article and its Supplementary Information . This systematic review is registered in PROSPERO (CRD42020211046).

Ten Threats to Global Health in 2019 (WHO, 2019); https://www.who.int/news-room/spotlight/ten-threats-to-global-health-in-2019

Patel, M. K. et al. Progress toward regional measles elimination - worldwide, 2000–2019. MMWR Morb. Mortal. Wkly Rep. 69 , 1700–1705 (2020).

Article   Google Scholar  

Suijkerbuijk, A. W. et al. Economic costs of measles outbreak in the Netherlands, 2013–2014. Emerg. Infect. Dis. 21 , 2067–2069 (2015).

Ghebrehewet, S. et al. The economic cost of measles: healthcare, public health and societal costs of the 2012–13 outbreak in Merseyside, UK. Vaccine 34 , 1823–1831 (2016).

Chovatiya, R. & Silverberg, J. I. Inpatient morbidity and mortality of measles in the United States. PLoS ONE 15 , e0231329 (2020).

Article   CAS   Google Scholar  

Feemster, K. A. & Szipszky, C. Resurgence of measles in the United States: how did we get here? Curr. Opin. Pediatr. 32 , 139–144 (2020).

Rechel, B., Richardson, E. & McKee, M. The Organization and Delivery of Vaccination Services in the European Union (The European Observatory on Health Systems and Policies, 2018). https://www.euro.who.int/__data/assets/pdf_file/0008/386684/vaccination-report-eng.pdf

Wilder-Smith, A. B. & Qureshi, K. Resurgence of measles in Europe: a systematic review on parental attitudes and beliefs of measles vaccine. J. Epidemiol. Glob. Health 10 , 46–58 (2020).

Bangura, J. B., Xiao, S., Qiu, D., Ouyang, F. & Chen, L. Barriers to childhood immunization in sub-Saharan Africa: a systematic review. BMC Public Health 20 , 1108 (2020).

Sweileh, W. M. Bibliometric analysis of global scientific literature on vaccine hesitancy in peer-reviewed journals (1990–2019). BMC Public Health 20 , 1252 (2020).

Frederiksen, L. S. F., Zhang, Y., Foged, C. & Thakur, A. The long road toward COVID-19 herd immunity: vaccine platform technologies and mass immunization strategies. Front. Immunol. 11 , 1817 (2020).

Randolph, H. E. & Barreiro, L. B. Herd immunity: understanding COVID-19. Immunity 52 , 737–741 (2020).

Dror, A. A. et al. Vaccine hesitancy: the next challenge in the fight against COVID-19. Eur. J. Epidemiol. 35 , 775–779 (2020).

MacDonald, N. E. Vaccine hesitancy: definition, scope and determinants. Vaccine 33 , 4161–4164 (2015).

Podsakoff, P. M., MacKenzie, S. B. & Podsakoff, N. P. Recommendations for creating better concept definitions in the organizational, behavioral, and social sciences. Organ. Res. Methods 19 , 159–203 (2016).

Shapiro, G. K. et al. A critical review of measures of childhood vaccine confidence. Curr. Opin. Immunol. 71 , 34–45 (2021).

Brewer, N. T., Chapman, G. B., Rothman, A. J., Leask, J. & Kempe, A. Increasing vaccination: putting psychological science into action. Psychol. Sci. Public Interest 18 , 149–207 (2017).

Bedford, H. et al. Vaccine hesitancy, refusal and access barriers: the need for clarity in terminology. Vaccine 36 , 6556–6558 (2018).

Orenstein, W. et al. Assessing the state of vaccine confidence in the United States: recommendations from the national vaccine advisory committee. Public Health Rep. 130 , 573–595 (2015).

Schmid, P., Rauber, D., Betsch, C., Lidolt, G. & Denker, M. L. Barriers of influenza vaccination intention and behavior - a systematic review of influenza vaccine hesitancy, 2005–2016. PLoS ONE 12 , e0170550 (2017).

Dubé, E. et al. “Nature does things well, why should we interfere?”: vaccine hesitancy among mothers. Qual. Health Res. 26 , 411–425 (2016).

Betsch, C. et al. Beyond confidence: development of a measure assessing the 5C psychological antecedents of vaccination. PLoS ONE 13 , e0208601 (2018).

Peretti-Watel, P., Larson, H. J., Ward, J. K., Schulz, W. S. & Verger, P. Vaccine hesitancy: clarifying a theoretical framework for an ambiguous notion. PLoS Curr. https://doi.org/10.1371/currents.outbreaks.6844c80ff9f5b273f34c91f71b7fc289 (2015).

Page, M. J. et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Brit. Med. J. 372 , n71 (2021).

Bertoncello, C. et al. Socioeconomic determinants in vaccine hesitancy and vaccine refusal in Italy. Vaccines 8 , 276 (2020).

Deml, M. J. et al. ‘Problem patients and physicians’ failures’: what it means for doctors to counsel vaccine hesitant patients in Switzerland. Soc. Sci. Med. 255 , 112946 (2020).

Dubé, E. et al. Vaccine hesitancy: an overview. Hum. Vaccin. Immunother. 9 , 1763–1773 (2013).

Dubé, E., Gagnon, D., Nickels, E., Jeram, S. & Schuster, M. Mapping vaccine hesitancy–country-specific characteristics of a global phenomenon. Vaccine 32 , 6649–6654 (2014).

Dubé, E. et al. Understanding vaccine hesitancy in Canada: results of a consultation study by the Canadian Immunization Research Network. PLoS ONE 11 , e0156118 (2016).

Gowda, C. & Dempsey, A. F. The rise (and fall?) of parental vaccine hesitancy. Hum. Vaccin. Immunother. 9 , 1755–1762 (2013).

Kestenbaum, L. A. & Feemster, K. A. Identifying and addressing vaccine hesitancy. Pediatr. Ann. 44 , e71–e75 (2015).

Larson, H. J. Negotiating vaccine acceptance in an era of reluctance. Hum. Vaccin. Immunother. 9 , 1779–1781 (2013).

Oduwole, E. O., Pienaar, E. D., Mahomed, H. & Wiysonge, C. S. Current tools available for investigating vaccine hesitancy: a scoping review protocol. BMJ Open 9 , e033245 (2019).

Opel, D. J. et al. Development of a survey to identify vaccine-hesitant parents: the parent attitudes about childhood vaccines survey. Hum. Vaccin. 7 , 419–425 (2011).

Smith, M. J. Promoting vaccine confidence. Infect. Dis. Clin. North Am. 29 , 759–769 (2015).

Dubé, È., Ward, J. K., Verger, P. & MacDonald, N. E. Vaccine hesitancy, acceptance, and anti-vaccination: trends and future prospects for public health. Annu Rev. Public Health 42 , 175–191 (2021).

Popper-Giveon, A. & Keshet, Y. Non-Vaccination Stage Model (NVST): the decision-making process among Israeli ultra-orthodox Jewish parents. Health https://doi.org/10.1177/1363459320988884 (2021).

Kumar, D., Chandra, R., Mathur, M., Samdariya, S. & Kapoor, N. Vaccine hesitancy: understanding better to address better. Isr. J. Health Policy Res . 5 , 2 (2016).

McIntosh, E. D., Janda, J., Ehrich, J. H., Pettoello-Mantovani, M. & Somekh, E. Vaccine hesitancy and refusal. J. Pediatr. 175 , 248–249.e1 (2016).

Díaz Crescitelli, M. E. et al. A meta-synthesis study of the key elements involved in childhood vaccine hesitancy. Public Health 180 , 38–45 (2020).

Larson, H. J. et al. Measuring vaccine hesitancy: the development of a survey tool. Vaccine 33 , 4165–4175 (2015).

Howard, M. C. A more comprehensive measure of vaccine hesitancy: creation of the Multidimensional Vaccine Hesitancy Scale (MVHS). J. Health Psychol . https://doi.org/10.1177/13591053211042062 (2021).

Turner, P. J., Larson, H., Dubé, È. & Fisher, A. Vaccine hesitancy: drivers and how the allergy community can help. J. Allergy Clin. Immunol. Pract. 9 , 3568–3574 (2021).

Freeman, D. et al. COVID-19 vaccine hesitancy in the UK: the Oxford coronavirus explanations, attitudes, and narratives survey (Oceans) II. Psychol. Med . https://doi.org/10.1017/s0033291720005188 (2020).

Su, Z. et al. A race for a better understanding of COVID-19 vaccine non-adopters. Brain Behav. Immun. Health 9 , 100159 (2020).

Sarathchandra, D., Navin, M. C., Largent, M. A. & McCright, A. M. A survey instrument for measuring vaccine acceptance. Prev. Med. 109 , 1–7 (2018).

Leask, J. et al. Communicating with parents about vaccination: a framework for health professionals. BMC Pediatr. 12 , 154 (2012).

Jankovic, S. Childhood vaccination in the twenty-first century: parental concerns and challenges for physicians. Arh. Farm. 69 , 452–468 (2019).

Hagood, E. A. & Mintzer Herlihy, S. Addressing heterogeneous parental concerns about vaccination with a multiple-source model: a parent and educator perspective. Hum. Vaccin. Immunother. 9 , 1790–1794 (2013).

Blaisdell, L. L., Gutheil, C., Hootsmans, N. A. & Han, P. K. Unknown risks: parental hesitation about vaccination. Med. Decis. Mak. 36 , 479–489 (2016).

Amin, A. B. et al. Association of moral values with vaccine hesitancy. Nat. Hum. Behav. 1 , 873–880 (2017).

Kotta, I., Kalcza-Janosi, K., Szabo, K. & Marschalko, E. E. Development and validation of the multidimensional COVID-19 vaccine hesitancy scale. Hum. Vaccin. Immunother. 18 , 1–10 (2021).

Sjögren, E., Ask, L. S., Örtqvist, Å. & Asp, M. Parental conceptions of the rotavirus vaccine during implementation in Stockholm: a phenomenographic study. J. Child Health Care 21 , 476–487 (2017).

Frew, P. M. et al. Development of a US trust measure to assess and monitor parental confidence in the vaccine system. Vaccine 37 , 325–332 (2019).

Smith, J. C., Appleton, M. & MacDonald, N. E. Building confidence in vaccines. Adv. Exp. Med. Biol. 764 , 81–98 (2013).

Larson, H. J., Schulz, W. S., Tucker, J. D. & Smith, D. M. Measuring vaccine confidence: introducing a global vaccine confidence index. PLoS Curr. https://doi.org/10.1371/currents.outbreaks.ce0f6177bc97332602a8e3fe7d7f7cc4 (2015).

Ramanadhan, S., Galarce, E., Xuan, Z., Alexander-Molloy, J. & Viswanath, K. Addressing the vaccine hesitancy continuum: an audience segmentation analysis of american adults who did not receive the 2009 H1N1 vaccine. Vaccines 3 , 556–578 (2015).

Rossen, I., Hurlstone, M. J., Dunlop, P. D. & Lawrence, C. Accepters, fence sitters, or rejecters: moral profiles of vaccination attitudes. Soc. Sci. Med. 224 , 23–27 (2019).

Rozbroj, T., Lyons, A. & Lucke, J. Vaccine-hesitant and vaccine-refusing parents’ reflections on the way parenthood changed their attitudes to vaccination. J. Community Health 45 , 63–72 (2020).

Schwartz, J. L. & Caplan, A. L. Vaccination refusal: ethics, individual rights, and the common good. Prim. Care 38 , 717–728 (2011).

Shay, L. A. et al. Parent-provider communication of HPV vaccine hesitancy. Pediatrics 141 , e20172312 (2018).

Oladejo, O. et al. Comparative analysis of the Parent Attitudes about Childhood Vaccines (PACV) short scale and the five categories of vaccine acceptance identified by Gust et al. Vaccine 34 , 4964–4968 (2016).

Bouchez, M. et al. Physicians’ decision processes about the HPV vaccine: a qualitative study. Vaccine 39 , 521–528 (2021).

Elwy, A. R. et al. Vaccine hesitancy as an opportunity for engagement: a rapid qualitative study of patients and employees in the U.S. Veterans Affairs healthcare system. Vaccine 9 , 100116 (2021).

CAS   Google Scholar  

Paris, C. et al. COVID-19 vaccine hesitancy among healthcare workers. Infect. Dis. Now 51 , 484–487 (2021).

Tram, K. H. et al. Deliberation, dissent, and distrust: understanding distinct drivers of COVID-19 vaccine hesitancy in the United States. Clin. Infect. Dis. https://doi.org/10.1093/cid/ciab633 (2021).

Vulpe, S. N. & Rughiniş, C. Social amplification of risk and “probable vaccine damage”: a typology of vaccination beliefs in 28 European countries. Vaccine 39 , 1508–1515 (2021).

Wang, M. W. et al. COVID-19 vaccination acceptance among healthcare workers and non-healthcare workers in china: a survey. Front. Public Health 9 , 709056 (2021).

Shapiro, G. K. et al. Using an integrated conceptual framework to investigate parents’ HPV vaccine decision for their daughters and sons. Prev. Med. 116 , 203–210 (2018).

Tatar, O., Shapiro, G. K., Perez, S., Wade, K. & Rosberger, Z. Using the precaution adoption process model to clarify human papillomavirus vaccine hesitancy in Canadian parents of girls and parents of boys. Hum. Vaccin. Immunother. 15 , 1803–1814 (2019).

Dubov, A. et al. Predictors of COVID-19 vaccine acceptance and hesitancy among healthcare workers in Southern California: not just “anti” vs. “pro” vaccine. Vaccines 9 , 1428 (2021).

Brunelli, L. et al. Parental trust and beliefs after the discovery of a six-year-long failure to vaccinate. Hum. Vaccin. Immunother. 17 , 583–587 (2020).

Armiento, R. et al. Impact of Australian mandatory ‘No Jab, No Pay’ and ‘No Jab, No Play’ immunisation policies on immunisation services, parental attitudes to vaccination and vaccine uptake, in a tertiary paediatric hospital, the Royal Children’s Hospital, Melbourne. Vaccine 38 , 5231–5240 (2020).

Berry, N. J. et al. Sharing knowledge about immunisation (SKAI): an exploration of parents’ communication needs to inform development of a clinical communication support intervention. Vaccine 36 , 6480–6490 (2018).

Bocquier, A. et al. Social differentiation of vaccine hesitancy among French parents and the mediating role of trust and commitment to health: a nationwide cross-sectional study. Vaccine 36 , 7666–7673 (2018).

Chang, K. & Lee, S. Y. Why do some Korean parents hesitate to vaccinate their children? Epidemiol. Health 41 , e2019031 (2019).

Danchin, M. H. et al. Vaccine decision-making begins in pregnancy: correlation between vaccine concerns, intentions and maternal vaccination with subsequent childhood vaccine uptake. Vaccine 36 , 6473–6479 (2018).

Forbes, T. A., McMinn, A., Crawford, N., Leask, J. & Danchin, M. Vaccination uptake by vaccine-hesitant parents attending a specialist immunization clinic in Australia. Hum. Vaccin. Immunother. 11 , 2895–2903 (2015).

Glanternik, J. R. et al. Evaluation of a vaccine-communication tool for physicians. J. Pediatr. 224 , 72–78.e1 (2020).

McDonald, P. et al. Exploring California’s new law eliminating personal belief exemptions to childhood vaccines and vaccine decision-making among homeschooling mothers in California. Vaccine 37 , 742–750 (2019).

Noyman-Veksler, G., Greenberg, D., Grotto, I. & Shahar, G. Parents’ malevolent personification of mass vaccination solidifies vaccine hesitancy. J. Health Psychol . https://doi.org/10.1177/1359105320903475 (2020).

Duong, M. C., Nguyen, H. T. & Duong, M. Evaluating COVID-19 vaccine hesitancy: a qualitative study from Vietnam. Diabetes Metab. Syndr. 16 , 102363 (2022).

Gatto, N. M. et al. Correlates of COVID-19 vaccine acceptance, hesitancy and refusal among employees of a safety net California County health system with an early and aggressive vaccination program: results from a cross-sectional survey. Vaccines 9 , 1152 (2021).

Moore, J. X. et al. Correlates of COVID-19 vaccine hesitancy among a community sample of African Americans living in the southern United States. Vaccines 9 , 879 (2021).

Murphy, J. et al. Psychological characteristics associated with COVID-19 vaccine hesitancy and resistance in Ireland and the United Kingdom. Nat. Commun. 12 , 29 (2021).

Musa, S. et al. A qualitative interview study with parents to identify barriers and drivers to childhood vaccination and inform public health interventions. Hum. Vaccin. Immunother. 17 , 3023–3033 (2021).

Steffens, M. S., Bullivant, B., Bolsewicz, K. T., King, C. & Beard, F. “to protect myself, my friends, family, workmates and patients… and to play my part”: COVID-19 vaccination perceptions among health and aged care workers in New South Wales, Australia. Int. J. Environ. Res. Public Health 18 , 8954 (2021).

Tavolacci, M. P., Dechelotte, P. & Ladner, J. COVID-19 vaccine acceptance, hesitancy, and resistancy among university students in France. Vaccines 9 , 654 (2021).

Buttenheim, A. M. et al. Vaccine exemption requirements and parental vaccine attitudes: an online experiment. Vaccine 38 , 2620–2625 (2020).

Corben, P. & Leask, J. Vaccination hesitancy in the antenatal period: a cross-sectional survey. BMC Public Health 18 , 566 (2018).

Gagneur, A. et al. Promoting vaccination in maternity wards ─ motivational interview technique reduces hesitancy and enhances intention to vaccinate, results from a multicentre non-controlled pre- and post-intervention RCT-nested study, Quebec, March 2014 to February 2015. Eur. Surveill. 24 , 1800641 (2019).

Lau, L. H. W., Lee, S. S. & Wong, N. S. The continuum of influenza vaccine hesitancy among nursing professionals in Hong Kong. Vaccine 38 , 6785–6793 (2020).

Nekrasova, E. et al. Vaccine hesitancy and influenza beliefs among parents of children requiring a second dose of influenza vaccine in a season: an American Academy of Pediatrics (AAP) Pediatric Research in Office Settings (PROS) study. Hum. Vaccin. Immunother. 16 , 1070–1077 (2020).

Verger, P. et al. Prevalence and correlates of vaccine hesitancy among general practitioners: a cross-sectional telephone survey in France, April to July 2014. Eur. Surveill. 21 , 30406 (2016).

Bianco, A. et al. Parental COVID-19 vaccine hesitancy: a cross-sectional survey in Italy. Expert Rev. Vaccines 21 , 541–547 (2022).

Edwards, B., Biddle, N., Gray, M. & Sollis, K. COVID-19 vaccine hesitancy and resistance: correlates in a nationally representative longitudinal survey of the Australian population. PLoS ONE 16 , e0248892 (2021).

Hsieh, Y. L., Rak, S., SteelFisher, G. K. & Bauhoff, S. Effect of the suspension of the J&J COVID-19 vaccine on vaccine hesitancy in the United States. Vaccine 40 , 424–427 (2022).

Umakanthan, S., Patil, S., Subramaniam, N. & Sharma, R. COVID-19 vaccine hesitancy and resistance in India explored through a population-based longitudinal survey. Vaccines 9 , 1064 (2021).

Zheng, W. et al. COVID-19 vaccine uptake and hesitancy among HIV-infected men who have sex with men in mainland China: a cross-sectional survey. Hum. Vaccin. Immunother. 17 , 4971–4981 (2021).

Baniak, L. M., Luyster, F. S., Raible, C. A., McCray, E. E. & Strollo, P. J. COVID-19 vaccine hesitancy and uptake among nursing staff during an active vaccine rollout. Vaccines 9 , 858 (2021).

Bou Hamdan, M., Singh, S., Polavarapu, M., Jordan, T. R. & Melhem, N. M. COVID-19 vaccine hesitancy among university students in Lebanon. Epidemiol. Infect. 149 , e242 (2021).

Castellano-Tejedor, C., Torres-Serrano, M. & Cencerrado, A. Unveiling Associations of COVID-19 vaccine acceptance, hesitancy, and resistance: a cross-sectional community-based adult survey. Int. J. Environ. Res. Public Health 18 , 12348 (2021).

Freeman, D. et al. Effects of different types of written vaccination information on COVID-19 vaccine hesitancy in the UK (OCEANS-III): a single-blind, parallel-group, randomised controlled trial. Lancet Public Health 6 , e416–e427 (2021).

Janssen, C. et al. Hesitancy towards COVID-19 vaccination among healthcare workers: a multi-centric survey in France. Vaccines 9 , 547 (2021).

Khaled, S. M. et al. Prevalence and potential determinants of COVID-19 vaccine hesitancy and resistance in Qatar: results from a nationally representative survey of Qatari nationals and migrants between December 2020 and January 2021. Vaccines 9 , 471 (2021).

Montagni, I. et al. Acceptance of a Covid-19 vaccine is associated with ability to detect fake news and health literacy. J. Public Health https://doi.org/10.1093/pubmed/fdab028 (2021).

Qunaibi, E., Basheti, I., Soudy, M. & Sultan, I. Hesitancy of Arab healthcare workers towards COVID-19 vaccination: a large-scale multinational study. Vaccines 9 , 446 (2021).

Reno, C. et al. Enhancing COVID-19 vaccines acceptance: results from a survey on vaccine hesitancy in northern Italy. Vaccines 9 , 378 (2021).

Rodríguez-Blanco, N. et al. Willingness to be vaccinated against COVID-19 in Spain before the start of vaccination: a cross-sectional study. Int. J. Environ. Res. Public Health 18 , 5272 (2021).

Leung, C. L. K. et al. Profiling vaccine believers and skeptics in nurses: a latent profile analysis. Int. J. Nurs. Stud. 126 , 104142 (2021).

Salerno, L., Craxì, L., Amodio, E. & Lo Coco, G. Factors affecting hesitancy to mRNA and viral vector COVID-19 vaccines among college students in Italy. Vaccines 9 , 927 (2021).

Zona, S. et al. Anti-COVID vaccination for adolescents: a survey on determinants of vaccine parental hesitancy. Vaccines 9 , 1309 (2021).

Alabbad, A. A., Alsaad, A. K., Al Shaalan, M. A., Alola, S. & Albanyan, E. A. Prevalence of influenza vaccine hesitancy at a tertiary care hospital in Riyadh, Saudi Arabia. J. Infect. Public Health 11 , 491–499 (2018).

Barello, S., Nania, T., Dellafiore, F., Graffigna, G. & Caruso, R. ‘Vaccine hesitancy’ among university students in Italy during the COVID-19 pandemic. Eur. J. Epidemiol. 35 , 781–783 (2020).

Bogart, L. M. et al. COVID-19 related medical mistrust, health impacts, and potential vaccine hesitancy among Black Americans living with HIV. J. Acquir. Immune Defic. Syndr. 86 , 200–207 (2020).

Brown, A. L. et al. Vaccine confidence and hesitancy in Brazil. Cad. Saude Publica 34 , e00011618 (2018).

Byström, E., Lindstrand, A., Bergström, J., Riesbeck, K. & Roth, A. Confidence in the National Immunization Program among parents in Sweden 2016 – a cross-sectional survey. Vaccine 38 , 3909–3917 (2020).

Costantino, C. et al. Determinants of vaccine hesitancy and effectiveness of vaccination counseling interventions among a sample of the general population in Palermo, Italy. Hum. Vaccin. Immunother. 16 , 2415–2421 (2020).

Deas, J., Bean, S. J., Sokolovska, I. & Fautin, C. Childhood vaccine attitudes and information sources among Oregon parents and guardians. Health Promot. Pract. 20 , 529–538 (2019).

Dempsey, A. F., Pyrzanowski, J., Campagna, E. J., Lockhart, S. & O’Leary, S. T. Parent report of provider HPV vaccine communication strategies used during a randomized, controlled trial of a provider communication intervention. Vaccine 37 , 1307–1312 (2019).

Detoc, M. et al. Intention to participate in a COVID-19 vaccine clinical trial and to get vaccinated against COVID-19 in France during the pandemic. Vaccine 38 , 7002–7006 (2020).

Du, F. et al. The determinants of vaccine hesitancy in China: a cross-sectional study following the Changchun Changsheng vaccine incident. Vaccine 38 , 7464–7471 (2020).

Dubé, E., Gagnon, D., Zhou, Z. & Deceuninck, G. Parental vaccine hesitancy in Quebec (Canada). PLoS Curr . https://doi.org/10.1371/currents.outbreaks.9e239605f4d320c6ad27ce2aea5aaad2 (2016).

Fisher, K. A. et al. Attitudes toward a potential SARS-CoV-2 vaccine: a survey of U.S. adults. Ann. Intern. Med. 173 , 964–973 (2020).

Gesser-Edelsburg, A., Walter, N., Shir-Raz, Y., Sassoni Bar-Lev, O. & Rosenblat, S. The behind-the-scenes activity of parental decision-making discourse regarding childhood vaccination. Am. J. Infect. Control 45 , 267–271 (2017).

Gowda, C., Schaffer, S. E., Kopec, K., Markel, A. & Dempsey, A. F. A pilot study on the effects of individually tailored education for MMR vaccine-hesitant parents on MMR vaccination intention. Hum. Vaccin. Immunother. 9 , 437–445 (2013).

Gowda, C., Schaffer, S. E., Kopec, K., Markel, A. & Dempsey, A. F. Does the relative importance of MMR vaccine concerns differ by degree of parental vaccine hesitancy?: an exploratory study. Hum. Vaccin. Immunother. 9 , 430–436 (2013).

Hadjipanayis, A. et al. Vaccine confidence among parents: large scale study in eighteen European countries. Vaccine 38 , 1505–1512 (2020).

Hirth, J. M., Fuchs, E. L., Chang, M., Fernandez, M. E. & Berenson, A. B. Variations in reason for intention not to vaccinate across time, region, and by race/ethnicity, NIS-Teen (2008–2016). Vaccine 37 , 595–601 (2019).

Karlsson, L. C. et al. The association between vaccination confidence, vaccination behavior, and willingness to recommend vaccines among Finnish healthcare workers. PLoS ONE 14 , e0224330 (2019).

Khodadadi, A. B., Redden, D. T. & Scarinci, I. C. HPV vaccination hesitancy among latina immigrant mothers despite physician recommendation. Ethn. Dis. 30 , 661–670 (2020).

Melot, B. et al. Knowledge, attitudes and practices about vaccination in Trentino, Italy in 2019. Hum. Vaccin. Immunother. 17 , 259–268 (2020).

Mereu, N. et al. Vaccination attitude and communication in early settings: an exploratory study. Vaccines 8 , 701 (2020).

Opel, D. J. et al. Characterizing providers’ immunization communication practices during health supervision visits with vaccine-hesitant parents: a pilot study. Vaccine 30 , 1269–1275 (2012).

Özceylan, G., Toprak, D. & Esen, E. S. Vaccine rejection and hesitation in Turkey. Hum. Vaccin. Immunother. 16 , 1034–1039 (2020).

Pahud, B. et al. A randomized controlled trial of an online immunization curriculum. Vaccine 38 , 7299–7307 (2020).

Quinn, S. C. et al. Breaking down the monolith: understanding flu vaccine uptake among African Americans. SSM Popul. Health 4 , 25–36 (2018).

Quinn, S. C. et al. Exploring racial influences on flu vaccine attitudes and behavior: results of a national survey of White and African American adults. Vaccine 35 , 1167–1174 (2017).

Quinn, S. C., Jamison, A. M., An, J., Hancock, G. R. & Freimuth, V. S. Measuring vaccine hesitancy, confidence, trust and flu vaccine uptake: results of a national survey of White and African American adults. Vaccine 37 , 1168–1173 (2019).

Repalust, A., Šević, S., Rihtar, S. & Štulhofer, A. Childhood vaccine refusal and hesitancy intentions in Croatia: insights from a population-based study. Psychol. Health Med. 22 , 1045–1055 (2017).

Rey, D. et al. Vaccine hesitancy in the French population in 2016, and its association with vaccine uptake and perceived vaccine risk-benefit balance. Eur. Surveill. 23 , 17–00816 (2018).

Santibanez, T. A. et al. Parental vaccine hesitancy and childhood influenza vaccination. Pediatrics 146 , e2020007609 (2020).

Schollin Ask, L. et al. Receiving early information and trusting Swedish child health centre nurses increased parents’ willingness to vaccinate against rotavirus infections. Acta Paediatr. 106 , 1309–1316 (2017).

Suryadevara, M., Handel, A., Bonville, C. A., Cibula, D. A. & Domachowske, J. B. Pediatric provider vaccine hesitancy: an under-recognized obstacle to immunizing children. Vaccine 33 , 6629–6634 (2015).

Taylor, S. et al. A proactive approach for managing COVID-19: the importance of understanding the motivational roots of vaccination hesitancy for SARS-CoV2. Front. Psychol. 11 , 575950 (2020).

Tran, B. X. et al. Media representation of vaccine side effects and its impact on utilization of vaccination services in Vietnam. Patient Prefer. Adherence 12 , 1717–1728 (2018).

Wilson, R. et al. Vaccine hesitancy and self-vaccination behaviors among nurses in southeastern France. Vaccine 38 , 1144–1151 (2020).

Yu, W. et al. Two media-reported vaccine events in China from 2013 to 2016: impact on confidence and vaccine utilization. Vaccine 38 , 5541–5547 (2020).

Abdel-Qader, D. H. et al. Pharmacists-physicians collaborative intervention to reduce vaccine hesitancy and resistance: a randomized controlled trial. Vaccine X 10 , 100135 (2022).

Abedin, M. et al. Willingness to vaccinate against COVID-19 among Bangladeshi adults: understanding the strategies to optimize vaccination coverage. PLoS ONE 16 , e0250495 (2021).

Abou Leila, R., Salamah, M. & El-Nigoumi, S. Reducing COVID-19 vaccine hesitancy by implementing organizational intervention in a primary care setting in Bahrain. Cureus 13 , e19282 (2021).

Google Scholar  

Aguilar Ticona, J. P. et al. Willingness to get the COVID-19 vaccine among residents of slum settlements. Vaccines 9 , 951 (2021).

Ahmad, K. K. Coronavirus disease 2019 vaccine hesitancy in the Kurdistan region: a cross-sectional national survey. Arch. Razi Inst. 76 , 751–759 (2021).

Al-Ayyadhi, N., Ramadan, M. M., Al-Tayar, E., Al-Mathkouri, R. & Al-Awadhi, S. Determinants of hesitancy towards COVID-19 vaccines in State of Kuwait: an exploratory internet-based survey. Risk Manage. Healthc. Policy 14 , 4967–4981 (2021).

Alfieri, N. L. et al. Parental COVID-19 vaccine hesitancy for children: vulnerability in an urban hotspot. BMC Public Health 21 , 1662 (2021).

Alibrahim, J. & Awad, A. COVID-19 vaccine hesitancy among the public in Kuwait: a cross-sectional survey. Int. J. Environ. Res. Public Health 18 , 8836 (2021).

Allington, D., McAndrew, S., Moxham-Hall, V. & Duffy, B. Coronavirus conspiracy suspicions, general vaccine attitudes, trust and coronavirus information source as predictors of vaccine hesitancy among UK residents during the COVID-19 pandemic. Psychol. Med . https://doi.org/10.1017/s0033291721001434 (2021).

Al-Sanafi, M. & Sallam, M. Psychological determinants of COVID-19 vaccine acceptance among healthcare workers in Kuwait: a cross-sectional study using the 5C and vaccine conspiracy beliefs scales. Vaccines 9 , 701 (2021).

Alshahrani, S. M. et al. Acceptability of COVID-19 vaccination in Saudi Arabia: a cross-sectional study using a web-based survey. Hum. Vaccin. Immunother. 17 , 3338–3347 (2021).

Al-Wutayd, O., Khalil, R. & Rajar, A. B. Sociodemographic and behavioral predictors of COVID-19 vaccine hesitancy in Pakistan. J. Multidiscip. Healthc. 14 , 2847–2856 (2021).

Alzeer, A. A. et al. The influence of demographics on influenza vaccine awareness and hesitancy among adults visiting educational hospital in Saudi Arabia. Saudi Pharm. J. 29 , 188–193 (2021).

Alzubaidi, H. et al. A mixed-methods study to assess COVID-19 vaccination acceptability among university students in the United Arab Emirates. Hum. Vaccin. Immunother. 17 , 4074–4082 (2021).

Anandraj, J., Krishnamoorthy, Y., Sivanantham, P., Gnanadas, J. & Kar, S. S. Impact of second wave of COVID-19 pandemic on the hesitancy and refusal of COVID-19 vaccination in Puducherry, India: a longitudinal study. Hum. Vaccin. Immunother. 17 , 5024–5029 (2021).

Arvanitis, M. et al. Factors associated with COVID-19 vaccine trust and hesitancy among adults with chronic conditions. Prev. Med. Rep. 24 , 101484 (2021).

Baccolini, V. et al. COVID-19 vaccine hesitancy among Italian university students: a cross-sectional survey during the first months of the vaccination campaign. Vaccines 9 , 1292 (2021).

Bacon, A. M. & Taylor, S. Vaccination hesitancy and conspiracy beliefs in the UK During the SARS-COV-2 (COVID-19) pandemic. Int. J. Behav. Med. 29 , 448–455 (2022).

Badr, H. et al. Overcoming COVID-19 vaccine hesitancy: insights from an online population-based survey in the United States. Vaccines 9 , 1100 (2021).

Batty, G. D., Deary, I. J., Fawns-Ritchie, C., Gale, C. R. & Altschul, D. Pre-pandemic cognitive function and COVID-19 vaccine hesitancy: cohort study. Brain Behav. Immun. 96 , 100–105 (2021).

Benham, J. L. et al. COVID-19 vaccine-related attitudes and beliefs in Canada: national cross-sectional survey and cluster analysis. JMIR Public Health Surveill. 7 , e30424 (2021).

Bolatov, A. K., Seisembekov, T. Z., Askarova, A. Z. & Pavalkis, D. Barriers to COVID-19 vaccination among medical students in Kazakhstan: development, validation, and use of a new COVID-19 vaccine hesitancy scale. Hum. Vaccin. Immunother. 17 , 4982–4992 (2021).

Butter, S., McGlinchey, E., Berry, E. & Armour, C. Psychological, social, and situational factors associated with COVID-19 vaccination intentions: A study of UK key workers and non-key workers. Br. J. Health Psychol. 27 , 13–29 (2022).

Chandani, S. et al. COVID-19 vaccination hesitancy in India: state of the nation and priorities for research. Brain Behav. Immun. Health 18 , 100375 (2021).

Chaudhary, F. A. et al. Factors influencing COVID-19 vaccine hesitancy and acceptance among the Pakistani population. Hum. Vaccin. Immunother. 17 , 3365–3370 (2021).

Chen, H. et al. Health belief model perspective on the control of COVID-19 vaccine hesitancy and the promotion of vaccination in China: web-based cross-sectional study. J. Med. Internet Res. 23 , e29329 (2021).

Cherian, V., Saini, N. K., Sharma, A. K. & Philip, J. Prevalence and predictors of vaccine hesitancy in an urbanized agglomeration of New Delhi, India. J. Public Health 44 , 70–76 (2022).

Contoli, B. et al. What is the willingness to receive vaccination against COVID-19 among the elderly in Italy? Data from the PASSI d’Argento surveillance system. Front. Public Health 9 , 736976 (2021).

Cordina, M., Lauri, M. A. & Lauri, J. Attitudes towards COVID-19 vaccination, vaccine hesitancy and intention to take the vaccine. Pharm. Pract. 19 , 2317 (2021).

Costantino, A. et al. COVID-19 vaccine acceptance among liver transplant recipients. Vaccines 9 , 1314 (2021).

Costantino, A. et al. COVID-19 vaccine: a survey of hesitancy in patients with celiac disease. Vaccines 9 , 511 (2021).

Coughenour, C., Gakh, M., Sharma, M., Labus, B. & Chien, L. C. Assessing determinants of COVID-19 vaccine hesitancy in Nevada. Health Secur. 19 , 592–604 (2021).

Crane, M. A., Faden, R. R. & Romley, J. A. Disparities in county COVID-19 vaccination rates linked to disadvantage and hesitancy. Health Aff. 40 , 1792–1796 (2021).

Danabal, K. G. M., Magesh, S. S., Saravanan, S. & Gopichandran, V. Attitude towards COVID 19 vaccines and vaccine hesitancy in urban and rural communities in Tamil Nadu, India - a community based survey. BMC Health Serv. Res. 21 , 994 (2021).

Doherty, I. A. et al. COVID-19 vaccine hesitancy in underserved communities of North Carolina. PLoS ONE 16 , e0248542 (2021).

Du, F. et al. Access to vaccination information and confidence/hesitancy towards childhood vaccination: a cross-sectional survey in China. Vaccines 9 , 201 (2021).

Du, M., Tao, L. & Liu, J. The association between risk perception and COVID-19 vaccine hesitancy for children among reproductive women in China: an online survey. Front. Med. 8 , 741298 (2021).

Ebrahimi, O. V. et al. Risk, trust, and flawed assumptions: vaccine hesitancy during the COVID-19 pandemic. Front. Public Health 9 , 700213 (2021).

Ehde, D. M., Roberts, M. K., Humbert, A. T., Herring, T. E. & Alschuler, K. N. COVID-19 vaccine hesitancy in adults with multiple sclerosis in the United States: a follow up survey during the initial vaccine rollout in 2021. Mult. Scler. Relat. Disord. 54 , 103163 (2021).

Ekstrand, M. L. et al. COVID-19 vaccine hesitancy among PLWH in South India: implications for vaccination campaigns. J. Acquir. Immune Defic. Syndr. 88 , 421–425 (2021).

Elizondo-Alzola, U. et al. Vaccine hesitancy among paediatric nurses: prevalence and associated factors. PLoS ONE 16 , e0251735 (2021).

Emerson, E. et al. Vaccine hesitancy among working-age adults with/without disability in the UK. Public Health 200 , 106–108 (2021).

Epstein, S. et al. Vaccination Against SARS-CoV-2 in neuroinflammatory disease: early safety/tolerability data. Mult. Scler. Relat. Disord. 57 , 103433 (2021).

Eyllon, M. et al. Associations between psychiatric morbidity and COVID-19 vaccine hesitancy: an analysis of electronic health records and patient survey. Psychiatry Res. 307 , 114329 (2022).

Fares, S., Elmnyer, M. M., Mohamed, S. S. & Elsayed, R. COVID-19 vaccination perception and attitude among healthcare workers in Egypt. J. Prim. Care Community Health 12 , 21501327211013303 (2021).

Fayed, A. A. et al. Willingness to receive the COVID-19 and seasonal influenza vaccines among the Saudi population and vaccine uptake during the initial stage of the national vaccination campaign: a cross-sectional survey. Vaccines 9 , 765 (2021).

Fazel, M. et al. Willingness of children and adolescents to have a COVID-19 vaccination: results of a large whole schools survey in England. eClinicalMedicine 40 , 101144 (2021).

Fernández-Penny, F. E. et al. COVID-19 vaccine hesitancy among patients in two urban emergency departments. Acad. Emerg. Med . 28 , 1100–1107 (2021).

Fisher, K. A. et al. Preferences for COVID-19 vaccination information and location: associations with vaccine hesitancy, race and ethnicity. Vaccine 39 , 6591–6594 (2021).

Gatwood, J., McKnight, M., Fiscus, M., Hohmeier, K. C. & Chisholm-Burns, M. Factors influencing likelihood of COVID-19 vaccination: a survey of Tennessee adults. Am. J. Health Syst. Pharm. 78 , 879–889 (2021).

Gbeasor-Komlanvi, F. A. et al. Prevalence and factors associated with COVID-19 vaccine hesitancy in health professionals in Togo, 2021. Public Health Pract. 2 , 100220 (2021).

Gendler, Y. & Ofri, L. Investigating the influence of vaccine literacy, vaccine perception and vaccine hesitancy on Israeli parents’ acceptance of the COVID-19 vaccine for their children: a cross-sectional study. Vaccines 9 (2021).

Gentile, A. et al. Vaccine hesitancy in Argentina: validation of WHO scale for parents. Vaccine 39 , 4611–4619 (2021).

Gerretsen, P. et al. Individual determinants of COVID-19 vaccine hesitancy. PLoS ONE 16 , e0258462 (2021).

Gerretsen, P. et al. Vaccine hesitancy is a barrier to achieving equitable herd immunity among racial minorities. Front. Med. 8 , 668299 (2021).

Griva, K. et al. Evaluating rRates and determinants of COVID-19 vaccine hesitancy for adults and children in the Singapore population: Strengthening Our Community’s Resilience against Threats from Emerging Infections (SOCRATEs) Cohort. Vaccines 9 , 1415 (2021).

Guaraldi, F. et al. Rate and predictors of hesitancy toward SARS-CoV-2 vaccine among type 2 diabetic patients: results from an Italian survey. Vaccines 9 , 460 (2021).

Hara, M., Ishibashi, M., Nakane, A., Nakano, T. & Hirota, Y. Differences in COVID-19 vaccine acceptance, hesitancy, and confidence between healthcare workers and the general population in Japan. Vaccines 9 , 1389 (2021).

Holeva, V., Parlapani, E., Nikopoulou, V. A., Nouskas, I. & Diakogiannis, I. COVID-19 vaccine hesitancy in a sample of Greek adults. Psychol. Health Med. 27 , 113–119 (2022).

Hong, J. et al. Knowledge about, attitude and acceptance towards, and predictors of intention to receive the COVID-19 vaccine among cancer patients in eastern China: a cross-sectional survey. J. Integr. Med. 20 , 34–44 (2021).

Hossain, M. B. et al. COVID-19 vaccine hesitancy among the adult population in Bangladesh: a nationwide cross-sectional survey. PLoS ONE 16 , e0260821 (2021).

Hossain, M. B. et al. Health belief model, theory of planned behavior, or psychological antecedents: what predicts covid-19 vaccine hesitancy better among the Bangladeshi adults? Front. Public Health 9 , 711066 (2021).

Huang, H. et al. COVID-19 vaccine uptake, acceptance, and hesitancy among persons with mental disorders during the second stage of China’s nationwide vaccine rollout. Front. Med. 8 , 761601 (2021).

Hussain, S. et al. Perceptions regarding COVID-19 vaccination among a representative Pakistani population coming to tertiary care cardiac hospital. Cureus 13 , e18654 (2021).

Hwang, S. E., Kim, W. H. & Heo, J. Socio-demographic, psychological, and experiential predictors of COVID-19 vaccine hesitancy in South Korea, October–December 2020. Hum. Vaccin. Immunother. 18 , 1–8 (2021).

Hyland, P. et al. Detecting and describing stability and change in COVID-19 vaccine receptibility in the United Kingdom and Ireland. PLoS ONE 16 , e0258871 (2021).

Issanov, A., Akhmetzhanova, Z., Riethmacher, D. & Aljofan, M. Knowledge, attitude, and practice toward COVID-19 vaccination in Kazakhstan: a cross-sectional study. Hum. Vaccin. Immunother. 17 , 3394–3400 (2021).

Jain, J. et al. COVID-19 vaccine hesitancy among medical students in India. Epidemiol. Infect. 149 , e132 (2021).

Kanyike, A. M. et al. Acceptance of the coronavirus disease-2019 vaccine among medical students in Uganda. Trop. Med. Health 49 , 37 (2021).

Kateeb, E. et al. Predictors of willingness to receive COVID-19 vaccine: cross-sectional study of Palestinian dental students. Vaccines 9 , 954 (2021).

Khairat, S., Zou, B. & Adler-Milstein, J. Factors and reasons associated with low COVID-19 vaccine uptake among highly hesitant communities in the US. Am. J. Infect. Control 50 , 262–267 (2022).

Khan, M. S. R., Watanapongvanich, S. & Kadoya, Y. COVID-19 vaccine hesitancy among the younger generation in Japan. Int. J. Environ. Res. Public Health 18 , 11702 (2021).

Khodadadi, A. B., Hansen, B., Kim, Y. I. & Scarinci, I. C. Latinx immigrant mothers’ perceived self-efficacy and intentions regarding human papillomavirus vaccination of their daughters. Womens Health Issues 32 , 293–300 (2022).

King, W. C., Rubinstein, M., Reinhart, A. & Mejia, R. Time trends, factors associated with, and reasons for COVID-19 vaccine hesitancy: a massive online survey of US adults from January–May 2021. PLoS ONE 16 , e0260731 (2021).

King, W. C., Rubinstein, M., Reinhart, A. & Mejia, R. COVID-19 vaccine hesitancy January–May 2021 among 18–64 year old US adults by employment and occupation. Prev. Med. Rep. 24 , 101569 (2021).

Ko, T., Dendle, C., Woolley, I., Morand, E. & Antony, A. SARS-COV-2 vaccine acceptance in patients with rheumatic diseases: a cross-sectional study. Hum. Vaccin. Immunother. 17 , 4048–4056 (2021).

Kollamparambil, U., Oyenubi, A. & Nwosu, C. COVID19 vaccine intentions in South Africa: health communication strategy to address vaccine hesitancy. BMC Public Health 21 , 2113 (2021).

Kozak, A. & Nienhaus, A. COVID-19 vaccination: status and willingness to be vaccinated among employees in health and welfare care in Germany. Int. J. Environ. Res. Public Health 18 , 6688 (2021).

Krishnamurthy, K. et al. COVID-19 vaccine intent among health care professionals of Queen Elizabeth Hospital, Barbados. J. Multidiscip. Healthc. 14 , 3309–3319 (2021).

Ku, L. The association of social factors and health insurance coverage with COVID-19 vaccinations and hesitancy, July 2021. J. Gen. Intern. Med. 37 , 409–414 (2022).

Kuhn, R. et al. COVID-19 vaccine access and attitudes among people experiencing homelessness from pilot mobile phone survey in Los Angeles, CA. PLoS ONE 16 , e0255246 (2021).

Kumar, R., Alabdulla, M., Elhassan, N. M. & Reagu, S. M. Qatar healthcare workers’ COVID-19 vaccine hesitancy and attitudes: a national cross-sectional survey. Front. Public Health 9 , 727748 (2021).

Lang, R. et al. COVID-19 vaccine attitudes and beliefs: a Canadian national cross-sectional survey and cluster analysis. JMIR Public Health Surveill. 7 , e30424 (2021).

Lee, M. & You, M. Direct and indirect associations of media use with COVID-19 vaccine hesitancy in South Korea: cross-sectional web-based survey. J. Med. Internet Res. 24 , e32329 (2022).

Li, M. et al. Hesitancy toward COVID-19 vaccines among medical students in Southwest China: a cross-sectional study. Hum. Vaccin. Immunother. 17 , 4021–4027 (2021).

Liddell, B. J. et al. Factors associated with COVID-19 vaccine hesitancy amongst refugees in Australia. Eur. J. Psychotraumatol. 12 , 1997173 (2021).

Lim, L. J., Lim, A. J. W., Fong, K. K. & Lee, C. G. Sentiments regarding COVID-19 vaccination among graduate students in Singapore. Vaccines 9 , 1141 (2021).

Liu, R. & Li, G. M. Hesitancy in the time of coronavirus: temporal, spatial, and sociodemographic variations in COVID-19 vaccine hesitancy. SSM Popul. Health 15 , 100896 (2021).

Longchamps, C. et al. COVID-19 vaccine hesitancy among persons living in homeless shelters in France. Vaccine 39 , 3315–3318 (2021).

Luk, T. T. et al. Prevalence and determinants of SARS-CoV-2 vaccine hesitancy in Hong Kong: a population-based survey. Vaccine 39 , 3602–3607 (2021).

Luo, H. et al. Willingness to get a COVID-19 vaccine and reasons for hesitancy among medicare beneficiaries: results from a national survey. J. Public Health Manage. Pract. 28 , 70–76 (2022).

Magon, A. et al. The effect of health literacy on vaccine hesitancy among Italian anticoagulated population during COVID-19 pandemic: the moderating role of health engagement. Hum. Vaccin. Immunother. 17 , 5007–5012 (2021).

Martinelli, M. & Veltri, G. A. Do cognitive styles affect vaccine hesitancy? A dual-process cognitive framework for vaccine hesitancy and the role of risk perceptions. Soc. Sci. Med . 289 , 114403 (2021).

McCabe, S. D. et al. Unraveling Attributes of COVID-19 vaccine hesitancy and uptake in the U.S.: a large nationwide study. Preprint at medRxiv https://doi.org/10.1101/2021.04.05.21254918 (2021).

McCarthy, M., Murphy, K., Sargeant, E. & Williamson, H. Examining the relationship between conspiracy theories and covid-19 vaccine hesitancy: a mediating role for perceived health threats, trust, and anomie? Anal. Soc. Issues Public Policy 22 , 106–129 (2022).

McElfish, P. A. et al. Sociodemographic determinants of COVID-19 vaccine hesitancy, fear of infection, and protection self-efficacy. J. Prim. Care Community Health 12 , 21501327211040746 (2021).

Milošević Đorđević, J., Mari, S., Vdović, M. & Milošević, A. Links between conspiracy beliefs, vaccine knowledge, and trust: anti-vaccine behavior of Serbian adults. Soc. Sci. Med . 277 , 113930 (2021).

Mohamad, O. et al. Factors associated with the intention of Syrian adult population to accept COVID19 vaccination: a cross-sectional study. BMC Public Health 21 , 1310 (2021).

Mohan, S., Reagu, S., Lindow, S. & Alabdulla, M. COVID-19 vaccine hesitancy in perinatal women: a cross sectional survey. J. Perinat. Med. 49 , 678–685 (2021).

Mohanan, L., Klugar, M., Pokorna, A. & Riad, A. COVID-19 vaccine booster hesitancy (VBH) of healthcare workers in Czechia: national cross-sectional study. Vaccines 9 , 1437 (2021).

Momplaisir, F. M. et al. Racial/ethnic differences in COVID-19 vaccine hesitancy among health care workers in 2 large academic hospitals. JAMA Netw. Open 4 , e2121931 (2021).

Monami, M. et al. COVID-19 vaccine hesitancy and early adverse events reported in a cohort of 7,881 Italian physicians. Ann. Ig. 34 , 344–357 (2022).

Moore, D. et al. Low COVID-19 vaccine hesitancy in Brazil. Vaccine 39 , 6262–6268 (2021).

Morris, J. L., Baniak, L. M., Luyster, F. S. & Dunbar-Jacob, J. Covid-19 vaccine confidence and hesitancy in nursing students and faculty at a large academic medical center. Nurs. Outlook 70 , 347–354 (2022).

Muhajarine, N. et al. COVID-19 vaccine hesitancy and refusal and associated factors in an adult population in Saskatchewan, Canada: evidence from predictive modelling. PLoS ONE 16 , e0259513 (2021).

Myers, A., Ipsen, C. & Lissau, A. COVID-19 vaccination hesitancy among Americans with disabilities aged 18-65: An exploratory analysis. Disabil. Health J. 15 , 101223 (2022).

Nowak, S. A., Gidengil, C. A., Parker, A. M. & Matthews, L. J. Association among trust in health care providers, friends, and family, and vaccine hesitancy. Vaccine 39 , 5737–5740 (2021).

Okubo, R., Yoshioka, T., Ohfuji, S., Matsuo, T. & Tabuchi, T. COVID-19 vaccine hesitancy and its associated factors in Japan. Vaccines 9 , 662 (2021).

Oliveira, B. et al. Prevalence and factors associated with covid-19 vaccine hesitancy in Maranhão, Brazil. Rev. Saude Publica 55 , 12 (2021).

Omar, D. I. & Hani, B. M. Attitudes and intentions towards COVID-19 vaccines and associated factors among Egyptian adults. J. Infect. Public Health 14 , 1481–1488 (2021).

Orangi, S. et al. Assessing the level and determinants of COVID-19 vaccine confidence in Kenya. Vaccines 9 , 936 (2021).

Pal, S. et al. COVID-19 vaccine hesitancy and attitude toward booster doses among US healthcare workers. Vaccines 9 , 1358 (2021).

Park, H. K., Ham, J. H., Jang, D. H., Lee, J. Y. & Jang, W. M. Political ideologies, government trust, and COVID-19 vaccine hesitancy in South Korea: a cross-sectional survey. Int. J. Environ. Res. Public Health 18 , 10655 (2021).

Piltch-Loeb, R. et al. COVID-19 vaccine concerns about safety, effectiveness, and policies in the United States, Canada, Sweden, and Italy among unvaccinated individuals. Vaccines 9 , 1138 (2021).

Purnell, M. et al. Exploring COVID-19 vaccine hesitancy at a rural historically black college and university. J. Am. Pharm. Assoc. 62 , 340–344 (2021).

Purvis, R. S. et al. Trusted sources of COVID-19 vaccine information among hesitant adopters in the United States. Vaccines 9 , 1418 (2021).

Qunaibi, E. A., Helmy, M., Basheti, I. & Sultan, I. A high rate of COVID-19 vaccine hesitancy in a large-scale survey on Arabs. eLife 10 , e68038 (2021).

Riad, A. et al. Prevalence and drivers of COVID-19 vaccine hesitancy among Czech university students: national cross-sectional study. Vaccines 9 , 948 (2021).

Lavoie, K. et al. Understanding national trends in COVID-19 vaccine hesitancy in Canada - April 2020 to March 2021. BMJ Open 12 , e059411 (2022).

Robertson, E. et al. Predictors of COVID-19 vaccine hesitancy in the UK household longitudinal study. Brain Behav. Immun. 94 , 41–50 (2021).

Rodriguez, R. M. et al. The rapid evaluation of COVID-19 vaccination in emergency departments for underserved patients study. Ann. Emerg. Med. 78 , 502–510 (2021).

Saluja, S. et al. Disparities in COVID-19 vaccine hesitancy among Los Angeles County adults after vaccine authorization. Prev. Med. Rep. 24 , 101544 (2021).

Shallal, A. et al. Evaluation of COVID-19 vaccine attitudes among Arab American healthcare professionals living in the United States. Vaccines 9 , 942 (2021).

Sirikalyanpaiboon, M. et al. COVID-19 vaccine acceptance, hesitancy, and determinants among physicians in a university-based teaching hospital in Thailand. BMC Infect. Dis. 21 , 1174 (2021).

Solak, Ç., Peker-Dural, H., Karlıdağ, S. & Peker, M. Linking the behavioral immune system to COVID-19 vaccination intention: the mediating role of the need for cognitive closure and vaccine hesitancy. Pers. Individ. Dif. 185 , 111245 (2022).

Spinewine, A. et al. Attitudes towards COVID-19 vaccination among hospital staff-understanding what matters to hesitant people. Vaccines 9 , 469 (2021).

Stojanovic, J. et al. Global trends and correlates of COVID-19 vaccination hesitancy: findings from the iCARE study. Vaccines 9 , 661 (2021).

Strathdee, S. A. et al. Correlates of coronavirus disease 2019 (COVID-19) vaccine hesitancy among people who inject drugs in the San Diego-Tijuana border region. Clin. Infect. Dis. https://doi.org/10.1093/cid/ciab975 (2021).

Szilagyi, P. G. et al. The role of trust in the likelihood of receiving a COVID-19 vaccine: results from a national survey. Prev. Med. 153 , 106727 (2021).

Szilagyi, P. G. et al. Likelihood of COVID-19 vaccination by subgroups across the US: post-election trends and disparities. Hum. Vaccin. Immunother. 17 , 3262–3267 (2021).

Tahir, A. I. et al. COVID-19 vaccine acceptance, hesitancy and refusal among Iraqi Kurdish population. Int. J. Health Sci. 16 , 10–16 (2022).

Teasdale, C. A. et al. Parental plans to vaccinate children for COVID-19 in New York city. Vaccine 39 , 5082–5086 (2021).

Thanapluetiwong, S., Chansirikarnjana, S., Sriwannopas, O., Assavapokee, T. & Ittasakul, P. Factors associated with COVID-19 vaccine hesitancy in Thai seniors. Patient Prefer. Adherence 15 , 2389–2403 (2021).

Tsai, R. et al. COVID-19 vaccine hesitancy and acceptance among individuals with cancer, autoimmune diseases, or other serious comorbid conditions: cross-sectional, internet-based survey. JMIR Public Health Surveill. 8 , e29872 (2022).

Uzochukwu, I. C. et al. COVID-19 vaccine hesitancy among staff and students in a Nigerian tertiary educational institution. Ther. Adv. Infect. Dis. 8 , 20499361211054923 (2021).

Vieira Rezende, R. P. et al. Characteristics associated with COVID-19 vaccine hesitancy: a nationwide survey of 1000 patients with immune-mediated inflammatory diseases. Vaccine 39 , 6454–6459 (2021).

Wang, S. X., Bell-Rogers, N., Dillard, D. & Harrington, M. A. COVID-19 vaccine hesitancy in Delaware’s underserved communities. Dela. J. Public Health 7 , 168–175 (2021).

Wang, X., Lin, L., Xu, J., Wang, W. & Zhou, X. Expectant parents’ vaccine decisions influenced by the 2018 Chinese vaccine crisis: a cross-sectional study. Prev. Med. 145 , 106423 (2021).

Waring, M. E. et al. Factors associated with mothers’ hesitancy to receive a COVID-19 vaccine. J. Behav. Med . https://doi.org/10.1007/s10865-021-00268-0 (2022).

Waters, A. R. et al. COVID-19 vaccine hesitancy among adolescent and young adult cancer survivors. JNCI Cancer Spectr. 5 , pkab049 (2021).

West, H. et al. COVID-19 vaccine hesitancy among temporary foreign workers from Bangladesh. Health Syst. Reform 7 , e1991550 (2021).

Willis, D. E. et al. COVID-19 vaccine hesitancy: race/ethnicity, trust, and fear. Clin. Transl. Sci. 14 , 2200–2207 (2021).

Willis, D. E., Presley, J., Williams, M., Zaller, N. & McElfish, P. A. COVID-19 vaccine hesitancy among youth. Hum. Vaccin. Immunother. 17 , 5013–5015 (2021).

Wiysonge, C. S. et al. COVID-19 vaccine acceptance and hesitancy among healthcare workers in South Africa. Expert Rev. Vaccines 21 , 549–559 (2022).

Wu, H., Ward, M., Brown, A., Blackwell, E. & Umer, A. COVID-19 vaccine intent in Appalachian patients with multiple sclerosis. Mult. Scler. Relat. Disord. 57 , 103450 (2021).

Wu, J. et al. COVID-19 vaccine hesitancy among Chinese population: a large-scale national study. Front. Immunol. 12 , 781161 (2021).

Xu, Y. et al. A cross-sectional survey on covid-19 vaccine hesitancy among parents from Shandong vs. Zhejiang. Front. Public Health 9 , 779720 (2021).

Zhang, M. X., Lin, X. Q., Chen, Y., Tung, T. H. & Zhu, J. S. Determinants of parental hesitancy to vaccinate their children against COVID-19 in China. Expert Rev. Vaccines 20 , 1339–1349 (2021).

Zhang, P. et al. Who is more likely to hesitate to accept COVID-19 vaccine: a cross-sectional survey in China. Expert Rev. Vaccines 21 , 397–406 (2022).

Zhao, Y. M. et al. Public willingness and determinants of COVID-19 vaccination at the initial stage of mass vaccination in China. Vaccines 9 , 1172 (2021).

Opel, D. J. et al. Validity and reliability of a survey to identify vaccine-hesitant parents. Vaccine 29 , 6598–6605 (2011).

Abd Halim, H., Abdul-Razak, S., Md Yasin, M. & Isa, M. R. Validation study of the Parent Attitudes About Childhood Vaccines (PACV) questionnaire: the Malay version. Hum. Vaccin. Immunother. 16 , 1040–1049 (2020).

Al-Regaiey, K. A. et al. Influence of social media on parents’ attitudes towards vaccine administration. Hum. Vaccin. Immunother. 18 , 1872340 (2021).

Alsuwaidi, A. R. et al. Vaccine hesitancy and its determinants among Arab parents: a cross-sectional survey in the United Arab Emirates. Hum. Vaccin. Immunother. 16 , 3163–3169 (2020).

Bagateli, L. E. et al. COVID-19 vaccine hesitancy among parents of children and adolescents living in Brazil. Vaccines 9 , 1115 (2021).

Beck, A., Bianchi, A. & Showalter, D. Evidence-based practice model to increase human papillomavirus vaccine uptake: a stepwise approach. Nurs. Womens Health 25 , 430–436 (2021).

Bianco, A., Mascaro, V., Zucco, R. & Pavia, M. Parent perspectives on childhood vaccination: how to deal with vaccine hesitancy and refusal? Vaccine 37 , 984–990 (2019).

Bonsu, N. E. M. et al. Understanding vaccine hesitancy among parents of children with autism spectrum disorder and parents of children with non-autism developmental delays. J. Child Neurol. 36 , 911–918 (2021).

Bulun, M. A. & Acuner, D. Turkish adaptation and reliability and validity study of parent attitudes about childhood vaccines survey. J. Pediatr. Res. 7 , 323–330 (2020).

Chang, J. & Kochel, R. Vaccine hesitancy and attributions for autism among racially and ethnically diverse groups of parents of children with autism spectrum disorder: a pilot study. Autism Res. 13 , 1790–1796 (2020).

Chung-Delgado, K., Valdivia Venero, J. E. & Vu, T. M. Vaccine hesitancy: characteristics of the refusal of childhood vaccination in a Peruvian population. Cureus 13 , e14105 (2021).

Clayton, K., Finley, C., Flynn, D. J., Graves, M. & Nyhan, B. Evaluating the effects of vaccine messaging on immunization intentions and behavior: evidence from two randomized controlled trials in Vermont. Vaccine 39 , 5909–5917 (2021).

Cole, J., Berman, S., Gardner, J., McGuire, K. & Chen, A. M. H. Implementation of a motivational interviewing-based decision tool to improve childhood vaccination rates: pilot study protocol. Res. Social Adm. Pharm. 17 , 619–624 (2020).

Cunningham, R. M., Guffey, D., Minard, C. G., Opel, D. J. & Boom, J. A. The effect of screening for vaccine hesitancy on the subsequent development of hesitancy: a randomized controlled trial, Houston, TX. Hum. Vaccin. Immunother. 17 , 1994–2000 (2021).

Daley, M. F., Narwaney, K. J., Shoup, J. A., Wagner, N. M. & Glanz, J. M. Addressing parents’ vaccine concerns: a randomized trial of a social media intervention. Am. J. Prev. Med . 55 , 44–54 (2018).

Eby, A. Z. Impacting parental vaccine decision-making. Pediatr. Nurs. 43 , 22–29, 34 (2017).

Goin-Kochel, R. P. et al. Beliefs about causes of autism and vaccine hesitancy among parents of children with autism spectrum disorder. Vaccine 38 , 6327–6333 (2020).

Henrikson, N. B. et al. Longitudinal trends in vaccine hesitancy in a cohort of mothers surveyed in Washington State, 2013–2015. Public Health Rep. 132 , 451–454 (2017).

Henrikson, N. B. et al. Physician communication training and parental vaccine hesitancy: a randomized trial. Pediatrics 136 , 70–79 (2015).

Hofstetter, A. M. et al. Parental vaccine hesitancy and declination of influenza vaccination among hospitalized children. Hosp. Pediatr. 8 , 628–635 (2018).

Kalok, A. et al. Vaccine hesitancy towards childhood immunisation amongst urban pregnant mothers in Malaysia. Vaccine 38 , 2183–2189 (2020).

Langkamp, D. L., Dusseau, A. & Brown, M. F. Vaccine hesitancy and low immunization rates in children with Down Syndrome. J. Pediatr. 223 , 64–67 (2020).

Mical, R., Martin-Velez, J., Blackstone, T. & Derouin, A. Vaccine hesitancy in rural pediatric primary care. J. Pediatr. Health Care 35 , 16–22 (2020).

Mohd Azizi, F. S., Kew, Y. & Moy, F. M. Vaccine hesitancy among parents in a multi-ethnic country, Malaysia. Vaccine 35 , 2955–2961 (2017).

Napolitano, F., D’Alessandro, A. & Angelillo, I. F. Investigating Italian parents’ vaccine hesitancy: a cross-sectional survey. Hum. Vaccin. Immunother. 14 , 1558–1565 (2018).

Olarewaju, V. O. et al. Application of the Parent Attitudes about Childhood Vaccines (PACV) survey in three national languages in Switzerland: exploratory factor analysis and Mokken scale analysis. Hum. Vaccin. Immunother. 17 , 2652–2660 (2021).

Opel, D. J. et al. The architecture of provider-parent vaccine discussions at health supervision visits. Pediatrics 132 , 1037–1046 (2013).

Opel, D. J. et al. The relationship between parent attitudes about childhood vaccines survey scores and future child immunization status: a validation study. JAMA Pediatr. 167 , 1065–1071 (2013).

Opel, D. J. et al. Impact of childhood vaccine discussion format over time on immunization status. Acad. Pediatr. 18 , 430–436 (2018).

Orr, C. & Beck, A. F. Measuring vaccine hesitancy in a minority community. Clin. Pediatr. 56 , 784–788 (2017).

Reuben, R., Aitken, D., Freedman, J. L. & Einstein, G. Mistrust of the medical profession and higher disgust sensitivity predict parental vaccine hesitancy. PLoS ONE 15 , e0237755 (2020).

Sabahelzain, M. M., van den Borne, B., Moukhyer, M. & Bosma, H. Determinants of measles vaccine hesitancy among Sudanese parents in Khartoum State, Sudan: a cross-sectional study. Vaccines 10 , 6 (2022).

Sahni, L. C. et al. Vaccine hesitancy and illness perceptions: comparing parents of children with autism spectrum disorder to other parent groups. Child. Health Care 35 , 385–402 (2020).

Sankaranarayanan, S., Jayaraman, A. & Gopichandran, V. Assessment of vaccine hesitancy among parents of children between 1 and 5 years of age at a tertiary care hospital in Chennai. Indian J. Community Med. 44 , 394–396 (2019).

Strelitz, B. et al. Parental vaccine hesitancy and acceptance of seasonal influenza vaccine in the pediatric emergency department. Vaccine 33 , 1802–1807 (2015).

Wagner, A. L. et al. Vaccine hesitancy and concerns about vaccine safety and effectiveness in Shanghai, China. Am. J. Prev. Med . 60 , S77–S86 (2020).

Williams, J. T. B., Rice, J. D. & O’Leary, S. T. Associations between religion, religiosity, and parental vaccine hesitancy. Vaccine X 9 , 100121 (2021).

Williams, S. E. et al. Screening tool predicts future underimmunization among a pediatric practice in Tennessee. Clin. Pediatr. 55 , 537–542 (2016).

Williams, S. E. et al. A randomized trial to increase acceptance of childhood vaccines by vaccine-hesitant parents: a pilot study. Acad. Pediatr. 13 , 475–480 (2013).

Yufika, A. et al. Parents’ hesitancy towards vaccination in Indonesia: a cross-sectional study in Indonesia. Vaccine 38 , 2592–2599 (2020).

Akhmetzhanova, Z., Sazonov, V., Riethmacher, D. & Aljofan, M. Vaccine adherence: the rate of hesitancy toward childhood immunization in Kazakhstan. Expert Rev. Vaccines 19 , 579–584 (2020).

Cunningham, R. M. et al. Development of a Spanish version of the parent attitudes about childhood vaccines survey. Hum. Vaccin. Immunother. 15 , 1106–1110 (2019).

Cunningham, R. M. et al. Prevalence of vaccine hesitancy among expectant mothers in Houston, Texas. Acad. Pediatr. 18 , 154–160 (2018).

Della Polla, G., Pelullo, C. P., Napolitano, F. & Angelillo, I. F. HPV vaccine hesitancy among parents in Italy: a cross-sectional study. Hum. Vaccin. Immunother. 16 , 2744–2751 (2020).

Dubé, E. et al. Overview of knowledge, attitudes, beliefs, vaccine hesitancy and vaccine acceptance among mothers of infants in Quebec, Canada. Hum. Vaccin. Immunother. 15 , 113–120 (2019).

Marshall, S., Moore, A. C., Sahm, L. J. & Fleming, A. Parent attitudes about childhood vaccines: point prevalence survey of vaccine hesitancy in an Irish population. Pharmacy 9 , 188 (2021).

Moyer-Gusé, E., Robinson, M. J. & McKnight, J. The role of humor in messaging about the MMR vaccine. J. Health Commun. 23 , 514–522 (2018).

Olarewaju, V. O. et al. The Youth Attitudes about Vaccines (YAV-5) scale: adapting the parent attitudes about childhood vaccines short scale for use with youth in German, French, and Italian in Switzerland, exploratory factor analysis and mokken scaling analysis. Hum. Vaccin. Immunother. 17 , 5183–5190 (2021).

Roberts, J. R. et al. Vaccine hesitancy among parents of adolescents and its association with vaccine uptake. Vaccine 33 , 1748–1755 (2015).

Ruggiero, K. M. et al. Parents’ intentions to vaccinate their children against COVID-19. J. Pediatr. Health Care 35 , 509–517 (2021).

Voo, J. Y. H. et al. Vaccine knowledge, awareness and hesitancy: a cross sectional survey among parents residing at Sandakan District, Sabah. Vaccines 9 , 1348 (2021).

Wallace, A. S. et al. Development of a valid and reliable scale to assess parents’ beliefs and attitudes about childhood vaccines and their association with vaccination uptake and delay in Ghana. Vaccine 37 , 848–856 (2019).

Wang, Y. & Zhang, X. Influence of parental psychological flexibility on pediatric COVID-19 vaccine hesitancy: mediating role of self-efficacy and coping style. Front. Psychol. 12 , 783401 (2021).

Whelan, S. O., Moriarty, F., Lawlor, L., Gorman, K. M. & Beamish, J. Vaccine hesitancy and reported non-vaccination in an Irish pediatric outpatient population. Eur. J. Pediatr. 180 , 2839–2847 (2021).

Williams, J. T. B. & O’Leary, S. T. Denver religious leaders’ vaccine attitudes, practices, and congregational experiences. J. Relig. Health 58 , 1356–1367 (2019).

Yörük, S. & Güler, D. Factors associated with pediatric vaccine hesitancy of parents: a cross-sectional study in Turkey. Hum. Vaccin. Immunother. 17 , 4505–4511 (2021).

Zhang, H. et al. Vaccine hesitancy among parents and its influencing factors: a cross-sectional study in Guangzhou, China. Hum. Vaccin. Immunother. 17 , 5153–5161 (2021).

Cataldi, J. R., Dempsey, A. F., Allison, M. A. & O’Leary, S. T. Impact of publicly available vaccination rates on parental school and child care choice. Vaccine 36 , 4525–4531 (2018).

Cataldi, J. R., Dempsey, A. F. & O’Leary, S. T. Measles, the media, and MMR: impact of the 2014–15 measles outbreak. Vaccine 34 , 6375–6380 (2016).

Cataldi, J. R. et al. Addressing personal parental values in decisions about childhood vaccination: measure development. Vaccine 37 , 5688–5697 (2019).

McDonald, C. et al. a consent support resource with benefits and harms of vaccination does not increase hesitancy in parents–an acceptability study. Vaccines 8 , 500 (2020).

Opel, D. J. et al. Previsit screening for parental vaccine hesitancy: a cluster randomized trial. Pediatrics 144 , e20190802 (2019).

Williams, J. T. B. et al. Adapting and piloting a vaccine hesitancy questionnaire in rural Guatemala. Vaccine 39 , 180–184 (2021).

Akbas Gunes, N. Parents’ perspectives about vaccine hesitancies and vaccine rejection, in the west of Turkey. J. Pediatr. Nurs. 53 , e186–e194 (2020).

Aldakhil, H., Albedah, N., Alturaiki, N., Alajlan, R. & Abusalih, H. Vaccine hesitancy towards childhood immunizations as a predictor of mothers’ intention to vaccinate their children against COVID-19 in Saudi Arabia. J. Infect. Public Health 14 , 1497–1504 (2021).

AlGoraini, Y. M. et al. Confidence toward vaccination as reported by parents of children admitted to a tertiary care hospital in Riyadh, Saudi Arabia: a cross sectional study. Vacunas 21 , 95–104 (2020).

Alsubaie, S. S. et al. Vaccine hesitancy among Saudi parents and its determinants. Result from the WHO SAGE working group on vaccine hesitancy survey tool. Saudi Med. J. 40 , 1242–1250 (2019).

Connors, J. T., Hodges, E. A., DʼAuria, J. & Windham, L. Implementing vaccine hesitancy screening for targeted education. J. Am. Assoc. Nurse Pract. 30 , 450–459 (2018).

Dasgupta, P., Bhattacherjee, S., Mukherjee, A. & Dasgupta, S. Vaccine hesitancy for childhood vaccinations in slum areas of Siliguri, India. Indian J. Public Health 62 , 253–258 (2018).

Johnson, K. D. et al. Combatting a “twin-demic”: a quantitative assessment of COVID-19 and influenza vaccine hesitancy in primary care patients. Health Promot. Perspect. 11 , 179–185 (2021).

Krishnamoorthy, Y. et al. Factors related to vaccine hesitancy during the implementation of Measles-Rubella campaign 2017 in rural Puducherry–a mixed-method study. J. Fam. Med. Prim. Care 8 , 3962–3970 (2019).

Masters, N. B., Tefera, Y. A., Wagner, A. L. & Boulton, M. L. Vaccine hesitancy among caregivers and association with childhood vaccination timeliness in Addis Ababa, Ethiopia. Hum. Vaccin. Immunother. 14 , 2340–2347 (2018).

Miko, D., Costache, C., Colosi, H. A., Neculicioiu, V. & Colosi, I. A. Qualitative assessment of vaccine hesitancy in Romania. Medicina 55 , 282 (2019).

Sharma, S., Akhtar, F., Singh, R. K. & Mehra, S. Understanding the three As (awareness, access, and acceptability) dimensions of vaccine hesitancy in Odisha, India. Clin. Epidemiol. Glob. Health 8 , 399–403 (2020).

Soysal, G., Durukan, E. & Akdur, R. The evaluation of vaccine hesitancy and refusal for childhood vaccines and the covid-19 vaccine in individuals aged between 18 and 25 years. Turkish J. Immunol. 9 , 120–127 (2021).

Khezong, A. et al. Association between adult vaccine hesitancy and parental acceptance of childhood COVID-19 vaccines: a web-based survey in a northwestern region in China. Vaccines 9 , 1088 (2021).

Akel, K. B., Masters, N. B., Shih, S. F., Lu, Y. & Wagner, A. L. Modification of a vaccine hesitancy scale for use in adult vaccinations in the United States and China. Hum. Vaccin. Immunother. 17 , 2639–2646 (2021).

Chen, M. et al. An online survey of the attitude and willingness of Chinese adults to receive COVID-19 vaccination. Hum. Vaccin. Immunother. 17 , 2279–2288 (2021).

Dolu, İ., Turhan, Z. & Yalnız Dilcen, H. COVID-19 vaccine acceptance is associated with vaccine hesitancy, perceived risk and previous vaccination experiences. Disaster Med. Public Health Prep . https://doi.org/10.1017/dmp.2021.370 (2021).

Domek, G. J. et al. Measuring vaccine hesitancy: field testing the WHO SAGE Working Group on Vaccine Hesitancy survey tool in Guatemala. Vaccine 36 , 5273–5281 (2018).

Gatwood, J. et al. Extent of and reasons for vaccine hesitancy in adults at high-risk for pneumococcal disease. Am. J. Health Promot . 35 , 908–916 (2021).

Han, Y. et al. Parental category B vaccine hesitancy and associated factors in China: an online cross-sectional survey. Expert Rev. Vaccines 21 , 145–153 (2021).

He, K., Mack, W. J., Neely, M., Lewis, L. & Anand, V. Parental perspectives on immunizations: impact of the COVID-19 pandemic on childhood vaccine hesitancy. J. Community Health 47 , 39–52 (2022).

Helmkamp, L. J. et al. A validated modification of the vaccine hesitancy scale for childhood, influenza and HPV vaccines. Vaccine 39 , 1831–1839 (2021).

Jones, D. L. et al. Severe acute respiratory syndrome coronavirus 2: vaccine hesitancy among underrepresented racial and ethnic groups with HIV in Miami, Florida. Open Forum Infect. Dis. 8 , ofab154 (2021).

Kasrine Al Halabi, C. et al. Attitudes of Lebanese adults regarding COVID-19 vaccination. BMC Public Health 21 , 998 (2021).

Kempe, A. et al. Parental hesitancy about routine childhood and influenza vaccinations: a national survey. Pediatrics 146 , e20193852 (2020).

Kim, H. W. et al. Perceptions of nurses on human papillomavirus vaccinations in the Republic of Korea. PLoS ONE 14 , e0211475 (2019).

Liu, P. L., Zhao, X. & Wan, B. COVID-19 information exposure and vaccine hesitancy: the influence of trust in government and vaccine confidence. Psychol. Health Med. https://doi.org/10.1080/13548506.2021.2014910 (2021).

Lu, F. & Sun, Y. COVID-19 vaccine hesitancy: the effects of combining direct and indirect online opinion cues on psychological reactance to health campaigns. Comput. Hum. Behav. 127 , 107057 (2022).

Lu, J. et al. Sensitivity to COVID-19 vaccine effectiveness and safety in Shanghai, China. Vaccines 9 , 472 (2021).

Lu, X. et al. Low willingness to vaccinate against herpes zoster in a Chinese metropolis. Hum. Vaccin. Immunother. 17 , 4163–4170 (2021).

Lu, X. et al. Gap between willingness and behavior in the vaccination against influenza, pneumonia, and herpes zoster among Chinese aged 50–69 years. Expert Rev. Vaccines 20 , 1147–1152 (2021).

Luyten, J., Bruyneel, L. & van Hoek, A. J. Assessing vaccine hesitancy in the UK population using a generalized vaccine hesitancy survey instrument. Vaccine 37 , 2494–2501 (2019).

Önal, Ö., Eroğlu, H. N., Evcil, F. Y., Kişioğlu, A. N. & Uskun, E. Validity and reliability of Turkish version of the Vaccine Hesitancy Scale. Turk. Arch. Pediatr. 56 , 230–235 (2021).

Ren, J. et al. The demographics of vaccine hesitancy in Shanghai, China. PLoS ONE 13 , e0209117 (2018).

Sabahelzain, M. M. et al. Psychometric properties of the adapted measles vaccine hesitancy scale in Sudan. PLoS ONE 15 , e0237171 (2020).

Sadaqat, W. et al. Determination of COVID-19 vaccine hesitancy among university students. Cureus 13 , e17283 (2021).

Shapiro, G. K. et al. The vaccine hesitancy scale: psychometric properties and validation. Vaccine 36 , 660–667 (2018).

Shen, X. et al. Assessing the COVID-19 vaccine hesitancy in the Chinese adults using a generalized vaccine hesitancy survey instrument. Hum. Vaccin. Immunother. 17 , 4005–4012 (2021).

Shih, S. F. et al. Vaccine hesitancy and rejection of a vaccine for the novel coronavirus in the United States. Front. Immunol. 12 , 558270 (2021).

Szilagyi, P. G. et al. Prevalence and characteristics of HPV vaccine hesitancy among parents of adolescents across the US. Vaccine 38 , 6027–6037 (2020).

Temsah, M. H. et al. Parental attitudes and hesitancy about COVID-19 vs. routine childhood vaccinations: a national survey. Front. Public Health 9 , 752323 (2021).

Thaker, J. The persistence of vaccine hesitancy: COVID-19 vaccination intention in New Zealand. J. Health Commun. 26 , 104–111 (2021).

Turhan, Z., Dilcen, H. Y. & Dolu, İ. The mediating role of health literacy on the relationship between health care system distrust and vaccine hesitancy during COVID-19 pandemic. Curr. Psychol . https://doi.org/10.1007/s12144-021-02105-8 (2021).

Vallis, M. & Glazer, S. Protecting individuals living with overweight and obesity: attitudes and concerns toward COVID-19 vaccination in Canada. Obesity 29 , 1128–1137 (2021).

Wagner, A. L. et al. Comparisons of vaccine hesitancy across five low- and middle-income countries. Vaccines 7 , 155 (2019).

Wagner, A. L., Shotwell, A. R., Boulton, M. L., Carlson, B. F. & Mathew, J. L. Demographics of vaccine hesitancy in Chandigarh, India. Front. Med. 7 , 585579 (2020).

Wang, Q. et al. Delays in routine childhood vaccinations and their relationship with parental vaccine hesitancy: a cross-sectional study in Wuxi, China. Expert Rev. Vaccines 21 , 135–143 (2021).

Wang, Q. et al. Vaccine hHesitancy: COVID-19 and influenza vaccine willingness among parents in Wuxi, China–a cross-sectional study. Vaccines 9 , 342 (2021).

Xu, Y. et al. COVID-19 vaccination attitudes with neuromyelitis optica spectrum disorders: vaccine hesitancy and coping style. Front. Neurol. 12 , 717111 (2021).

Yeşiltepe, A., Aslan, S. & Bulbuloglu, S. Investigation of perceived fear of COVID-19 and vaccine hesitancy in nursing students. Hum. Vaccin. Immunother. 17 , 5030–5037 (2021).

Duong, T. V. et al. Oxford COVID-19 vaccine hesitancy in school principals: impacts of gender, well-being, and coronavirus-related health literacy. Vaccines 9 , 958 (2021).

Guaracha-Basáñez, G. et al. COVID-19 vaccine hesitancy among Mexican outpatients with rheumatic diseases. Hum. Vaccin. Immunother. 17 , 5038–5047 (2021).

Joshi, A. et al. COVID-19 vaccine hesitancy in healthcare workers amidst the second wave of the pandemic in India: a single centre study. Cureus 13 , e17370 (2021).

Zhang, X., Guo, Y., Zhou, Q., Tan, Z. & Cao, J. The mediating roles of medical mistrust, knowledge, confidence and complacency of vaccines in the pathways from conspiracy beliefs to vaccine hesitancy. Vaccines 9 , 1342 (2021).

Pomares, T. D. et al. Association of cognitive biases with human papillomavirus vaccine hesitancy: a cross-sectional study. Hum. Vaccin. Immunother. 16 , 1018–1023 (2020).

Ashkenazi, S. et al. The relationship between parental source of information and knowledge about measles / measles vaccine and vaccine hesitancy. Vaccine 38 , 7292–7298 (2020).

Biswas, N., Mustapha, T., Khubchandani, J. & Price, J. H. The nature and extent of COVID-19 vaccination hesitancy in healthcare workers. J. Community Health 46 , 1244–1251 (2021).

Boattini, M. et al. Rubella serosurvey and factors related to vaccine hesitancy in childbearing women in Italy. Prev. Med. Rep. 15 , 100945 (2019).

Browne, S. K. et al. Coronavirus disease 2019 (COVID-19) vaccine hesitancy among physicians, physician assistants, nurse practitioners, and nurses in two academic hospitals in Philadelphia. Infect. Control Hosp. Epidemiol. https://doi.org/10.1017/ice.2021.410 (2021).

Dybsand, L. L., Hall, K. J. & Carson, P. J. Immunization attitudes, opinions, and knowledge of healthcare professional students at two Midwestern universities in the United States. BMC Med. Educ. 19 , 242 (2019).

Gao, X., Li, H., He, W. & Zeng, W. COVID-19 vaccine hesitancy among medical students: the next COVID-19 challenge in Wuhan, China. Disaster Med. Public Health Prep . https://doi.org/10.1017/dmp.2021.291 (2021).

Gerber, J. E. et al. Vaccinomics: a cross-sectional survey of public values. Hum. Vaccin. Immunother. 17 , 2999–3015 (2021).

Giambi, C. et al. Parental vaccine hesitancy in Italy – results from a national survey. Vaccine 36 , 779–787 (2018).

Guay, M., Gosselin, V., Petit, G., Baron, G. & Gagneur, A. Determinants of vaccine hesitancy in Quebec: a large population-based survey. Hum. Vaccin. Immunother. 15 , 2527–2533 (2019).

Harapan, H. et al. Vaccine hesitancy among communities in ten countries in Asia, Africa, and South America during the COVID-19 pandemic. Pathog. Glob. Health 116 , 236–243 (2022).

Hornsey, M. J., Lobera, J. & Díaz-Catalán, C. Vaccine hesitancy is strongly associated with distrust of conventional medicine, and only weakly associated with trust in alternative medicine. Soc. Sci. Med. 255 , 113019 (2020).

Hu, Y., Chen, Y., Liang, H. & Wang, Y. Reliability and validity of a survey to identify vaccine hesitancy among parents in Changxing county, Zhejiang province. Hum. Vaccin. Immunother. 15 , 1092–1099 (2019).

Johnson, D. K. et al. Combating vaccine hesitancy with vaccine-preventable disease familiarization: an interview and curriculum intervention for college students. Vaccines 7 , 39 (2019).

Kassianos, G. et al. Motors of influenza vaccination uptake and vaccination advocacy in healthcare workers: a comparative study in six European countries. Vaccine 36 , 6546–6552 (2018).

Klassen, A. C. et al. Formative research to address vaccine hesitancy in Tajikistan. Vaccine 39 , 1516–1527 (2021).

Liu, Y. et al. COVID-19 vaccination in people living with HIV (PLWH) in China: a cross sectional study of vaccine hesitancy, safety, and immunogenicity. Vaccines 9 , 1458 (2021).

Paoli, S. et al. Assessing vaccine hesitancy among healthcare workers: a cross-sectional study at an Italian paediatric hospital and the development of a healthcare worker’s vaccination compliance index. Vaccines 7 , 201 (2019).

Rahman, M., Hossain, A., Sufian, A. & Anwar, N. COVID-19 vaccine demand, hesitancy, and nationalism: a case of protection motivation behavior in Bangladesh. J. Infect. Dev. Ctries 15 , 1388–1395 (2021).

Raude, J. et al. Opening the ‘Vaccine Hesitancy’ black box: how trust in institutions affects French GPs’ vaccination practices. Expert Rev. Vaccines 15 , 937–948 (2016).

Schwarzinger, M., Watson, V., Arwidson, P., Alla, F. & Luchini, S. COVID-19 vaccine hesitancy in a representative working-age population in France: a survey experiment based on vaccine characteristics. Lancet Public Health 6 , e210–e221 (2021).

Stoeckel, F., Carter, C., Lyons, B. A. & Reifler, J. Association of vaccine hesitancy and immunization coverage rates in the European Union. Vaccine 39 , 3935–3939 (2021).

Tomljenovic, M., Petrovic, G., Antoljak, N. & Hansen, L. Vaccination attitudes, beliefs and behaviours among primary health care workers in northern Croatia. Vaccine 39 , 738–745 (2021).

Verger, P. et al. Vaccine hesitancy among hospital staff physicians: a cross-sectional survey in France in 2019. Vaccine 39 , 4481–4488 (2021).

Verger, P. et al. Vaccine hesitancy among general practitioners and its determinants during controversies: a national cross-sectional survey in France. EBioMedicine 2 , 891–897 (2015).

Carranza, D., Dub, T. & Sivelä, J. Vaccine Hesitancy and Uptake. From Research and Practices to Implementation (Finnish Institute for Health and Welfare (THL), 2021); https://eu-jav.com/public_deliverable/deliverable-8-1-vaccine-hesitancy-and-uptake-from-research-and-practices-to-implementation/

Merriam-Webster Dictionary Hesitancy https://www.merriam-webster.com/dictionary/hesitancy (2021).

Merriam-Webster Dictionary Vaccine Hesitancy https://www.merriam-webster.com/dictionary/vaccine%20hesitancy (2021).

Siani, A. et al. Investigating the determinants of vaccine hesitancy within undergraduate students’ social sphere. Z. Gesundh. Wiss . https://doi.org/10.1007/s10389-021-01538-6 (2021).

Strong, H., Reiter-Purtill, J., Howarth, T., West-Smith, L. & Zeller, M. H. Early COVID-19 vaccine hesitancy characteristics in mothers following bariatric surgery. Obes. Surg. 32 , 852–860 (2022).

Lavail, K. H. & Kennedy, A. M. The role of attitudes about vaccine safety, efficacy, and value in explaining parents’ reported vaccination behavior. Health Educ. Behav. 40 , 544–551 (2013).

Frew, P. M. et al. Development of a measure to assess vaccine confidence among men who have sex with men. Expert Rev. Vaccines 17 , 1053–1061 (2018).

World Bank Country and Lending Groups (The World Bank, 2021); https://datahelpdesk.worldbank.org/knowledgebase/articles/906519-world-bank-country-and-lending-groups

Hong, Q. N. et al. Mixed Methods Appraisal Tool (MMAT), version 2018 (Canadian Intellectual Property Office, Industry Canada, 2018).

Hong, Q. N. et al. Reporting the Results of the MMAT (Version 2018) http://mixedmethodsappraisaltoolpublic.pbworks.com/w/file/fetch/140056890/Reporting%20the%20results%20of%20the%20MMAT.pdf (2020).

Download references

Acknowledgements

J.L.A.H. and M.E.J.L.H. received funding from The Netherlands Organisation for Health Research and Development (ZonMw project number 839190002). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We thank J. van Haren for her valuable contribution in sorting and organizing the data of this systematic review.

Author information

Authors and affiliations.

Radboud University Medical Center, Radboud Institute for Health Sciences, Primary and Community Care, Nijmegen, the Netherlands

Daphne Bussink-Voorend, Jeannine L. A. Hautvast & Olga Visser

Behavioural Science Institute, Radboud University, Nijmegen, the Netherlands

Lisa Vandeberg

Radboud University Medical Center, Radboud Institute for Health Sciences, IQ Healthcare, Nijmegen, the Netherlands

Marlies E. J. L. Hulscher

You can also search for this author in PubMed   Google Scholar

Contributions

D.B.-V., J.L.A.H., O.V. and M.E.J.L.H. designed the project and analysed the data. D.B.-V., J.L.A.H., L.V., O.V. and M.E.J.L.H. interpreted the data. The manuscript, figures and tables were drafted by D.B.-V. and edited by J.L.A.H., L.V., O.V. and M.E.J.L.H.

Corresponding author

Correspondence to Daphne Bussink-Voorend .

Ethics declarations

Competing interests.

The authors declare no competing interests.

Peer review

Peer review information.

Nature Human Behaviour thanks Chuanxi Fu, Amalie Dyda and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary information.

Supplementary methods, PRISMA 2020 for abstracts checklist, PRISMA 2020 checklist.

Reporting Summary

Peer review file, supplementary table 1.

Overview and characteristics of included studies.

Supplementary Table 2

Overview of extracted excerpts.

Supplementary Table 3

Overview of vaccine hesitancy subpopulations.

Supplementary Table 4

Overview of studies describing a measurement of vaccine hesitancy.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Cite this article.

Bussink-Voorend, D., Hautvast, J.L.A., Vandeberg, L. et al. A systematic literature review to clarify the concept of vaccine hesitancy. Nat Hum Behav 6 , 1634–1648 (2022). https://doi.org/10.1038/s41562-022-01431-6

Download citation

Received : 25 November 2021

Accepted : 13 July 2022

Published : 22 August 2022

Issue Date : December 2022

DOI : https://doi.org/10.1038/s41562-022-01431-6

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Factors associated with sars-cov-2 vaccine hesitancy after stroke: a cross-sectional study.

  • Ying-Hai Zhu
  • Xiu-Qing Yao

BMC Public Health (2024)

  • Cameron O’Neill Byerley

Scientific Reports (2024)

Vaccine hesitancy and trust in sub-Saharan Africa

  • Kerstin Unfried

Association of personality traits and socio-environmental factors with COVID-19 pandemic-related conspiratorial thinking in the D-A-CH region

  • Jakob Weitzer
  • Gerald Steiner

SN Social Sciences (2024)

Modelling the impact of opinion flexibility on the vaccination choices during epidemics

  • Rossella Della Marca
  • Marco Menale

Ricerche di Matematica (2024)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

literature review on immunization

  • Research article
  • Open access
  • Published: 28 July 2021

Factors influencing childhood immunisation uptake in Africa: a systematic review

  • Abubakar Nasiru Galadima 1 ,
  • Nor Afiah Mohd Zulkefli 1 ,
  • Salmiah Md Said 1 &
  • Norliza Ahmad 1  

BMC Public Health volume  21 , Article number:  1475 ( 2021 ) Cite this article

31k Accesses

37 Citations

Metrics details

Vaccine preventable diseases are still the most common cause of childhood mortality, with an estimated 3 million deaths every year, mainly in Africa and Asia. An estimate of 29% deaths among children aged 1–59 months were due to vaccine preventable diseases. Despite the benefits of childhood immunisation, routine vaccination coverage for all recommended Expanded Programme on Immunization vaccines has remained poor in some African countries, such as Nigeria (31%), Ethiopia (43%), Uganda (55%) and Ghana (57%). The aim of this study is to collate evidence on the factors that influence childhood immunisation uptake in Africa, as well as to provide evidence for future researchers in developing, implementing and evaluating intervention among African populations which will improve childhood immunisation uptake.

We conducted a systematic review of articles on the factors influencing under-five childhood immunisation uptake in Africa. This was achieved by using various keywords and searching multiple databases (Medline, PubMed, CINAHL and Psychology & Behavioral Sciences Collection) dating back from inception to 2020.

Out of 18,708 recorded citations retrieved, 10,396 titles were filtered and 324 titles remained. These 324 abstracts were screened leading to 51 included studies. Statistically significant factors found to influence childhood immunisation uptake were classified into modifiable and non-modifiable factors and were further categorised into different groups based on relevance. The modifiable factors include obstetric factors, maternal knowledge, maternal attitude, self-efficacy and maternal outcome expectation, whereas non-modifiable factors were sociodemographic factors of parent and child, logistic and administration factors.

Different factors were found to influence under-five childhood immunisation uptake among parents in Africa. Immunisation health education intervention among pregnant women, focusing on the significant findings from this systematic review, would hopefully improve childhood immunisation uptake in African countries with poor coverage rates.

Peer Review reports

Vaccine Preventable Diseases (VPDs) are still the most common cause of childhood mortality with an estimated 3 million deaths every year, mainly in Africa and Asia [ 1 ]. A study conducted by the World Health Organization (WHO) and United Nations International Children’s Emergency Fund (UNICEF) in 2014, reported that an estimate of 29% deaths among children aged 1–59 months were due to vaccine preventable diseases [ 2 ]. In 2014, there were 24.1 million reported cases of pertussis, with the African region accounting for the highest proportion of 7.8 million (33%) cases [ 3 ].

Immunisation is considered to be one of the most successful and cost-effective public health sustainable interventions for human beings against diseases that affect our health [ 4 ]. Routine immunisation plays a key role to significantly reduce child mortality due to vaccine preventable diseases. WHO revealed that immunisation has been estimated to prevent 3 million deaths globally every year [ 5 ]. Between the years 2000 and 2016, a decrease of 84% in the measles mortality rate was recorded worldwide due to measles vaccination [ 6 ]. Likewise, a reduction in pertussis mortality was also recorded globally from 390,000 deaths in 1999 among children younger than 5 years of age to 160,700 deaths in 2014 as a result of vaccine effectiveness against pertussis [ 6 , 7 ].

According to the Expanded Programme on Immunization (EPI), every child in Africa must receive one dose of Bacillus Calmette Guerin (BCG), Oral Polio Vaccine (OPV0) and Hepatitis B Vaccine (HBV1) at birth, Penta1 & OPV1 at 6 weeks of age, Penta2 & OPV2 at 10 weeks of age, Penta3 & OPV3 at 14 weeks of age and measles and yellow fever at 9 months of age. Despite the benefits of childhood immunisation, routine vaccination coverage for all recommended EPI vaccines has remained poor in some African countries such as Nigeria (31%; 2018), Ethiopia (43%; 2019), Uganda (55%; 2016) and Ghana (57%; 2014). The coverage is higher in some of the African countries, such as in Tanzania in the year 2016 and Kenya in 2014 (75 and 78%, respectively) [ 8 , 9 , 10 , 11 , 12 , 13 ]. Diphtheria Pertussis and Tetanus (DPT3) coverage is also low in African countries such as Nigeria (50%) [ 8 ]. However, these coverages are still below the targets endorsed by WHO in the 2012 Global Vaccine Action Plan, which aimed to ensure delivery of universal access to immunisation with associated targets reaching 90% of the national vaccination coverage and at least 80% vaccination coverage in every district [ 14 ].

Previous observational studies conducted among African countries and other parts of the world highlighted various factors that influenced childhood immunisation uptake. These factors are socio-demographic factors including maternal age, maternal educational status, paternal educational status, mother’s marital status, maternal occupation, family income, wealth index and ethnicity [ 15 , 16 , 17 , 18 ] and obstetric factors including antenatal care follow-up, postnatal care follow-up, preceding birth interval and place of delivery [ 18 , 19 , 20 , 21 ]. Despite the poor childhood immunisation uptake in African countries, no current systematic review has been conducted that focuses on describing, in detail, the factors influencing childhood immunisation uptake in Africa. Therefore, the aim of this study is to collate evidence on the factors that influence childhood immunisation uptake in Africa as well as to provide evidence for future researchers in developing, implementing and evaluating intervention among African populations, which will improve childhood immunisation uptake. This study will assist in developing health promotional programs and policies on childhood immunisation in Africa.

This systematic review was conducted and reported in accordance with published Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) [ 22 ].

Electronic database search

We designed and implemented a comprehensive systematic literature search with the assistance of an experienced librarian using a well-developed strategy. The following databases were searched on the same date, from date of inception to 26th of October 2020: Medline, PubMed, CINAHL and Psychology & Behavioral Science Collection. The search strategy comprised a combination of medical subheading (MeSH) terms and keywords: childhood immunization uptake, factors, influencing or affecting, child or newborn or infant or baby, immunisation or vaccines or vaccination or pentavalent vaccine or Penta vaccine or Bacillus Calmette Guerin vaccine or BCG or Diphtheria Tetanus and Pertussis or DTP or Oral Polio vaccine or OPV or Measles vaccine or Yellow fever vaccine or Pneumococcal Conjugate vaccine or PCV or Hepatitis B vaccine or Hep B vaccine, uptake or adherence or compliance or coverage (Supplemental file 1 ).

The search strategy was developed in Medline and adapted for the other databases in order to account for differences in indexing. We restricted it to humans in the search process. Reference lists of included studies were also searched.

We included any observational (cross-sectional, case-control and cohort), mixed method study (convergent, exploratory and explanatory type) and qualitative study design (phenomenological study and case study) conducted in Africa published in the English language with findings relating to childhood immunisation uptake. The participants of these studies were caregivers with children under 5 years of age. The included studies also reported data on association between possible predictors and childhood immunisation or provided details of any non-compliance (vaccine refusal). We also included peer-reviewed full text publications reporting an association between at least one individual factor and uptake of childhood immunisation. Moreover, no restrictions were imposed on the year of publication.

We excluded articles without any description of study design, intervention studies (randomised controlled trial and quasi experimental design), review articles or systematic reviews and studies which made no mention of any of the Expanded Programme on Immunization target vaccines according to the National Program on Immunization (Table  1 ).

Description of study outcomes

Parental socio-demographic factors: maternal age, maternal education, paternal education, income, place of residence, maternal occupation and religion; child socio-demographic factors: child age and gender; religion and cultural beliefs; obstetric history: place of delivery, antenatal care follow-up and postnatal care follow-up; health care system: distance to hospital and availability of vaccine; knowledge; outcome expectation and self-efficacy (Table  2 ).

Selection of studies

All searched articles were evaluated for eligibility in order to be included in the review. The main researcher first removed all duplicates and screened the titles using Microsoft Excel. Abstract screening was then conducted independently by two reviewers. The full text of any article considered potentially relevant was also retrieved independently by the two reviewers. Consensus was reached by discussion or involvement of a third reviewer when there were differences of opinion.

Data extraction

A data extraction form was designed and piloted for this review. The form was used to extract the following data: study characteristics such as authors’ name and year of publication, study location, method, study design, sample size, results and factors. Data was extracted independently by the two reviewers to ensure accuracy. Any difference of opinion was resolved by discussion or involvement of a third reviewer.

Categorisation of factors associated with immunisation coverage

The qualitative synthesis of the factors influencing childhood immunisation uptake among parents of children under-five years of age are categorised into two groups, namely modifiable and non-modifiable factors. Modifiable factors are considered to be factors that are intrinsic to mothers/caregivers (can be changed through health education intervention targeting mothers/caregivers) while non-modifiable factors are considered to be factors that are extrinsic to mothers/caregivers (cannot be changed through health education intervention targeting mothers/caregivers). This classification would enable researchers to plan an appropriate health education intervention, with the use of appropriate health behavioural theory.

Assessment of methodological quality

ANG and NZ assessed the methodological quality for each of the included studies using the criteria outlined in the Joanna Briggs Institute (JBI) for both quantitative and qualitative studies [ 30 ], which includes 8 items for cross-sectional study design and 10 items for both case-control and qualitative study designs respectively . Any disagreement was resolved by discussion. In this review, we grouped articles into three categories: high, moderate and low quality for their methodological qualities using the JBI critical appraisal tools; studies scoring >  70% were considered as high quality, 60–69% were moderate quality and < 60% were low quality. This review may be prone to publication bias as grey literature/unpublished literature and studies that are not published in the English language were not considered.

Data synthesis

No meta-analysis was conducted, however, the results obtained from the systematic review were synthesised and placed in a logic framework. The various results obtained from the systematic review were shown in the logic framework and discussed in a narrative synthesis manner. The descriptive information and data for each of the included studies were captured using a table formulated by the researchers. These included study author, year and location; method; study design; sample size; results and factors. ANG synthesis ed the data and created a table with input from both NZ and SS. ANG and NZ reported factors under two different categories namely: modifiable and non-modifiable factors.

In this review, a wide range of AOR and 95% confidence intervals (upper limit and lower limit) from different studies were reported and confidence interval that does not include 1 and p -value less than 0.05 with AOR greater than 1 are considered as statistically significant.

Of 18,708 recorded citations retrieved, 8279 duplicates and 33 articles published in languages other than the English language were removed. After screening 10,396 titles, 324 records remained and their abstracts were screened. Seventy-nine records remained after abstracts were screened and their full texts were assessed for eligibility. We excluded 44 studies in total, of which 16 studies were not conducted in Africa, 12 studies did not make mention of the EPI vaccine, 9 were review studies and 7 were intervention studies. In total, 51 studies were included, of which 35 were from the main database search, 8 from forward citation tracking and 8 from reference tracking. All the included studies looked into factors influencing childhood immunisation uptake in one of the African countries (Fig.  1 ). Table  3 contained a summary of the factors that influence under-five childhood immunisation uptake among mothers/caregivers. The studies used different study designs to investigate the factors that influence under-five childhood immunisation uptake among parents.

figure 1

PRISMA flow diagram for the identification, screening, eligibility, and inclusion of studies

The majority of our epidemiological studies achieved at least 70% scores which is considered high [ 16 , 18 , 20 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 40 , 42 , 43 , 44 , 45 , 48 , 50 , 51 , 52 , 55 , 57 , 58 , 59 , 61 , 62 , 63 , 64 , 65 , 66 , 67 , 68 , 69 ] and the overall risk of bias was moderate in 10 studies (Table 3 ) [ 15 , 19 , 39 , 46 , 47 , 49 , 53 , 54 , 56 , 60 ]. Out of the 10 studies, 6 had a moderate risk of bias in the confounding domain and 4 had a moderate risk of bias in the outcome assessment domain. However, none of the included epidemiological studies demonstrated either a serious or low risk of bias < 60%.

Study locations

The studies were conducted in various countries within Africa, namely Angola, Cameroon, Congo and Mozambique with one study each [ 25 , 32 , 33 , 44 ]. Burkina Faso, Gambia, Ghana and Tanzania contributed two studies each [ 24 , 28 , 31 , 39 , 40 , 41 , 53 , 55 ]. Kenya and Uganda contributed three studies each [ 19 , 26 , 42 , 43 , 54 , 63 ] and Ethiopia had 14 studies [ 16 , 20 , 23 , 29 , 34 , 35 , 36 , 37 , 38 , 59 , 65 , 66 , 67 , 69 ]. Most of the studies were conducted in Nigeria with a total number of 19 studies [ 15 , 18 , 27 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 56 , 57 , 58 , 60 , 61 , 62 , 64 , 68 ]. The majority of the study respondents were parents of children who were within the childhood immunisation schedule range. The studies focused on factors that influence or determine childhood immunisation uptake. The majority of these studies were carried out mainly in West Africa (Nigeria) due to poor childhood immunisation uptake in the nation. This demonstrated that most of the researches were still attempting to determine the main factors that play a role in influencing childhood immunisation uptake, which would enable researchers to address the issues for the development of future health education intervention programs.

Study type and respondents

The majority of the studies were a cross-sectional study design with a total number of 42 studies, 3 qualitative study design [ 38 , 50 , 64 ], 4 mixed method [ 23 , 37 , 41 , 68 ] and 2 case-control [ 34 , 59 ]. The number of study respondents varied across the studies with 14 being the smallest and 7815 the largest (Table 3 ).

Factors associated with immunisation coverage

Modifiable factors.

The factors that are considered modifiable in this review are obstetric factors, maternal knowledge, maternal outcome expectation, maternal attitude, maternal self-efficacy and environmental factors. These factors were found to statistically influence childhood immunisation uptake in Africa and reported a wide range of AOR and 95% confidence intervals (upper limit and lower limit) from different studies.

In respect to obstetric factors, place of delivery was evident in five studies which are all statistically significant with AOR  2.11–3.13 and 95% confidence intervals (CI): 1.09, 7.13) [ 26 , 44 , 63 , 65 , 67 ] and three studies also reported postnatal care follow-up with AOR 1.8–5.8; 95% CI: 1.21, 3.16 [ 20 , 59 , 63 ]. Six studies revealed antenatal care follow-up with AOR  2.4–3.7; 95% CI: 1.1, 10.0 [ 16 , 19 , 65 , 66 , 67 , 69 ], likewise maternal tetanus toxoid was reported in two studies with AOR  2.43–3.2; 95% CI: 1.10, 10.00 [ 27 , 36 ].

Many studies reported factors relating to maternal knowledge, for example, knowledge on child immunisation (AOR = 3.3; 95% CI: 1.87, 7.43) [ 34 , 67 ], knowledge on preventive objective of immunisation ( p  < 0.05) [ 31 ], lack of knowledge on vaccines which accounted for 50% reason for non-compliance [ 57 ], knowledge on child vaccination schedule, which was reported in three studies with AOR  2.9–4.3; 95% CI: 1.42, 8.0 [ 26 , 35 , 59 ]. Likewise, awareness on immunisation and immunisation programs were also revealed in three studies with AOR  = 1.9–2.8; 95% CI: 1.44, 2.49 [ 20 , 25 , 37 ]. Two studies reported the impact of knowledge of VPDs (AOR  = 2.5; 95% CI: 1.5, 4.2) [ 16 , 61 ]. For maternal attitude and self-efficacy, three studies reported availability of child immunisation records with AOR  = 2.38–7.7; 95% CI: 1.43, 10. 06) [ 20 , 31 , 47 ]. Likewise, four studies documented fear of vaccine side effects (AOR  = 1.92; 95% CI: 1.01, 3.70) [ 23 , 57 , 60 , 62 ], negative beliefs towards childhood immunisation uptake [ 46 ], good perception of immunisation (AOR  = 2.60; 95% CI: 1.50, 4.51) [ 49 ] and confidence towards vaccine safety ( p  < 0.05) [ 46 ].

With regards to maternal outcome expectation, three studies reported knowledge on benefit of childhood immunisation uptake (AOR  = 5.51; 95% CI: 1.52, 19.94) [ 23 , 35 , 54 ] and one study documented severity of VPDs ( p  = 0.045) [ 45 ]. With regards to environmental factors, two studies reported religious belief (OR  = 1.65: 95% CI: 1.15, 2.36) [ 44 , 64 ]. Likewise, two studies also attributed cultural belief [ 56 , 60 ].

Non-modifiable factors

In this review, parental socio-demographic factors (maternal age, maternal education, paternal education, maternal marital status, area of residence, wealth index, number of siblings, religion, ethnicity and family income), child socio-demographic factors (gender, age) and environmental factors (distance to health facility, mode of transportation, accessibility of vaccination site, satisfaction with vaccine services, quality of vaccine provider clients relationship and availability of vaccine) are considered as non-modifiable factors. These factors were found to statistically influence childhood immunisation uptake in Africa and reported a wide range of AOR and 95% confidence intervals (upper limit and lower limit) from different studies.

In respect to parental socio-demographic factors, three studies reported maternal age with AOR  = 1.08–9.54; 95% CI: 1.08, 18.09; p  < 0.05 [ 23 , 36 , 38 ], nine studies reported maternal educational status which were all statistically significant with AOR  = 3.55–7.50; 95% CI: 1.02, 10.60 [ 15 , 18 , 26 , 55 , 65 , 66 , 67 , 69 ], three studies reported paternal education (AOR  = 3.1; 95% CI: 1.3, 7.4) [ 16 , 48 , 59 ], three studies reported maternal occupation and mothers’ marital status respectively (AOR  = 1.62–3.01; 95% CI: 1.0, 5.8) [ 16 , 24 , 58 , 65 ]. Another two studies reported area of residence (AOR  = 2.70; 95% CI: 1.52, 4.81) [ 18 , 26 ], wealth index was documented in three studies (AOR  = 1.4–2.38; 95% CI: 1.06, 3.73) [ 20 , 65 , 69 ], three studies reported family income (AOR  = 3.2; 95% CI: 1.4, 7.4) [ 16 , 31 , 59 ], one study reported number of siblings (APR  = 1.42; 95% CI: 1.05, 1.71) [ 25 ], two studies reported religion ( p  < 0.05) [ 19 , 44 ], three studies documented nomadic lifestyle (OR  = 11.06; 95% CI: 4.29, 28.54) [ 26 , 43 , 68 ] and ethnicity was also reported in two studies (AOR  = 2.47; 95% CI: 1.28, 4.76) [ 26 , 51 ]. Regarding child socio-demographic factors, child age was reported in two studies ( p  < 0.05) [ 25 , 56 ] whilst one study reported gender (OR  = 2.98; 95% CI: 1.21, 7.35) [ 46 ]. In relation to environmental factors, distance to health facility was attributed in four studies which were all statistically significant with AOR  = 2.11–3.10; 95% CI: 1.46, 6.30 [ 16 , 26 , 44 , 67 ], one study reported mode of transportation (AOR  = 1.54; 95% CI: 1.20, 1.97) [ 40 ], accessibility of vaccination site was also documented in one study (AOR  = 4.54; 95% CI: 2.34, 8.77) [ 29 ], four studies reported satisfaction with vaccine services (AOR  = 2.63; 95% CI: 1.1, 6.3) [ 28 , 56 , 60 , 64 ], quality of vaccine provider clients relationship was recorded in one study (AOR  = 1.86; 95% CI: 1.03, 3.50) [ 28 ] and vaccine availability in three studies [ 56 , 60 , 68 ].

The main reason for conducting this systematic review was to identify the factors that influence under-five childhood immunisation uptake among parents. In this review, many factors have been found to influence childhood immunisation uptake and are categorised and reviewed below.

Parental socio-demographic factors

Maternal age was revealed to be a factor influencing childhood immunisation uptake in a case-control study conducted in Ethiopia that involved 548 children aged between 12 to 23 months, in which mothers over 19 years of age were approximately 10 times more likely to have their children fully immunised compared to mothers under 19 years of age [ 23 ]. This may be due to knowledge gained over time on the importance of immunisation by mothers over 19 years of age, combined with the negative impact on children due to lack of immunisation. This finding is supported by two studies conducted in Ethiopia [ 36 , 37 ].

In this review, maternal education was the most common reported parental socio-demographic factor found to influence childhood immunisation. Mothers with at least a primary or secondary school education were found to be approximately eight times more likely to have their children fully immunised compared to mothers with no formal education [ 56 ]. This is more likely due to the fact that as the educational status of mothers improves, the seeking behaviour of children may perhaps increase, which in turn have positive impacts towards childhood immunisation uptake [ 56 ]. Furthermore, this could also be a result of changes that accompany maternal education, such as changes in attitudes, traditions and beliefs, increased autonomy and control over household resources which enhance healthcare seeking [ 26 ]. Similar findings have been reported in many studies [ 15 , 18 , 26 , 55 , 65 , 66 , 67 , 69 ].

From a cross-sectional study conducted in the Sinana District of Ethiopia, consisting of 591 children aged 12–23 months, paternal education was also found to be statistically associated with the child immunisation status. Children with fathers who had a secondary or higher educational level, were three times more likely to be fully vaccinated compared with children whose fathers had no formal education [ 16 , 48 , 59 ]. As the educational status of fathers improves, the seeking behaviour of children may perhaps increase, which in turn may have positive impacts towards childhood immunisation uptake [ 48 ].

Results from a cross-sectional study conducted in the Sinana District of Ethiopia, consisting of 591 children aged 12–23 months, has found maternal occupation to be statistically associated with child immunisation uptake. The proportion of children who were not fully vaccinated was found to be higher among mothers who were housewives [ 16 ]. Mothers whose occupation was in the farming sector, were less likely to be totally dependent on their spouses in terms of financial support and were found to be almost twice as likely to complete the immunisation of their children, compared with mothers who were housewives. A likely reason for this would be that mothers would not be reliant on their spouses to provide transport fees in order to take their children to be immunised. They most probably have been exposed to information on the benefit of childhood immunisation uptake through access to media [ 16 ]. This finding is supported by another cross-sectional study conducted in Ethiopia [ 65 ].

The marital status of a mother was also reported to have an influence towards childhood immunisation. In a descriptive cross-sectional study conducted in Ghana involving 280 mothers, it was found that divorced mothers were 3 times less likely to complete immunisation schedules of their children compared to mothers who were married [ 24 ]. In a cross-sectional study conducted in Nigeria involving 232 mothers (children aged 12–23 months), married women were observed to have a significantly adequate knowledge of immunisation which may increase the likelihood of achieving a higher rate of immunised children compared with their counterparts who were either single/divorced/widowed or separated [ 58 ]. The marital status of a mother may enhance her knowledge in the sense that those that are married may have more access to education compared to single mothers who may have other responsibilities and would instead tend to put their education aside in order to meet the needs of their children [ 24 ]. The supportive role of their partners may also enhance her knowledge if both partners jointly try to find ways to better the health status of their offspring [ 58 ].

A cross-sectional study from this review that was conducted in Kenya, which consisted of 298 mothers with children aged 12–23 months, demonstrated that area of residence statistically influenced childhood immunisation uptake. Children who lived in urban areas were 12 times more like to be vaccinated compared to children living in rural communities [ 26 ]. In Nigeria, results obtained using NDHS data involving 5754 children aged between 12 and 23 months, also revealed a statistical association between area of residence and child immunisation status. Children residing in urban areas were more likely to be vaccinated compared to children living in rural communities. This may probably be attributed to the fact that parents living in urban areas were more likely to be educated which may increase their knowledge towards the benefit of childhood immunisation uptake compared to parents living in rural areas [ 18 ].

Three different cross-sectional studies conducted in Ethiopia among children aged 12–23 months found wealth index to be a factor influencing childhood immunisation uptake. Children born to mothers from a rich index group were found to be twice as likely to be fully vaccinated compared with children from mothers from a poor wealth index group [ 20 , 65 , 69 ]. Rich parents may have more access to media and probably have more exposure to information on the benefit of childhood immunisation uptake.

Family income was found to be another factor influencing childhood immunisation uptake in a cross-sectional study conducted in Burkina Faso consisting of 591 children aged between 12 and 23 months. The study found that if the income of a family is greater than 1000 ETB or 52 USD, it increases the tendency of having children fully vaccinated in that family by approximately three times when compared to a poor family with a lesser income [ 16 ]. The family with a higher income may have easier access to Immunisation Centres due to accessible effective transportation options and would have less financial challenges when compared with families with a lower income. This finding was also supported by two cross-sectional study designs [ 31 , 59 ].

The number of siblings was also found to be a factor affecting childhood immunisation uptake in a cross-sectional study conducted in Angola involving 1209 children under 5 years of age. A family comprising 2 to 3 siblings were more likely to vaccinate their children compared with a family with less than 2 siblings [ 25 ]. This may be due to experience gained over time on the importance of immunisation as well as the medical complications that have occurred in children due to lack of immunisation.

Religion has been revealed by studies to be a factor influencing childhood immunisation uptake. From a cross-sectional study conducted in Uganda using Uganda Demographic Health Survey data, childhood immunisation uptake was affected by religious affiliations. Children from Muslim families had a lesser chance of been fully vaccinated compared with children from Catholic families [ 19 ]. Likewise, in Mozambique parents with no religious affiliation were found to be twice as likely not to complete their childhood immunisation uptake [ 44 ]. This could be due to the circulation of false information obtained via religious networks. This false information may be linked to negative beliefs of vaccines, for example, the belief that vaccines are composed of anti-fertility drugs [ 70 ].

Nomadic lifestyle was found to be associated with child immunisation uptake in Kenya. Children born to a family who practice a nomadic lifestyle, were found to be 11 times more likely not to be fully vaccinated compared with children born to a family that do not practice a nomadic lifestyle [ 26 , 43 , 68 ]. The family who practices a nomadic lifestyle may constantly change their location, switching from one place to another where immunisation services may not be readily accessible [ 26 ].

Ethnicity was found to be a factor affecting childhood immunisation uptake in Nigeria in which children belonging to the Igbo ethnic group were about three times more likely to be fully vaccinated compared to children belonging to an ethnic group such as Hausa, Yoruba and others [ 51 , 56 ]. These disparities could be attributed to the factors prevalent at community level, for example, in the Hausa community there is low level of education, high poverty, poor utilisation of antenatal care and home delivery and all these factors are associated with poor immunisation uptake. It could also be due to a misconception regarding the safety of vaccines and fear of vaccine side effects [ 51 ]. These findings are consistent with the findings from a systematic review conducted to determine the significant factors of adherence among parents or caregivers of under-five childhood immunisation schedules, even though the review did not specifically focus on Africa [ 71 ].

Child socio-demographic factors

Child gender was found to be associated with child immunisation uptake in Ibadan, Nigeria where a male child is about three times more likely to be immunised compared with a female child [ 46 ]. This may be attributed to the beliefs of parents that immunisation will have negative impacts on their daughters when they reach child bearing age.

A contradictory finding was revealed with regards to influence of child age on immunisation uptake. In Nigeria, higher immunisation uptake was observed in children above 1 year of age (84.4%) compared with children below 1 year (63.6%) and this could be due to the fact that some mothers delay child immunisation until their children reach a certain age, due to negative beliefs that their children are too young to be immunised [ 56 ]. In Angola, higher immunisation uptake was seen in children who were 1 year of age or less compared with children that were above 1 year of age. This could be attributed to the lectures received by mothers towards benefits of timely childhood immunisation uptake during their antenatal and postnatal care. It is likely that the content of health education delivered to the mothers has changed with more emphasis placed on the benefit of childhood immunisation uptake [ 25 ].

Obstetric factors

Antenatal care follow-up (ANC) was found to be a factor influencing childhood immunisation uptake in a cross-sectional study conducted in Ethiopia that involved 591 children aged between 12 and 23 months. The study showed mothers who frequently attend ANC during their pregnancy were about four times more likely to have their children fully vaccinated compared with mothers who did not attend ANC regularly [ 16 ]. This finding is supported by the findings of a study conducted in Uganda and four more studies from Ethiopia [ 19 , 65 , 66 , 67 , 69 ]. Mothers who frequented health facilities during pregnancy may have received counselling on childhood immunisation where the importance of timely childhood immunisation uptake may be prioritised regularly [ 16 ].

The postnatal check-up was also found to be an influencing factor towards childhood uptake where children who received a check-up within 2 months after birth were twice more likely to be fully vaccinated compared to those who did not receive a check-up after delivery [ 20 , 59 , 63 ]. This can be attributed to educational sessions that mothers were exposed to during postnatal visits where the importance of timely immunisation of the baby may be emphasised [ 20 ].

The maternal tetanus toxoid (MTT) vaccine was also noted to be a factor influencing childhood immunisation uptake. Mothers who received at least one dose of the MTT vaccine were three times more likely to have their children fully immunised compared with mothers who did not receive any dose of the MTT vaccine [ 27 , 36 ]. This could be attributed to the knowledge that mothers may have obtained regarding the benefit of childhood immunisation uptake during the MTT vaccination in their health centre [ 27 ].

Children who were delivered in hospitals were more likely to have a complete vaccination status compared with children delivered at home [ 26 , 44 , 63 ]. Mothers who had hospital deliveries may receive advice after delivery where the importance of timely immunisation of the baby may be emphasised and therefore, they are more likely to receive their vaccines [ 44 ].

Maternal knowledge

Having a good maternal knowledge on child immunisation was revealed to be a predictor for childhood immunisation uptake in a case-control study conducted in Northern Ethiopia. Children of mothers with a good knowledge on childhood immunisation were found to be three times more likely to be completely immunised compared with children whose mothers had a poor knowledge on childhood immunisation [ 34 ]. This finding is consistent with the previous systematic review conducted in Sub-Saharan Africa [ 72 ].

In Burkina Faso, mothers with a knowledge on preventive objectives of immunisation were more likely to have their children immunised compared with mothers with limited knowledge [ 31 ]. In a household-level cluster survey consisting of 7815 children conducted in Nigeria that involved 40 polio high-risk districts of Nigeria, lack of maternal knowledge regarding vaccines was found to be the main reason contributing to poor childhood immunisation uptake which accounted for 50% of reasons for non-vaccination [ 57 ].

Maternal knowledge on vaccines and VPDs were also shown to influence childhood immunisation uptake in Southeast Ethiopia, where children whose mothers had a good knowledge on vaccines and VPDs were found to be three times more likely to be fully vaccinated compared with children of mothers who had a poor knowledge of vaccines and VPDs [ 16 ]. A similar finding was also found in Nigeria [ 61 ]. Maternal knowledge on child vaccine schedules was revealed to statistically influence child immunisation uptake where mothers who had knowledge on schedules of vaccines were found to be four times more likely to fully immunise their children compared with mothers who had no knowledge of vaccine schedules [ 26 , 35 , 59 ]. Mothers with knowledge of immunisation schedules may know the exact time for each childhood immunisation uptake and they might also know the benefit of timely immunisation uptake for their children. Parents who were aware of immunisation and immunisation programs were three times more likely to have their children immunised compared with their counterparts [ 20 , 25 , 37 ]. Childhood immunisation uptake can be negatively affected by the level of knowledge of mothers. The more knowledge they acquire, the higher the tendency of increasing their confidence towards childhood immunisation uptake.

Maternal attitude and self-efficacy

Studies have shown that mothers who have their child immunisation records were more likely to have their children fully immunised compared with mothers without child immunisation records [ 20 , 31 , 47 ]. In Nigeria, the poor attitude of mothers accounted for 16% of the reasons for poor childhood immunisation uptake [ 57 ]. Among 248 defaulting mothers in Ibadan, Nigeria, more than half of this group reported that the reason for defaulting was that they considered childhood immunisation to be a waste of time [ 46 ]. The children whose mothers had a positive perception towards vaccine side effects, were twice more likely to be fully immunised compared with children whose mothers had a negative perception towards vaccine side effects [ 23 ]. Many studies also reported fear of vaccine side effects influenced immunisation uptake [ 57 , 60 , 62 ]. Mothers who lacked confidence in vaccine safety, were less likely to have their children immunised [ 46 ]. Mothers with a good perception on immunisation were three times more likely to have their children fully immunised compared with mothers with a poor perception on immunisation [ 49 ]. The attitudes of mothers towards childhood immunisation uptake is influenced by their perception. This in turn can decrease their confidence regarding childhood immunisation uptake.

Maternal outcome expectations

Mothers who knew the benefits of childhood immunisation were six times more likely to have their children fully immunised compared with their counterparts [ 23 , 35 ]. Having an expectation towards the protection that follows childhood immunisation significantly influences childhood immunisation [ 54 ]. Furthermore, knowing the seriousness of VPDs was also found to be a predictor for non-compliance [ 45 ]. The more knowledge mothers acquired with regards to the benefit of child immunisation and consequences of not immunising a child, the higher the tendency of increasing their practice confidence towards childhood immunisation uptake. This finding is consistent with the findings from a systematic review previously conducted that aimed to identify factors associated with immunisation coverage in poor urban areas and slums [ 73 ].

Environmental factors

Environmental factors are classified as social factors and health care systems or logistic factors. Religious belief was revealed to be one of the social factors influencing childhood immunisation uptake in Mozambique. Mothers who considered immunisation as unacceptable in their religion were less likely to have their children fully immunised compared with mothers who did not consider immunisation as unacceptable in their religion [ 44 ]. Lack of adequate involvement by religious and traditional leaders in immunisation activities was found to be a reason for immunisation failure in Borno State, Nigeria [ 64 ].

In Nigeria, cultural beliefs against immunisation are found to be destructive towards childhood immunisation uptake [ 56 , 60 ]. This could probably be due to the circulation of false information via the use of either family or religious networks towards vaccines. For example, beliefs that vaccines were composed of anti-fertility drugs and therefore could destroy the eggs of females and cause damage to the reproductive system [ 70 ]. Traditional and religious leaders are highly respected and are generally regarded and accepted as the custodians of traditions entrusted to them to provide traditional guidance to their respective communities. Therefore, their involvement in immunisation activities will help increase immunisation acceptance and uptake since the community trust their views on various matters [ 64 ].

In Kenya, distance to health facilities was found to affect childhood immunisation uptake among 298 respondents. Children belonging to mothers or caregivers who travelled a short distance to the health facility for immunisation were 18 times more likely to be fully vaccinated compared with children whose mothers or caregivers travelled further to a health facility for their children’s immunisations [ 26 ]. This was supported by three studies conducted in Mozambique and Ethiopia [ 16 , 44 , 67 ]. These findings were in line with the previous systematic review conducted in Sub-Saharan Africa [ 72 ]. The mode of transportation for immunisation was also found to be an influencing factor where mothers who make use of public transport were twice more likely to have their children fully immunised compared to mothers who walked [ 40 ].

Accessibility of vaccination sites was found to be a predictor for childhood immunisation uptake in Southern Ethiopia. Mothers who considered immunisation sites to be accessible were 5 times more likely to have their children fully immunised compared to mothers who did not consider it accessible [ 29 ].

Satisfaction with vaccine services was also found to influence childhood immunisation uptake in Tanzania among 380 mothers of children aged 12–23 months. Mothers who are satisfied with vaccine services were about three times more likely to have their children vaccinated compared with mothers who were unsatisfied with vaccine services [ 28 , 56 , 60 , 64 ]. The way vaccine providers behave could either enhance or discourage mothers from taking their children for vaccinations [ 28 ].

The quality of the vaccine provider and client relationship was also found to be a predictor for childhood immunisation uptake in Tanzania among 380 participants. Mothers who had a positive perception towards quality of vaccine provider and client relationship were twice more likely to have their children fully immunised compared to mothers who had a negative perception towards quality of vaccine provider and client relationship [ 28 ]. This could probably be due to the way vaccine providers behave which may either enhance or discourage mothers from taking their children for vaccinations [ 28 ].

In Nigeria, the unavailability of vaccines when required was also found to be another reason for defaulting on childhood immunisation uptake [ 56 , 60 , 68 ]. Mothers may have spent a considerable amount of money in order to access health care on several occasions. However, the service was not always available which resulted in them becoming discouraged and they failed to complete the immunisation uptake of their children [ 56 ].

The findings of the study will benefit policy makers in making decisions and formulating appropriate guidelines and policies with regards to childhood immunisation uptake in Africa. These findings can also enable other researchers to plan an appropriate health education intervention encompassing an appropriate health behavioural theory to address the factors affecting childhood immunisation uptake among African countries. The strengthening of childhood immunisation policies, strategies and further research examining the causal relationship on childhood immunisation in Africa are required.

Social cognitive theory (SCT) is one of the health theories commonly used in health education interventions [ 74 , 75 ] which describes human behaviour through the influence of personal and environmental factors [ 76 ]. The theory accounts for human behaviour, cognition and environment and is the only health theory that takes into account reciprocal interaction, unlike other theories such as Information-Motivation-Behavioural Skills model [ 74 , 77 ]. In this systematic review, there are many factors that has been found to influence childhood immunisation uptake in Africa such as personal factors (parental sociodemographic factors, obstetric factors, knowledge gaps, negative attitudes, outcome expectation and lack of self-efficacy) and environmental factors (social factors and healthcare system) [ 16 , 18 , 19 , 20 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 , 49 , 50 , 51 , 52 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 , 62 , 63 , 64 , 70 ]. Therefore, using this theory may enable researchers to address both the personal and environmental factors influencing childhood immunisation uptake.

The majority of our included studies achieved at least high scores [ 16 , 18 , 20 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 40 , 42 , 43 , 44 , 45 , 48 , 50 , 51 , 52 , 55 , 57 , 58 , 59 , 61 , 62 , 63 , 64 , 65 , 66 , 67 , 68 , 69 ] and the overall risk of bias was moderate in ten studies [ 15 , 19 , 39 , 46 , 47 , 49 , 53 , 54 , 56 , 60 ]. Moreover, none of the included studies revealed either a serious or low risk of bias.

Our study has acknowledged some limitations. This review may be prone to publication bias as grey literature/unpublished literature and studies that are not published in the English language were not considered. Conducting a quantitative meta-analysis in this systematic review may be very important for analysing quantitative trends. The majority of the literature cited in this review is observational in nature and therefore, this study is lacking evidence for causation. Furthermore, many studies relied on survey data which may increase the risk of nonresponse bias or selection bias.

In conclusion, various factors influencing childhood immunisation uptake in Africa were identified from this systematic review. The factors were categorised into two main groups, modifiable and non-modifiable factors which were later divided further. The modifiable factors (obstetric history, maternal knowledge, maternal attitude and self-efficacy and maternal outcome expectation) were revealed as having a direct relationship with the childhood immunisation uptake. Many factors and results attained from this review could enable the researchers to further understand and develop necessary intervention in order to address the issue of the factors influencing childhood immunisation uptake. Finally, we recommend an immunisation health education intervention among pregnant women focusing on the significant findings from this systematic review which may hopefully improve childhood immunisation uptake among countries with poor coverage in Africa.

Availability of data and materials

All data generated during this study are included in this article.

Abbreviations

Adjusted Odd Ratio

Confidence Interval

Adjusted Prevalence Rate

Ethiopia Demographic Health Surveillance

Nigeria Demographic Health Surveillance

Farafenni Health and Demographic Surveillance

Nairobi Urban Health and Demographic Surveillance System

Tanzania Health and Demographic Surveillance System

Nouna Health and Demographic Surveillance System

Uganda Demographic Health Surveillance System

Tetanus Toxoid

Antenatal Care

Postnatal Care

Vaccine Preventable Diseases

Expanded Program on Immunization

National Program on Immunization

Routine Immunization

Health Extension Workers

Oral Polio Vaccine

Expanded Programme on Immunization

Adjusted Prevalence Ratio

Centre for Global Development. Making markets for vaccines: from ideas to actions. Washington DC: Centre for Global Development; 2005.

Google Scholar  

World Health Organization, UNICEF. Global immunization data, 2014. http://www.who.int/immunization/monitoring_surveillance/global_immunization_data.pdf?ua=1 .

Yeung K, Duclos P, Nelson E, Hutubessy R. An update of the global burden of pertussis in children younger than 5 years: a modelling study. Lancet Infect Dis. 2017;17(9):974–80. https://doi.org/10.1016/s1473-3099(17)30390-0 .

Article   PubMed   Google Scholar  

Awosika D. Access to immunization and other public health interventions through the pharmacists. West Afr J Pharm. 2012;23(1):3–11.

World Health Organization, 2020. Immunization coverage. Key facts. Retrieved from https://www.who.int/news-room/fact-sheets/detail/immunization-coverage

World Health Organization, 2018. Immunization. Retrievd from http://www.who.int/topics/immunization/en/

Crowcroft N, Stein C, Duclos P, Birmingham M. How best to estimate the global burden of pertussis? Lancet Infect Dis. 2003;3(7):413–8. https://doi.org/10.1016/S1473-3099(03)00669-8 .

Article   CAS   PubMed   Google Scholar  

Nigeria Demographic and Health Survey. Key indicators report. Nigeria: National Population Commission Abuja; 2018.

Ethiopia Mini Demographic and Health Survey 2019. Key indicators. Ethiopian Public health Institute Addis Ababa. The Federal Ministry of Health Addis Ababa.

Uganda Demographic and Health Survey. Key indicators report. Uganda: Uganda Bureau of Statistics Kampala; 2016.

Ghana Demographic and Health Survey. Ghana statistical service Accra, Ghana. Ghana: Ghana Health Service Accra; 2014.

Tanzania Demographic and Health Survey and Malaria Indicator Survey 2016. Final Report. Ministry of Health, Community Development, Gender, Elderly and Children Dar Es Salaam. Ministry of Health Zanzibar. National Bureau of Statistics Dar Es Salaam.

Kenya Demographic and Health Survey. Kenya National Bureau of statistics Nairobi, Kenya. Kenya: Ministry of Health Nairobi; 2014.

World Health Organization 2015. Immunization, vaccines and biologicals. Retrieved from https://www.who.int/immunization/global_vaccine_action_plan/en/

Gidado S, Nguku P, Biya O, Waziri N, Mohammed A, Nsubuga P, et al. Determinants of routine immunization coverage in Bungudu, Zamfara state, northern Nigeria, may 2010. Pan Afr Med J. 2014;18. https://doi.org/10.11604/pamj.supp.2014.18.1.4149 .

Legesse E, Dechasa W. An assessment of child immunization coverage and its determinants in Sinana District, Southeast Ethiopia. BMC Pediatr. 2015;15(1):31.

Article   PubMed   PubMed Central   Google Scholar  

Kitamura. Factors affecting childhood immunization in Lao People's Democratic Republic: A cross-sectional study from nationwide, population-based, multistage cluster sampling. BioSci Trends. 2013;7(4):178–85.

PubMed   Google Scholar  

Adedokun S, Uthman O, Adekanmbi V, Wiysonge C. Incomplete childhood immunization in Nigeria: a multilevel analysis of individual and contextual factors. BMC Public Health. 2017;17(1):236.

Bbaale E. Factors Influencing Childhood Immunization in Uganda. J Health Popul Nutr. 2013;31(1):118–9.

Lakew Y, Bekele A, Biadgilign S. Factors influencing full immunization coverage among 12–23 months of age children in Ethiopia: evidence from the national demographic and health survey in 2011. BMC Public Health. 2015;15(1):728.

Rahman M, Obaida-Nasrin S. Factors affecting acceptance of complete immunization coverage of children under five years in rural Bangladesh. Salud Pública de México. 2010;52(2).

Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ. 2009;21(339):b2535.

Article   Google Scholar  

Negussie A, Kassahun W, Assegid S, Hagan A. Factors associated with incomplete childhood immunization in Arbegona district, southern Ethiopia: a case – control study. BMC Public Health. 2015;16(1):27.

Anokye R, Acheampong E, Budu-Ainooson A, Edusei A, Okyere P, Dogbe J, et al. Socio-demographic determinants of childhood immunization incompletion in Koforidua, Ghana. BMC Res Notes. 2018;11(1):656.

Oliveira M, Martinez E, Rocha J. Factors associated with vaccination coverage in children < 5 years in Angola. Rev Saude Publica. 2014;48(6):906–15. https://doi.org/10.1590/s0034-8910.2014048005284 .

Kiptoo E. Factors Influencing Low Immunization Coverage Among Children Between 12–23 Months in East Pokot, Baringo Country, Kenya. Int J Vaccines & Vaccination. 2015;1(2).

Adedire E, Ajayi I, Fawole O, Ajumobi O, Kasasa S, Wasswa P, et al. Immunisation coverage and its determinants among children aged 12–23 months in Atakumosa-west district, Osun State Nigeria: a cross-sectional study. BMC Public Health. 2016;16(1):905.

Chambongo P, Nguku P, Wasswa P, Semali I. Community vaccine perceptions and its role on vaccination uptake among children aged 12-23 months in the Ileje District, Tanzania: a cross section study. Pan Afr Med J. 2016;23. https://doi.org/10.11604/pamj.2016.23.162.8925 .

Animaw W, Taye W, Merdekios B, Tilahun M, Ayele G. Expanded program of immunization coverage and associated factors among children age 12–23 months in Arba Minch town and Zuria District, Southern Ethiopia, 2013. BMC Public Health. 2014;14(1):464.

Moola S, Munn Z, Tufanaru C, Aromataris E, Sears K, Sfetcu R, et al. Systematic reviews of etiology and risk. In: Aromataris E, Munn Z, editors. Faculty of Health Sciences, University Adelaide Australia: Joanna Briggs Institute Reviewer’s Manual: The Joanna Briggs Institute; 2017.

Sanou A, Simboro S, Kouyaté B, Dugas M, Graham J, Bibeau G. Assessment of factors associated with complete immunization coverage in children aged 12–23 months: a cross-sectional study in Nouna district, Burkina Faso. BMC Int Health Human Rights. 2009;9(S1):S10.

Chiabi A, Nguefack F, Njapndounke F, Kobela M, Kenfack K, Nguefack S, et al. Vaccination of infants aged 0 to 11 months at the Yaounde Gynaeco-obstetric and pediatric hospital in Cameroon: how complete and how timely? BMC Pediatr. 2017;17(1):206.

Mwamba G, Yoloyolo N, Masembe Y, Nsambu M, Nzuzi C, Tshekoya P, et al. Vaccination coverage and factors influencing routine vaccination status in 12 high risk health zones in the province of Kinshasa City, Democratic Republic of Congo (DRC), 2015. Pan Afr Med J. 2017;27. https://doi.org/10.11604/pamj.supp.2017.27.3.11930 .

Aregawi H, Gebrehiwot T, Abebe Y, Meles K, Wuneh A. Determinants of defaulting from completion of child immunization in Laelay Adiabo District, Tigray region, northern Ethiopia: a case-control study. PLoS One. 2017;12(9):e0185533. https://doi.org/10.1371/journal.pone.0185533 .

Article   CAS   PubMed   PubMed Central   Google Scholar  

Etana B, Deressa W. Factors associated with complete immunization coverage in children aged 12–23 months in Ambo Woreda, Central Ethiopia. BMC Public Health. 2012;12(1):566.

Mohamud A, Feleke A, Worku W, Kifle M, Sharma H. Immunization coverage of 12–23 months old children and associated factors in Jigjiga District, Somali National Regional State, Ethiopia. BMC Public Health. 2014;14(1):865.

Abebe A, Mengistu T, Mekuria A. Measles case, immunization coverage and its determinant factors among 12–23 month children, in Bassona Worena Woreda, Amhara Region, Ethiopia, 2018. BMC Res Notes. 2019;12(1):17.

Article   CAS   Google Scholar  

Zewdie A, Letebo M, Mekonnen T. Reasons for defaulting from childhood immunization program: a qualitative study from Hadiya zone, Southern Ethiopia. BMC Public Health. 2016;16(1):1240.

Payne S, Townend J, Jasseh M, Lowe Jallow Y, Kampmann B. Achieving comprehensive childhood immunization: an analysis of obstacles and opportunities in the Gambia. Health Policy Plan. 2013;29(2):193–203. https://doi.org/10.1093/heapol/czt004 .

Odutola A, Afolabi M, Ogundare E, Lowe-Jallow Y, Worwui A, Okebe J, et al. Risk factors for delay in age-appropriate vaccinations among Gambian children. BMC Health Serv Res. 2015;15(1):1240.

Bosu W, Ahelegbe D, Edum-Fotwe E, Bainson KA, Kobina Turkson P. Factors influencing attendance to immunization sessions for children in a rural district of Ghana. Acta Trop. 1997;68(3):259–67. https://doi.org/10.1016/S0001-706X(97)00094-6 .

Mutua M, Kimani-Murage E, Ettarh R. Childhood vaccination in informal urban settlements in Nairobi, Kenya: Who gets vaccinated? BMC Public Health. 2011;11(1):6.

Pertet A, Kaseje D, Otieno-Odawa C, Kirika L, Wanjala C, Ochieng J, et al. Under vaccination of children among Maasai nomadic pastoralists in Kenya: is the issue geographic mobility, social demographics or missed opportunities? BMC Public Health. 2018;18(1):1389.

Jani J, De Schacht C, Jani I, Bjune G. Risk factors for incomplete vaccination and missed opportunity for immunization in rural Mozambique. BMC Public Health. 2008;8(1):161.

Umeh G, Nomhwange T, Shamang A, Zakari F, Musa A, Dogo P, et al. Attitude and subjective wellbeing of non-compliant mothers to childhood oral polio vaccine supplemental immunization in Northern Nigeria. BMC Public Health. 2018;18(1):231.

Oladokun R, Adedokun O, Lawoyin O. Children not receiving adequate immunization in Ibadan, Nigeria: what reasons and beliefs do their mothers have. Niger J Clin Pract. 2010;13(2):173–8.

CAS   PubMed   Google Scholar  

Babalola S. Determinants of the uptake of the full dose of diphtheria–pertussis–tetanus vaccines (DPT3) in northern Nigeria: a multilevel analysis. Matern Child Health J. 2008;13(4):550–8. https://doi.org/10.1007/s10995-008-0386-5 .

Odusanya O, Alufohai E, Meurice F, Ahonkhai V. Determinants of vaccination coverage in rural Nigeria. BMC Public Health. 2008;8(1):381.

Taiwo L, Idris S, Abubakar A, Nguku P, Nsubuga P, Gidado S, et al. Factors affecting access to information on routine immunization among mothers of under 5 children in Kaduna state Nigeria, 2015. Pan Afr Med J. 2017;27. https://doi.org/10.11604/pamj.2017.27.186.11191 .

Oku A, Oyo-Ita A, Glenton C, Fretheim A, Eteng G, Ames H, et al. Factors affecting the implementation of childhood vaccination communication strategies in Nigeria: a qualitative study. BMC Public Health. 2017;17(1):200.

Antai D. Inequitable childhood immunization uptake in Nigeria: a multilevel analysis of individual and contextual determinants. BMC Infect Dis. 2009;9(1):181.

Ijarotimi I, Fatiregun A, Adebiyi O, Ilesanmi O, Ajumobi O. Urban–rural differences in immunisation status and associated demographic factors among children 12-59 months in a southwestern state, Nigeria. PLOS ONE. 2018;13(11):e0206086. https://doi.org/10.1371/journal.pone.0206086 .

Semali I. Trends in Immunization Completion and Disparities in the Context of Health Reforms: The case study of Tanzania. BMC Health Serv Res. 2010;10(1):299.

Vonasek B, Bajunirwe F, Jacobson L, Twesigye L, Dahm J, Grant M, et al. Do maternal knowledge and attitudes towards childhood immunizations in rural Uganda correlate with complete childhood vaccination? Ann Global Health. 2016;82(3):567. https://doi.org/10.1016/j.aogh.2016.04.523 .

Kagoné M, Yé M, Nébié E, Sie A, Schoeps A, Becher H, et al. Vaccination coverage and factors associated with adherence to the vaccination schedule in young children of a rural area in Burkina Faso. Glob Health Action. 2017;10(1):1399749. https://doi.org/10.1080/16549716.2017.1399749 .

Duru C, Iwu A, Uwakwe K, Diwe K, Merenu I, Emerole C, et al. Assessment of immunization status, coverage and determinants among under 5-year-old children in Owerri, Imo state, Nigeria. OALib. 2016;03(06):1–17.

Gunnala R, Ogbuanu I, Adegoke O, Scobie H, Uba B, Wannemuehler K, et al. Routine vaccination coverage in northern Nigeria: results from 40 district-level cluster surveys, 2014-2015. PLoS One. 2016;11(12):e0167835. https://doi.org/10.1371/journal.pone.0167835 .

Chris-Otubor G, Dangiwa D, Ior D, Anukam C. Assessment of knowledge, attitudes and practices of mothers in Jos north regarding immunization. Int Organ Scientific Res J Pharm. 2015;5(6):34–45.

Tadesse H, Deribew A, Woldie M. Predictors of defaulting from completion of child immunization in south Ethiopia. A case control study. BMC Public Health. 2009;9(1). https://doi.org/10.1186/1471-2458-9-150 .

Kio O, Agbede O, Mkpuruoma J. Knowledge beliefs and practices regarding immunization among nursing mothers in Nigeria. J Health Med Nursing. 2016;24:145–9.

Awosan K, Ibrahim M, Yunusa E, Isah B, Raji M, Abubakar N. Knowledge, Attitude and Compliance with Full Immunization of Children Against Vaccine Preventable Diseases among Pregnant Mothers in Sokoto, Nigeria. Int J Contemp Med Res [IJCMR]. 2018;5(6).

Ekure E, Balogun M, Mukhtar Y, Ojo O, Emodi I, et al. Community knowledge, attitude and practice of childhood immunization in Southwest Nigeria. Nigerian J Paediatr. 2013;40(2):106–11. https://doi.org/10.4314/njp.v40i2.1 .

Canavan M, Sipsma H, Kassie G, Bradley E. Correlates of complete childhood vaccination in east african countries. PLoS One. 2014;9(4):e95709. https://doi.org/10.1371/journal.pone.0095709 .

Omotara A, Okujagu F. Assessment of knowledge, attitude and practice of stakeholders towards immunization in Borno state, Nigeria: A qualitative approach. J Comm Med Health Educ. 2012;2(9). https://doi.org/10.4172/2161-0711.1000181 .

Tamirat K, Sisay M. Full immunization coverage and its associated factors among children aged 12–23 months in Ethiopia: further analysis from the 2016 Ethiopia demographic and health survey. BMC Public Health. 2019;19(1):1019.

Kinfe Y, Gebre H, Bekele A. Factors associated with full immunization of children 12–23 months of age in Ethiopia: a multilevel analysis using 2016 Ethiopia Demographic and health Survey. PLoS One. 2019;14(11):e0225639. https://doi.org/10.1371/journal.pone.0225639 .

Girmay A, Dadi A. Full immunization coverage and associated factors among children aged 12-23 months in a hard-to-reach areas of Ethiopia. Int J Pediatr. 2019;2019:1–8. https://doi.org/10.1155/2019/1924941 .

Akwataghibe N, Ogunsola E, Broerse J, Popoola O, Agbo A, Dieleman M. Exploring factors influencing immunization utilization in Nigeria—a mixed methods study. Front Public Health. 2019;7. https://doi.org/10.3389/fpubh.2019.00392 .

Mekonnen Z, Gelaye K, Martin W, Binyam T. Timely completion of vaccination and its determinants among children in northwest, Ethiopia: a multilevel analysis. BMC Public Health. 2020;20:908.

Jegede A. What led to the Nigerian boycott of the polio vaccination campaign? PLoS Med. 2007;4(3):e73. https://doi.org/10.1371/journal.pmed.0040073 .

Zulkefli N, Mohammad R, Juni M. Adherence among parents on under-five childhood immunization schedule: a review. Int J Public Health Clin Sci. 2016;3(6).

Bangura J, Xiao S, Qiu D, Ouyang F, Chen L. Barriers to childhood immunization in sub-Saharan Africa: A systematic review. BMC Public Health. 2020;20(1):1108.

Crocker-Buque T, Mindra G, Duncan R, Mounier-Jack S. Immunization, urbanization and slums – a systematic review of factors and interventions. BMC Public Health. 2017;17(1):556.

Bandura A. Social cognitive theory of mass communication. Media effects. Adv Theory Res. 2008:94–124.

Bagherniya M, Sharma M, Darani FM, Maracy MR, Safarian M, Birgani RA. School-based nutrition education intervention using social cognitive theory for overweight and obese Iranian adolescent girls: a cluster randomized controlled trial; 2018. https://doi.org/10.1177/0272684X17749566 .

Book   Google Scholar  

Bandura A. Social foundations of thought and action: a social cognitive theory: Englewood Cliffs Prentice-Hall; 1986.

Fisher J, Fisher W. Changing AIDS risk behavior. Psychol Bull. 1992;111(3):455–74.

Download references

Acknowledgements

The authors gratefully acknowledge electronic database searches done using Sultan Abdul Samad Library, Universiti Putra Malaysia.

No external funding was received for this systematic review.

Author information

Authors and affiliations.

Department of Community Health, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Malaysia

Abubakar Nasiru Galadima, Nor Afiah Mohd Zulkefli, Salmiah Md Said & Norliza Ahmad

You can also search for this author in PubMed   Google Scholar

Contributions

ANG conceived the study with NZ, and SS advised on the methods. ANG undertook the searches and titles for screening. ANG, NZ and SS agreed final inclusions and undertook data extraction. ANG, NZ, SS and NA contributed to write-up and editing of the final paper. All authors have read and approved the final version of this manuscript.

Corresponding author

Correspondence to Nor Afiah Mohd Zulkefli .

Ethics declarations

Ethics approval and consent to participate.

Not applicable.

Consent for publication

Competing interests.

The authors declare that they have no competing interests.

Additional information

Publisher’s note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: supplemental file 1..

Database search terms used; Fig. 1: PRISMA flow diagram for the identification, screening, eligibility, and inclusion of studies; Table 1: Eligibility criteria table; Table 2: Operational definition; Table 3: Summary of systematic review factors influencing childhood immunisation uptake.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ . The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/ ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Cite this article.

Galadima, A.N., Zulkefli, N.A.M., Said, S.M. et al. Factors influencing childhood immunisation uptake in Africa: a systematic review. BMC Public Health 21 , 1475 (2021). https://doi.org/10.1186/s12889-021-11466-5

Download citation

Received : 26 June 2020

Accepted : 06 July 2021

Published : 28 July 2021

DOI : https://doi.org/10.1186/s12889-021-11466-5

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Childhood immunisation

BMC Public Health

ISSN: 1471-2458

literature review on immunization

AIP Publishing Logo

  • Previous Article
  • Next Article

Vaccine safety and efficacy: A literature review

[email protected]

[email protected]

  • Article contents
  • Figures & tables
  • Supplementary Data
  • Peer Review
  • Reprints and Permissions
  • Cite Icon Cite
  • Search Site

Zijun He , Mengshu He , Emily Yuan; Vaccine safety and efficacy: A literature review. AIP Conf. Proc. 27 April 2021; 2350 (1): 020011. https://doi.org/10.1063/5.0049198

Download citation file:

  • Ris (Zotero)
  • Reference Manager

To introduce vaccine safety and efficacy, the two main public concerns for vaccine use. This is a review of the literature including but not limited to scientific publications and government documents that are related to vaccine safety and efficacy. The publication dates range from 1984 to 2020. Vaccine safety and efficacy are the two main concerns of vaccine use. The Food and Drug Administration (FDA) has a rigid policy for vaccine licensure and strict surveillance after vaccine deployment to ensure the safety of the vaccine. Vaccine efficacy is a critical criterion of the vaccine pre-licensure clinical trials and post-licensure surveillance. Double-blind, randomized, and clinical controlled studies and case-controlled studies are the two main methods to evaluate the vaccine efficacy. In this study, knowledge of vaccine safety and efficacy from numerous studies and researches are combined to provide an overall view and facilitate the understanding of vaccines. Vaccine administration is essentially a parental or personal decision. It is important to inform the public about the importance and benefits of vaccinations. As the number of vaccines increases, there is a need for a universal framework for vaccine assessment to facilitate international communications and encourage interdisciplinary studies. Vaccine inventions for contagious diseases like COVID-19 are urgent.

Citing articles via

Publish with us - request a quote.

literature review on immunization

Sign up for alerts

  • Online ISSN 1551-7616
  • Print ISSN 0094-243X
  • For Researchers
  • For Librarians
  • For Advertisers
  • Our Publishing Partners  
  • Physics Today
  • Conference Proceedings
  • Special Topics

pubs.aip.org

  • Privacy Policy
  • Terms of Use

Connect with AIP Publishing

This feature is available to subscribers only.

Sign In or Create an Account

  • Research article
  • Open access
  • Published: 21 June 2008

Too little but not too late: Results of a literature review to improve routine immunization programs in developing countries

  • Tove K Ryman 1 ,
  • Vance Dietz 1 &
  • K Lisa Cairns 1  

BMC Health Services Research volume  8 , Article number:  134 ( 2008 ) Cite this article

17k Accesses

58 Citations

6 Altmetric

Metrics details

Globally, immunization services have been the center of renewed interest with increased funding to improve services, acceleration of the introduction of new vaccines, and the development of a health systems approach to improve vaccine delivery. Much of the credit for the increased attention is due to the work of the GAVI Alliance and to new funding streams. If routine immunization programs are to take full advantage of the newly available resources, managers need to understand the range of proven strategies and approaches to deliver vaccines to reduce the incidence of diseases. In this paper, we present strategies that may be used at the sub-national level to improve routine immunization programs.

We conducted a systematic review of studies and projects reported in the published and gray literature. Each paper that met our inclusion criteria was rated based on methodological rigor and data were systematically abstracted. Routine-immunization – specific papers with a methodological rigor rating of greater than 60% and with conclusive results were reported.

Greater than 11,000 papers were identified, of which 60 met our inclusion criteria and 25 papers were reported. Papers were grouped into four strategy approaches: bringing immunizations closer to communities (n = 11), using information dissemination to increase demand for vaccination (n = 3), changing practices in fixed sites (n = 4), and using innovative management practices (n = 7).

Immunization programs are at a historical crossroads in terms of developing new funding streams, introducing new vaccines, and responding to the global interest in the health systems approach to improving immunization delivery. However, to complement this, actual service delivery needs to be strengthened and program managers must be aware of proven strategies. Much was learned from the 25 papers, such as the use of non-health workers to provide numerous services at the community level. However it was startling to see how few papers were identified and in particular how few were of strong scientific quality. Further well-designed and well-conducted scientific research is warranted. Proposed areas of additional research include integration of additional services with immunization delivery, collaboration of immunization programs with new partners, best approaches to new vaccine introduction, and how to improve service delivery.

Peer Review reports

Immunizations are a cornerstone of public health: the World Health Organization (WHO) estimates, that in 2006, immunizations saved two to three million lives [ 1 ]. Nonetheless, in that same year 1.4 million children are estimated to have died from vaccine preventable diseases (measles, Haemophilus influenzae type B [Hib], pertussis, tetanus, yellow fever, and poliomyelitis), a reflection of the incomplete coverage with existing vaccines that persists in many parts of the world [ 1 ]. In 2006, of the 157 WHO member states defined as "developing", only 42 (27%) had three doses of diptheria-pertussis-tetanus (DPT) coverage greater than 80% in all districts [ 2 ]. At the same time, new opportunities exist to strengthen immunization coverage in developing countries, such as increased funding through platforms such as the Global Immunization Vision and Strategy (GIVS), as well as novel ideas for integration with other health services. These recent developments have encouraged a macro-analytic approach to ensure that systems function so that children receive needed vaccines. While these new approaches are welcomed, at the micro level, immunization service delivery in health facilities needs to be strengthened.

Immunization programs need continued support with proven strategies and fresh approaches to reduce the incidence of diseases that may be prevented through the use of traditional vaccines, and to permit the effective introduction of new vaccines. There are 23 new or improved vaccines for children and adolescents in development [ 3 ]. Integrating these vaccines into routine programs will substantially increase the needed expenditure on routine immunizations. To fully take advantage of these new vaccines it is essential to identify and utilize proven strategies for improving routine immunization programs at the service delivery level. Despite the attention that global immunization has attracted in recent years in terms of the introduction of new vaccines and the strengthening of health systems, there is a clear need to ensure that program managers are aware of what strategies at the health facility level will be needed to strengthen programs. To help identify these strategies a review of gray literature and a systematic review of published literature were conducted. In this paper, we present the strategies that may be used at the community or facility level that have been shown to strengthen routine immunization programs. This review builds on two similar reviews of immunization service strategies in developing countries which were published between 2004 and 2005, one of published literature and the other of gray literature [ 4 , 5 ]. Although similar to this review, additional papers not identified by the previous reviews have been included as our approach was broader and included all papers reporting on a strategy used to strengthen routine services. For the other reviews, the existence of primary data evaluating the effectiveness or cost-effectiveness of the strategy to improve coverage was required. With this review, primary data on effectiveness of the strategy was not required for inclusion, as we wanted to identify all possible strategies since effectiveness is not always generalizable.

Although routine immunization programs and mass campaigns are complementary strategies used to increase immunization coverage, this review focused on routine immunization programs. This information will be of interest to immunization managers at national and sub-national levels, as well as those interested in increasing population coverage with other recommended health interventions.

Literature Search

We searched on-line library journal databases using 42 terms (Table 1 ) for papers published in English, French, or Spanish from 1975 through 2004, in total 11,235 papers were identified. We also searched the gray literature by requesting information from 35 websites including WHO regional databases, dissertation, theses and gray literature database websites and contacting 31 experts in the field of which 20 replied and 11 provided references. In total, close to 11,500 papers were identified, the vast majority of which were not routine immunization specific. Based on a review of titles, abstracts and executive summaries, 264 papers were collected. These papers were reviewed by one person and narrowed down to those that presented a study or project conducted to improve routine immunization programs among humans in a low- or middle-income country. Papers assessing immunization campaigns were excluded. Only 60 papers (50 published and 10 unpublished) met these criteria; these papers were then systematically reviewed.

Review Methods

The 60 papers identified through our literature search were first classified into one of three methodological groups: observational studies, studies with evaluation before and after the intervention, and studies with comparison groups. Next, each paper was rated based on a standardized assessment unique to each methodology and data were systematically abstracted. This rating was based on whether elements considered critical to the scientific quality of the study and to the reader's ability to understand adequately the intervention and its impact were present. No effort was made to validate the methods reported, for example that the sample size was correct, rather instead to confirm that the data necessary to verify methods were presented in the paper and that the methods seemed appropriate. The elements assessed included information about and appropriateness of the target population, the use of randomization in the study if appropriate, the presence of clearly defined study questions and outcomes, the identification of possible confounders, the quality of data analysis, evidence of sufficient time to evaluate the intervention, and a discussion of study limitations and how study results compared to published literature (Table 2 ). The rating ascribed to each paper that described studies with before-and-after interventions or with comparison groups represented a consensus between two reviewers while observational studies were rated by a single reviewer. The same rating was applied to all paper types, however only one reviewer was used for observational studies as many of the criteria we used in the assessment were not applicable for observational studies (e.g. methods for obtaining controls, sample size calculations, accounting for confounders, etc). Papers with a score of more than 60% (number of assessment elements reported in the paper over total elements assessed) were reported and papers with a score below this cut-off were excluded due to difficulty in interpreting the results that they presented. Furthermore papers were excluded that were found to present inconclusive results or focused on strategies to improve the overall health systems as opposed to a particular routine immunization strategy.

Only 25 papers met our criteria for inclusion in this review (Table 3 ). All of the gray literature papers were excluded; most of these papers lacked detailed information or methodology details and so received too low a score to be included. The remaining papers were grouped by the general approach used to improve immunization programs (Figure 1 ). There were numerous groupings which could have been used to organize the findings, and some papers inevitably overlap. Ultimately, we chose categories that we felt would be most beneficial and the most "user-friendly" for national and sub-national program managers to identify strategies. The four groups were: bringing immunizations closer to communities (n = 11), using information dissemination to increase demand for vaccinations (n = 3), changing practices in fixed sites (n = 4), and using innovative management practices (n = 7). In the selected papers, the most consistently reported outcome indicator was the percentage change in fully vaccinated children (FVC), although some papers used other outcomes such as percentage change in vaccination coverage for specific antigens, dropout from routine immunizations as measured by coverage for an early vaccine when compared to that of a later vaccine, or timeliness of vaccination for a specified antigen.

figure 1

Review Methods . see attached file 1.

Bringing immunizations closer to the community

The studies included in this category used non-health workers to encourage people to seek immunization services, or increased access to immunization services by bringing services to communities, and additionally in some cases by increasing demand through educating communities.

Many of these papers documented how the involvement of community members can improve immunization utilization. For example, strengthening demand for immunization services was part of the Integrated Child Development Services Program in India which began in 1975. In this program one village woman for every 1000 population was selected to provide health information to village residents, maintain lists of women and children who needed immunizations, motivate families to bring children for immunizations, assist with immunizations, and follow-up on immunization side effects, as well as to provide other community services. After more than five years of implementation, the proportion of vaccinated children was higher in the intervention group than in the control group for every antigen, ranging from a 35% difference for DPT3 vaccine to a 43% difference for Bacille Calmette-Guerin (BCG) [ 6 ].

In Bangladesh and South Africa, tools were developed to assist community workers in tracking their home visits. An observational study in Bangladesh evaluated using semi-literate and illiterate local women in an urban setting to track defaulters using a color-coded tracking system, to refer them to services and accompany mothers to immunization clinics. During the 13-month intervention (1987–1988), 87% percent of children referred by these volunteers completed the recommended immunization series and 96% of women that were referred received tetanus vaccine [ 7 ]. A similar program in South Africa evaluated giving record cards to Village Health Workers (VHWs) to record home visits over a one-year period (1988) in an intervention district. VHWs used the cards to identify children to visit, document visit frequency, and track health interventions including immunizations. Sixty-seven percent of children born during the program had completed their third dose of Oral Polio Vaccine (OPV) by eight months of age compared with 50% in the cohort of children (13 to 24 months) born before the program was implemented. However, coverage with measles vaccine by 10 months of age among children aged 13 to 24 months was higher compared to children exposed to the program [ 8 ].

In two papers the use of home visits for education and/or service delivery was evaluated. In Ghana, non-health workers conducted door-to-door visits and referred all children less than five years of age to routine immunization clinics. In addition, a health worker conducted home visits for children who failed to finish their immunization series. Over a six-month period (7/1991–2/1992), the percentage of FVC increased from 60% to 85% in the intervention group, whereas in the control group coverage increased from 61% to 67% [ 9 ]. In Mexico, trained community members were used to conduct home visits during which immunization education was provided along with needed vaccines. This intervention increased the percentage of FVC less than one year of age from 21% to 77% in five months (1994), compared with the control group where coverage increased only from 30% to 35% [ 10 ].

Other successful strategies focused on increasing access to immunization services. In Kenya, school buildings were utilized as immunization centers, with an educational component provided by schoolchildren who circulated immunization information within their communities. Furthermore, mobile teams were used to increase access. Coverage outcomes varied according to population density. In high population density areas the percentage of FVC increased from 54% to 82% and in low density areas it increased from 25% to 57% over an unspecified period. Coverage at follow-up in comparison high density areas was 69% compared to the 82% and in low population density areas 27% compared to the 57% [ 11 ]. In a district of Papua New Guinea, health post staff were trained in administering immunizations to permit vaccines to be given closer to rural communities. In this study, conducted between 1983 and 1987, measles coverage increased from 4% to 75% in the intervention district, compared with the control district where coverage increased from 5% to 58% [ 12 ]. In Nigeria, access to immunization services was improved by increasing the number of locations offering immunizations and adding mobile clinics in the evenings. The area in which this intervention was conducted saw an increase in FVC from 5% to 43% over a two-year period (1984–1986) [ 13 ].

Conflict areas are generally difficult to reach because of security concerns. Three papers evaluated strategies that provided immunizations in conflict areas of Mozambique. Strategies involved using bush planes to gain access to people, providing incentives to attract people to immunization sites, going house-to-house to motivate parents to bring their children for immunization, and working with communities to coordinate provision of services [ 14 – 16 ].

Using information dissemination to increase demand for vaccination

Information can be provided through numerous channels to either increase awareness of the benefits of immunization or to promote participation. These strategies increase demand for vaccination without changing the service delivery. Mass communication campaigns have the potential to reach large numbers of people, if access to the type of media selected is good. In the Philippines, a mass media campaign focusing on measles vaccination delivered through routine services was evaluated. An increase in the percentage of FVC from 54% in 1989 to 65% in 1990 was reported; this increase was attributed to the impact of the media campaign [ 17 ]. In Bangladesh, an increase in immunization coverage was linked to the use of inter-personal communication among mothers participating in a non-government organization (NGO) credit program that encouraged child immunization without providing additional immunization services. Increased coverage of several antigens was reported among the children of women who participated in the NGO program relative to the children of women who did not participate in the NGO program. For example, in 1995, measles coverage was 68% among children of participants compared with 59% in children of non-participants [ 18 ]. In the West Bank providing information at the local level through training community members regarding immunizations and providing resource rooms with information on immunization did not increase vaccine coverage, however the timeliness of immunizations, defined as children receiving vaccines at the appropriate age, improved (1985–1996) [ 19 ].

Changing practices in fixed sites

Improved quality of health facility practices can increase coverage through reducing dropout (children that start the vaccination series, but did not complete the series) and missed opportunities (children that were available for vaccination, but that were not vaccinated). In Ethiopia, the use of reminder stickers for parents resulted in decreasing dropout between DPT1 and DPT2 to seven percent in the intervention district compared with 13% in the comparison district during 1992 [ 20 ]. A study conducted in Sudan compared two methods to reduce missed opportunities for vaccination: moving the immunization location close to the consultation room in the health facility to provide immediate immunizations to children who had recently been seen in consultation, and having the physician write a prescription for immunizations during curative visits. Each method resulted in an increase of 32% more children being vaccinated during the intervention week than during the week prior to the intervention [ 21 ]. An urban Nigerian health center increased coverage of children fully vaccinated by one year of age by 18% in 1982 through reducing wait times by creating a quick immunization line [ 22 ]. In a Mexican children's hospital the missed opportunities for immunization were reduced by immunizing all hospitalized children who were not up-to-date with their vaccines. This led to the number of childhood immunizations delivered monthly increasing from 150 to 600 in 1991 [ 23 ].

Using innovative management practices

Reviewed papers addressed two management issues: who should manage immunization systems and how systems might be improved to provide the highest quality services. In Cambodia, increased coverage and improved equity were achieved by contracting immunization services to NGOs in selected districts (1997–2000). Although these districts had higher immunization coverage and improved equity compared to immunization programs run by the Ministry of Health, the annual per capita cost of contracting out services was almost twice that of providing services through the Ministry of Health [ 24 ].

Coverage can also be increased through better use of data and community information. In Bolivia, high-risk populations in selected communities were visited biannually from 1992 to 1994 and members of these populations assisted in identifying their priority health problems. Among these targeted populations, 78% percent of children aged 12 to 23 months were fully vaccinated in established programs compared with only eight percent in comparable populations in control communities [ 25 ]. Similarly, in Papua New Guinea health staff met to determine how best to improve services. They redefined health facility catchment areas and built a reporting system to collect accurate and meaningful data. These interventions were associated with an increased coverage of DPT2 from 64% in 1980 to 89% in 1984 [ 26 ].

Other management methods have been used to improve service quality. In Indonesia, experienced nurses in well-performing health centers peer-trained nurses in poorly-performing health centers (1993–1994). This intervention was low-cost and was associated with increases in coverage for all antigens, such as an increase of 25 percentage points in measles coverage in health centers participating in this program as compared to health centers that did not participate [ 27 ]. In Madagascar, in 2000, the use of auto-disable syringes was found to improve the availability of immunizations. Health workers were more willing to vaccinate on non-immunization days since the additional work of syringe and needle sterilization was not required. Missed opportunities for vaccination were thus reduced and coverage was increased. Use of auto-disable syringes also improved injection safety [ 28 ]. In Nicaragua, food incentives were introduced (1985) to create demand for immunization services. Mobile outreach without food incentives had 63% attendance but when food incentives were added, attendance increased to 102%. The coverage >100% was described as most likely occurring because of census errors, as mechanisms were put in place to reduce the opportunity for ineligible children to receive the food incentives. A static clinic achieved 94% attendance with food incentives [ 29 , 30 ].

A striking finding from this literature review was the paucity of well-conducted studies examining ways in which routine immunization programs in developing countries may be improved through interventions at the community or facility level. Despite an exhaustive literature search through which we identified greater than 11,000 papers, only 25 were ultimately eligible for inclusion in this review, of which only four projects were conducted in the last ten years. Furthermore, many of these 25 papers were of only moderate scientific quality. This may be in part because scientific research was not the primary purpose of the activity that many of the papers reported. Nonetheless, this situation is surprising in light of the fact that the Expanded Program on Immunization (EPI) has existed for more than a quarter of a century, and the importance and cost-effectiveness of achieving high population coverage with vaccines has been repeatedly recognized [ 31 ].

Although every paper included in this review aimed to show an improvement in immunization coverage, a wide range of indicators were used to measure success. A meta-analysis could not be conducted due to the variety of indicators reported. Some strategies were implemented in areas where baseline coverage was relatively high, thus limiting the potential increase in coverage. Other strategies were evaluated in places with low baseline coverage, and thus had the potential to result in large coverage increases. For these reasons, it is difficult to determine which strategies were most successful. Furthermore, some strategies may be more successful in certain social or health care settings than others. It is challenging to determine the generalizability of the findings, as less than half of the papers included a complete discussion of findings including topics such as comparison of findings from other similar studies.

The 25 papers identified reveal how community and facility-based strategies to strengthen routine immunization programs may result not only in increased vaccination coverage, but in other benefits. For example, projects designed to increase coverage were associated with improved timeliness of vaccination, improved knowledge regarding vaccines, improved quality, and increased equity.

Evidence from the papers suggests non-health workers can provide numerous services including education, mobilization, and tracking of target populations. Often these non-health workers are very successful because of their community knowledge, the respect they are given by the community, and the fact that they have access to community members who may not be reached by mass media such as radio or television. Community members can be used to promote specific antigens based on their expertise; for example, Traditional Birth Attendants (TBAs) may be best at increasing coverage of vaccines delivered early in life (i.e., BCG, DPT1). Home visits by non-health – worker volunteers can be very successful at motivating parents to utilize immunization services. During house visits, these volunteers can identify families not utilizing services; these families can then be followed-up by health workers.

In general, the sustainability of interventions as perceived by papers' authors was not addressed in the papers reviewed, and few programs were evaluated for enough time to determine sustainability. Given the number of interventions relying on volunteers, sustainability is of particular concern. Researchers should evaluate the residual impact of the intervention, to better understand the sustainability of the project. For example evaluating if the program resulted in change in infrastructure or practices that would continue to improve immunization coverage after the project was over. In this paper, no data have been presented on the cost-effectiveness of the various interventions discussed. These data have not been included since the two other reviews, previously mentioned [ 4 , 5 ], have reviewed and published the cost-effectiveness, as well as effectiveness, of various immunization service strategies.

Our literature review had a number of limitations. Although we attempted to conduct a thorough search for papers, those not readily available through databases, or on the web may have been missed. Furthermore the literature-gathering process was conducted through the use of a computer from an office; a more complete review may have been achieved through visiting locations to access literature in person [ 5 ]. The methods used to assess the quality of papers and thus determine their eligibility for inclusion in this review may have been biased toward published papers as many gray literature papers did not discuss the study methodology used in enough detail to allow it to be assessed. As such, no gray literature papers were included in this review. Furthermore, the majority of papers, published and unpublished, reported positive results, thus excluding opportunities to learn from unsuccessful interventions.

Additional Research

Although a wide range community and facility-based strategies to strengthen immunization programs were covered in the papers reviewed, there remain many areas for further research. Some of the topics we felt were lacking from the papers reviewed may not be specific to immunization and as such were not identified through our search strategy. For example, we found few studies related to health facility management, facility staffing, or community financing of health facilities. However, it seems likely that such studies exist. Topic areas which we identified as important and were likely not missed due to our search strategy were highlighted as needing further research (Table 4 ). Attention to these areas will be important as immunization services are integrated with other health interventions and new vaccines are introduced. Studies should strive to use rigorous scientific methods, for example by calculating minimum sample sizes based on clearly articulated assumptions, assessing confounders, using control areas when appropriate, using randomization to select intervention areas, and using statistical tests as indicated in data analysis. Furthermore, the results of these studies should be widely disseminated. In addition to peer-reviewed publication, studies can be disseminated through Interagency Coordinating Committees, newsletters, WHO regional bulletins, and press releases.

This paper summarizes the literature in terms of what is reported to have been successful in improving routine immunization programs through community and facility-based interventions over the past thirty years. Information obtained from well conducted scientific studies will be crucial to assist program managers to implement strategies to achieve high coverage. These activities coupled with the attention being given to the health systems approached advocated by the GAVI Alliance and other donors will be crucial to ensure that all levels of the immunization system function effectively. The potential health improvements from vaccines will continue to increase as new vaccines become available and as the price of these vaccines becomes more affordable. Nonetheless, as the true impact of vaccination depends heavily on the ability of immunization programs to reach every targeted individual without clear local delivery strategies countries will not be in a position to take full advantage of the potential for reduction of disease. Data from these papers confirms the need for well managed immunization programs providing high quality accessible services in conjunction with community demand. These key elements are also the basis of the WHO Reaching Every District (RED) strategy [ 32 ], a broad, all-encompassing approach covering five areas of immunization programs. Findings from this paper also indicate that all elements of an immunization program need to be addressed. For example, easily-accessed, high-quality services will not be utilized if community demand is lacking. With this in mind, it is critical to build upon lessons from the past and to continue to conduct research on how high vaccination coverage can be achieved in every community.

World Health Organization [Internet]: Immunization surveillance, assessment and monitoring: vaccine preventable diseases. [2002 data, cited 2004]., [ http://www.who.int/immunization_monitoring/diseases/en/ ]

World Health Organization [Internet]: Immunization surveillance, assessment and monitoring: vaccine preventable diseases. [2005 data, cited Jan 2007]., [ http://www.who.int/immunization_monitoring/data/SlidesGlobalImmunization.pdf ]

Infectious Diseases in Children [Internet]: Vaccines for Children in the Pipeline. [2006 data, cited Jan 2007]., [ http://www.idinchildren.com/200607/vaccines.pdf ]

Pegurri E, Fox-Rushby JA, Walker D: Effects, costs and cost-effectiveness of interventions to expand coverage of immunisation services in developing countries: A systematic review of the published literature. Vaccine. 2005, 23 (13): 1624-1635. 10.1016/j.vaccine.2004.02.029.

Article   PubMed   Google Scholar  

Batt K, Fox-Rushby J, Castillo-Riquelme M: The costs, effects and cost-effectiveness of strategies to increase coverage of routine immunizations in low- and middle- income countries: systematic review of the grey literature. Bulletin of the World Health Organization. 2004, 82 (9): 689-696.

PubMed   PubMed Central   Google Scholar  

Tandon BN, Gandhi N, the Integrated Child Development Consultants: Immunization coverage in India for areas served by the Integrated Child-Development Services programme. Bulletin of the World Health Organization. 1992, 70 (4): 461-465.

CAS   PubMed   PubMed Central   Google Scholar  

Hughart N, Silimperi DR, Khatun J, Stanton B: A new EPI strategy to reach high risk urban children in Bangladesh: urban volunteers. Trop Geogr Med. 1992, 44 (1–2): 142-148.

CAS   PubMed   Google Scholar  

Kuhn L, Zwarenstein M: Evaluation of a village health worker programme: the use of village health worker retained records. International Journal of Epidemiology. 1990, 19 (3): 685-692. 10.1093/ije/19.3.685.

Article   CAS   PubMed   Google Scholar  

Brugha RF, Kevany JP: Maximizing immunization coverage through home visits: a controlled trial in an urban area of Ghana. Bulletin of the World Health Organization. 1996, 74 (5): 517-524.

Calderón-Ortiz R, Mejía-Mejía J: Estrategia de contratación permanente dentro del Programa de Vacunación Universal. Salud Pública de México. 1996, 38 (4): 243-248.

PubMed   Google Scholar  

Anonymous: Expanded Programme on Immunization: Study of feasibility, coverage and cost of maintenance immunization for children by district mobile teams in Kenya. Weekly Epidemiological Record. 1977, 52 (24): 197-204.

Google Scholar  

Alto WA, Alk S, Pinau D, Polume H: Improving immunization coverage, a comparison between traditional MCH teams and MCH teams plus Aid Post Orderlies. P N G Med J. 1989, 32 (2): 97-100.

Oruamabo RS, Okoji GO: Immunisation status of children in Port Harcourt before and after commencing the Expanded Programme on Immunisation. Public Health. 1987, 101 (6): 447-452. 10.1016/S0033-3506(87)80007-0.

Coninx R, Dupuy C, Hermann C, Ribeiro GC, Margot M, Lucic K: Vaccination of the civilian population in a country at war: it can be done; it can also be evaluated. The ICRC experience in Mozambique. Journal of Tropical Pediatrics. 1998, 44 (3): 186-188. 10.1093/tropej/44.3.186.

Cutts FT, Kortbeek S, Malalane R, Penicele P, Gingell K: Developing appropriate strategies for EPI: A case study from Mozambique. Health Policy & Planning. 1988, 3 (4): 291-301. 10.1093/heapol/3.4.291.

Article   Google Scholar  

Cutts FT, Phillips M, Kortbeek S, Soares A: Door-to-door canvassing for immunization program acceleration in Mozambique: Achievements and costs. International Journal of Health Services. 1990, 20 (4): 717-725.

Zimicki S, Hornik RC, Verzosa CC, Hernandez JR, deGuzman E, Dayrit M, Fausto A, Lee MB, Abad M: Improving vaccination coverage in urban areas through a health communication campaign: the 1990 Philippine experience. Bulletin of the World Health Organization. 1994, 72 (3): 409-422.

Amin R, Li YP: NGO-promoted women's credit program, immunization coverage, and child mortality in rural Bangladesh. Women & Health. 1997, 25 (1): 71-87. 10.1300/J013v25n01_05.

Article   CAS   Google Scholar  

Tulchinsky T, Al Zeer AM, Abu Mounshar J, Subeih T, Schoenbaum M, Roth M, Gamulka B, Abenueze M, Acker C: A successful, preventive-oriented village health worker program in Hebron, the West Bank, 1985–1996. J Public Health Manag Pract. 1997, 3 (4): 57-67.

Berhane Y, Pickering J: Are reminder stickers effective in reducing immunization dropout rates in Addis Ababa, Ethiopia?. J Trop Med Hyg. 1993, 96 (3): 139-145.

Loevinsohn BP, Gareaballah E: Missed opportunities for immunization during visits for curative care: a randomized cross-over trial in Sudan. Bulletin of the World Health Organization. 1992, 70 (3): 335-339.

Ekunwe EO: Expanding immunization coverage through improved clinic procedures. World Health Forum. 1984, 5 (4): 361-363.

Avila-Figueroa C, Navarrete-Navarro S, Ramírez-Galván L, Baltazar-López A, López-Serrano M, Santos-Preciado JI: Inmunizaciones en niños hospitalizados y de consulta externa: reducción de las oportunidades perdidas de vacunación. Bol Med Hospital Infant Mex. 1992, 49 (5): 271-274.

CAS   Google Scholar  

Schwartz JB, Bhushan I: Improving immunization equity through a public-private partnership in Cambodia. Bulletin of the World Health Organization. 2004, 82 (9): 661-667.

Perry H, Robison N, Chavez D, Taja O, Hilari C, Shanklin D, Wyon J: The census-based, impact-oriented approach: its effectiveness in promoting child health in Bolivia. Health Policy and Planning. 1998, 13 (2): 140-151. 10.1093/heapol/13.2.140.

van Zwanenberg TD, Hull C: Improving immunisation: coverage in a province in Papua New Guinea. Br Med J (Clin Res Ed). 1988, 296 (6637): 1654-1656.

Robinson JS, Burkhalter BR, Rasmussen B, Sugiono R: Low-cost on-the-job peer training improved immunization coverage in Indonesia. Bulletin of the World Health Organization. 2001, 79 (2): 150-158.

Drain PK, Ralaivao JS, Rakotonandrasana A, Carnell MA: Introducing auto-disable syringes to the national immunization programme in Madagascar. Bulletin of the World Health Organization. 2003, 81 (8): 553-560.

Loevinsohn BP, Loevinsohn ME: Improvement in coverage of primary health care in a developing country through use of food incentives. The Lancet. 1986, 1 (8493): 1314-1316. 10.1016/S0140-6736(86)91231-6.

Loevinsohn BP, Loevinsohn ME: Well child clinics and mass vaccination campaigns: an evaluation of strategies for improving the coverage of primary health care in a developing country. American Journal of Public Health. 1987, 77 (11): 1407-1411.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Hadler SC, Cochi SL, Bilous J, Cutts FT: Chapter 55: Vaccination Programs in Developing Countries. Vaccines. Edited by: Plotkin SA, Orenstein WA. 2004, Philadelphia, Pennsylvania, USA: Elsevier Inc., 1407-1441. fourth

World Health Organization [Internet]: The RED Strategy. [cited 2006]., [ http://www.who.int/immunization_delivery/systems_policy/red/en/ ]

Pre-publication history

The pre-publication history for this paper can be accessed here: http://www.biomedcentral.com/1472-6963/8/134/prepub

Download references

Acknowledgements

The authors are grateful to Karen Wilkins, Elizabeth Luman, Steve Hadler, Philip Brachman, and Aaron Wallace.

Author Note: The findings and conclusions in this report are those of the author(s) and do not necessarily represent the views of the Centers for Disease Control and Prevention.

Author information

Authors and affiliations.

Global Immunization Division, Centers for Disease Control and Prevention, 1600 Clifton Road MS-E05, Atlanta, GA, 30333, USA

Tove K Ryman, Vance Dietz & K Lisa Cairns

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Tove K Ryman .

Additional information

Competing interests.

The authors declare that they have no competing interests.

Authors' contributions

TKR: Made substantial contributions to conception and design, acquisition of data, analysis and interpretation of data. Has also been involved in drafting the manuscript, revised it critically for important intellectual content and given final approval for the version to be published. VD: Made substantial contributions to conception and design, acquisition of data, analysis and interpretation of data. Has also been involved in drafting the manuscript, revised it critically for important intellectual content and given final approval for the version to be published. KLC: Has been involved in drafting the manuscript, revised it critically for important intellectual content and given final approval for the version to be published.

Tove K Ryman, Vance Dietz and K Lisa Cairns contributed equally to this work.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions.

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article.

Ryman, T.K., Dietz, V. & Cairns, K.L. Too little but not too late: Results of a literature review to improve routine immunization programs in developing countries. BMC Health Serv Res 8 , 134 (2008). https://doi.org/10.1186/1472-6963-8-134

Download citation

Received : 17 August 2007

Accepted : 21 June 2008

Published : 21 June 2008

DOI : https://doi.org/10.1186/1472-6963-8-134

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Immunization Program
  • Immunization Coverage
  • Routine Immunization
  • Immunization Service
  • Intervention District

BMC Health Services Research

ISSN: 1472-6963

literature review on immunization

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Front Immunol

COVID-19 Vaccination and the Rate of Immune and Autoimmune Adverse Events Following Immunization: Insights From a Narrative Literature Review

Naim mahroum.

1 International School of Medicine, Istanbul Medipol University, Istanbul, Turkey

2 Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel

3 St. George School of Medicine, University of London, London, United Kingdom

Aviran Ohayon

Ravend seida, abdulkarim alwani, mahmoud alrais, magdi zoubi, nicola luigi bragazzi.

4 Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, Canada

Despite their proven efficacy and huge contribution to the health of humankind, vaccines continue to be a source of concern for some individuals around the world. Vaccinations against COVID-19 increased the number of distressed people and intensified their distrust, particularly as the pandemic was still emerging and the populations were encouraged to be vaccinated under various slogans like “back to normal life” and “stop coronavirus”, goals which are still to be achieved. As fear of vaccination-related adverse events following immunization (AEFIs) is the main reason for vaccine hesitancy, we reviewed immune and autoimmune AEFIs in particular, though very rare, as the most worrisome aspect of the vaccines. Among others, autoimmune AEFIs of the most commonly administered COVID-19 vaccines include neurological ones such as Guillain-Barre syndrome, transverse myelitis, and Bell’s palsy, as well as myocarditis. In addition, the newly introduced notion related to COVID-19 vaccines, “vaccine-induced immune thrombotic thrombocytopenia/vaccine-induced prothrombotic immune thrombotic thrombocytopenia” (VITT/VIPITT)”, is of importance as well. Overviewing recent medical literature while focusing on the major immune and autoimmune AEFIs, demonstrating their rate of occurrence, presenting the cases reported, and their link to the specific type of COVID-19 vaccines represented the main aim of our work. In this narrative review, we illustrate the different vaccine types in current use, their associated immune and autoimmune AEFIs, with a focus on the 3 main COVID-19 vaccines (BNT162b2, mRNA-1273, and ChAdOx1). While the rate of AEFIs is extremely low, addressing the issue in this manner, in our opinion, is the best strategy for coping with vaccine hesitancy.

Introduction

Considered one of the fastest processes of manufacturing vaccines ever, “Coronavirus Disease 2019” (COVID-19) vaccines were discovered, studied, and produced in terms of months since the declaration of COVID-19 as a pandemic by the World Health Organization (WHO) ( 1 ). While the burden of the outbreak alongside the support of the governments were the main motivations for the accelerated vaccine production, the process itself aroused concerns regarding the safety of the COVID-19 vaccines and contributed to mistrust in health authorities as well as in the vaccines themselves ( 2 , 3 ). Interestingly, distrust in COVID-19 vaccines was reported even before the vaccines were available ( 4 ). It is of great importance to mention that vaccines are highly effective in reducing the burden of infectious diseases as the history clearly shows ( 5 , 6 ), and the COVID-19 pandemic is likely to follow a similar trend after the roll-out of the COVID-19 vaccination has been fully implemented, globally. However, the fear of the lay public for adverse events following immunization (AEFIs), especially those of severe and long-term features, may constitute the main obstacle in successfully fighting against COVID-19 vaccine hesitancy ( 7 , 8 ). This is especially true for autoimmune and immune-mediated AEFIs such as idiopathic thrombocytopenia (ITP), or immune thrombocytopenia, Guillain-Barré syndrome (GBS), and transverse myelitis (TM), among others. Therefore, it is crucial to address the mounting concern by reviewing the past, present, and possible future AEFIs of vaccines in general and COVID-19 vaccines in particular, especially as millions were already vaccinated around the world, and more data has accumulated regarding the safety profile of the COVID-19 vaccines ( 9 ). Addressing AEFIs of autoimmune nature covers, to a great extent, the concerns generated by the public explicitly considering vaccine booster programs endorsed by the health authorities. The importance of facing and addressing public concerns in dealing with vaccine hesitancy cannot be overemphasized ( 10 ).

We hereby present a detailed, narrative literature review covering the various types of vaccines, their immune-mediated AEFIs, and we focus on the autoimmune AEFIs described in correlation with the main COVID-19 vaccines in use worldwide, that is to say, those most administered (approximately 95% of those delivered). The rarity of the AEFIs among vaccinated people, and the risk of severe complications that COVID-19 carries, are the key factor for fighting against vaccine hesitancy.

Types of Vaccines, Adverse Events Following Immunization, and COVID-19 Related Vaccines

Vaccines that were approved or in use against COVID-19 can be classified as follows:

Inactivated Vaccines

Inactivated or killed vaccines contain a pathogen that has been inactivated after being grown in a culture. The first utilized was the cholera vaccine introduced in 1896 ( 11 ). There are two subtypes of inactivated vaccines: the traditional whole-cell vaccine and the acellular vaccine. The latter is a more modern version and contains only 3-4 antigens rather than the hundreds of microbial antigens present in the whole-cell vaccine. This antigen reduction produces a more specific immune response, thereby reducing potential AEFIs ( 12 ), such as febrile responses ( 13 ). While traditional whole virion-inactivated influenza vaccines and whole cell pertussis vaccines commonly cause febrile responses within 24 hours of immunization, the split virion influenza vaccine and the acellular pertussis vaccine showed a decreased rate of febrile illness ( 14 , 15 ). Concerning inactivated vaccines against COVID-19, the vaccine developed by the Beijing Institute of Biological Products, known as BBIBP-CorV, and referred to as Sinopharm, is an inactivated vaccine. In a randomized, double-blind, dose-escalation, controlled phases I and II trial of the BBIBP-CorV vaccine, an acceptable safety profile and a robust humoral response to the coronavirus were reported ( 16 ). The study was conducted among 18-80 years old, healthy individuals negative for serum-specific IgM or IgG antibodies against both N and S proteins of the virus. The inactivated BBIBP-CorV vaccine was immunogenic and elicited strong humoral responses, with a 100% seroconversion rate in all groups. However, lower seroconversion rates were found in the group of individuals aged 60 and older, probably due to the atrophy/ageing of the immune system ( 17 ). The degree of local and systemic AEFIs was generally mild and occurred most after the first dose of vaccination ( 18 ).

Viral Vector-Based Vaccines

Viral vectors were first introduced in relation to gene and cancer therapy but, since then, have been adapted to vaccine development ( 19 ). The utility of vector-based vaccines is determined by the capacity of viruses to infect cells ( 20 ). High efficiency gene transduction, extremely selective gene delivery to target cells, generation of powerful immunological responses, and improved cellular immunity are the advantages of viral vector-based vaccines ( 20 ). Multiple viruses ranging from very complex large DNA viruses such as poxviruses, to simple RNA viruses such as parainfluenza virus, have been deployed as viral vectors ( 21 ). Viral vector vaccines facilitate intracellular antigen expression and trigger a highly cytotoxic T-lymphocyte response ( 22 ). A significant emphasis was made on COVID-19 vaccines based on viral vectors like the AstraZeneca, Janssen/Johnson & Johnson, and CanSino vaccines ( 19 ), which are based on adenoviruses ( 23 ). In addition to the vaccines listed above, Sputnik V, also known as Gam-COVID-Vac, is a COVID-19 adenovirus viral vector vaccine ( 24 ). Injection site pain is considered the most common local AEFI of viral vector vaccines, together with headache, fatigue, muscle pain, malaise, chills, and joint pain ( 25 ).

mRNA Vaccines

Because of their high potency, ability to evolve quickly, and potential for low-cost manufacturing and safe delivery, mRNA vaccines were found to be a promising alternative to traditional vaccine techniques. In 1990, the first effective application of in vitro transcribed (IVT) mRNA in animals was described, when reporter gene mRNAs were injected into mice and protein synthesis was detected ( 26 ). However, this finding did not lead to an increase in the mRNA vaccine investment, due to concerns regarding mRNA instability, inefficient in vivo delivery, and high innate immunogenicity. Subsequently, over the past decade, the use of mRNA vaccines has been gradually implemented due to their low potential risk of infection and insertional mutagenesis ( 27 ). Actually, the concept behind the mRNA vaccine is to deliver the mRNA of the pathogen into the human body in order to trigger an immune response and produce antibodies against the pathogen. There are two types of mRNA vaccines available for prevention of infectious diseases: self-amplifying or replicon RNA vaccines (SAM) and non-replicating mRNA vaccines. SAM vaccines have the benefit of producing their own adjuvants in the form of dsRNA structures, replication intermediates, and other motifs, which may explain their high potency. Directly injectable, non-replicating mRNA vaccines are promising vaccination products due to their convenient way of administration and low cost, especially in resource-constrained environments ( 28 ). Two of the most widely distributed COVID-19 vaccines, namely the BNT162b2 (BioNTech/Pfizer) and the mRNA-1273 (Moderna), are mRNA-based vaccines. The BNT162b2 vaccine, for instance, proved to be around 95% efficient in preventing the disease, with a relatively high safety profile ( 29 ). However, mild and short-term AEFIs were reported such as pain at the injection site, fatigue, and headache. The incidence of serious AEFIs was low.

Roll-out of COVID-19 Vaccines

As herd immunity is achieved once a large portion of the population is immune against an infectious agent preventing it from widely spreading, it is a must in overcoming the COVID-19 pandemic ( 30 ). In addition, herd immunity is also necessary to protect individuals who are unable to get vaccinated such as infants, children under the recommended age, and immunocompromised individuals ( 31 ). In turn, people become immune against a specific infection in two ways, either through acquiring the infection naturally from a pathogen, or passively by vaccination. While COVID-19 can be severe and fatal, especially in people at risk such as the elderly, those with chronic cardiovascular and respiratory disease, among others ( 32 ), vaccinations is paramount in terms of preventing disease acquisition as well as its spread. The US Food and Drug Administration (FDA) authorized the first vaccine for COVID-19 on December 11 th , 2020 ( 33 ). More specifically, the first vaccine authorized was BNT162b2 (BioNTech/Pfizer COVID-19 vaccine) under the Emergency Use Authorizations (EUA). The United Kingdom (UK) Medicines and Healthcare products Regulatory Agency (MHRA) had already authorized the use of the same vaccine, some days before, on December 2 nd , 2020 ( 34 ). Subsequently, the European Union (EU) commission authorized the vaccine on December 21 st , 2020 ( 35 ). Shortly afterward, the mRNA-1273 COVID-19 vaccine (Moderna) was authorized ( 36 ). Later on, the ChAdOx1 vaccine (AstraZeneca) was authorized as well by the UK MHRA ( 37 ). The three mentioned vaccines were the most commonly used vaccines across the globe. By the end of November, 2021, approximately 55% of the world population has received at least one dose of a COVID-19 vaccine, 7.9 billion doses have been given globally, and around 27 million new doses are administered daily ( 38 ). Asia had the most doses administered at 5.09 billion doses, followed by Europe with 942 million doses, North America with 739.54 million doses, South America with 581.31 million doses, Africa with 235.45 million doses, and finally Oceania with 49.67 million doses.

Autoimmune Adverse Events Following Immunization and Their Correlation With Vaccines in General and COVID-19 Vaccines in Particular

Autoimmune neurological adverse events following immunization, guillain-barre’ syndrome (gbs).

GBS is an autoimmune disorder characterized by an immune-mediated nerve damage generally triggered by an infectious agent leading to cross-reactive antibodies attacking axonal antigens and resulting in demyelinating polyneuropathy ( 39 ). Despite its rarity, GBS is considered as the most common cause of acute flaccid paralysis worldwide ( 40 ), with about 100,000 people developing the disorder every year ( 41 ). The disease is manifested by ascending and symmetrical flaccid paralysis leading to paresthesia, autonomic dysfunction, and respiratory muscle paralysis ( 42 ). In some instances, cranial nerve involvement is also seen, causing facial diplegia ( 43 ). The complications of GBS can be fatal, including respiratory and cardiac failure ( 44 ). The mortality rate of GBS varies widely from 1-18% ( 45 , 46 ). Numerous reports have suggested a possible relationship between GBS and vaccines. However, solid evidence was not established ( 47 , 48 ). During the COVID-19 pandemic, various reports addressed the correlation between COVID-19 and GBS; nevertheless, a concrete causal relationship is yet to be established ( 40 ).

BNT162b2 (BioNTech/Pfizer) and GBS

Several case reports were published regarding GBS following the BNT162b2 vaccine. A 82-years-old female presented two weeks following her first dose ( 49 ), and a 67-year-old male seven days after the first shot ( 50 ). In addition, a 71-year-old male with Miller-Fisher syndrome (MFS), a rare variant of GBS, presented 18 days following his first dose of BNT162b2 vaccine ( 22 ). GBS was also documented following the second dose of the vaccine. For instance, a 25-year-old female developed a clinical picture of GBS few days following her second dose of the vaccine, and a 73-year-old male twenty days following the second dose of the vaccine ( 51 ). Furthermore, Ben David and colleagues ( 52 ) conducted a retrospective cohort study aimed to assess the safety of mRNA-based COVID-19 vaccine in previously diagnosed cases of GBS between 2000-2020. Based on a database from a health organization serving more than 2.5 million members, the authors found that out of 702 members who had a diagnosis of GBS, 579 received at least one vaccine dose, and only one patient presented with a relapse of GBS several days following the second dose, which represents a minimal risk. In addition, Shasha et al. ( 53 ) investigated AEFIs profile following BNT162b2 vaccine in a sample size of over 400,000 individuals, and found that only one individual in the vaccinated group had GBS versus none in the control group. Similarly, seven cases of GBS were reported following the first dose of BNT162b2 mRNA vaccine among approximately 4 million recipients (incidence of 0.18/100,000), while no cases recorded following the second dose. The study was conducted over a period of 30 days following vaccination concluding that among recipients of the BNT162b2 mRNA vaccine, GBS may occur at the expected community-based rate ( 54 ).

mRNA-1273 (Moderna) and GBS

To the best of our knowledge, only two case reports have been published regarding GBS following Moderna, mRNA-1273 COVID-19 vaccine. Both of the cases developed after the second dose. In the first case ( 55 ), the symptoms appeared two days after vaccination presented by an axonal variant of GBS. In turn, in the second case ( 56 ), the symptoms appeared 6 weeks following the vaccination, while the electrophysiological test identified a mixed axonal and demyelinating type of GBS. Furthermore, among 16 cases of acute-onset polyradiculoneuropathy that presented within 4 weeks after first dose of SARS-CoV2 vaccines, only one person received the mRNA-1273 vaccine, whereas 14 received the ChAdOx1 vaccine ( 57 ).

ChAdOx1 (AstraZeneca) and GBS

During July 2021, the European Medicines agency (EMA) recommended that GBS should be added as a warning sign on the ChAdOx1 vaccine product information, despite being unable to decisively conclude about a causal association ( 58 ). This action was in part due to multiple reports of GBS cases occurring within a month following the first dose of the vaccine ( 59 – 67 ). The series of cases caused a major concern among leading experts. In fact, the GBS cases reported after the ChAdOx1 vaccine often presented as facial diplegia and paresthesia, a rather rare manifestation of the condition, with clinical improvement after corticosteroid and intravenous immune globulins (IVIG) therapy. The cases had no known exposure to the coronavirus and tested negative upon admission. However, as a recent study found that the COVID-19 infection was unlikely to cause GBS ( 68 ), the significance is debatable. Other baseline characteristics, such as age, gender and associated morbidity differed between cases, and scholars are therefore unable to make a clear-cut connection between them. The incidence of GBS was estimated to be approximately 0.89 to 1.89 cases per 100,000 person-years ( 69 ). A recent report issued by the UK Health Security Agency concluded that the risk of developing GBS after the first dose of ChAdOx1vaccine adds 5.6 extra cases of GBS per million doses, while having no association with respect to the second dose ( 70 ). As the condition has also been linked to the Ad26.COV2.S (Janssen/Johnson & Johnson) COVID-19 vaccine ( 71 ), another adenoviral vector vaccination, further investigations into the pathogenesis are required as large studies about GBS rate are lacking, and conclusions should not be drawn without further analysis.

Transverse Myelitis

TM is an immune-mediated acute or a subacute inflammatory disease of the spinal cord accompanied with motor, sensory, and autonomic symptoms ( 72 ). The clinical presentation of TM varies depending on the level of the spinal cord involved as the disease is manifested below the affected segment. Patients presenting with TM can be paraplegic, most having urinary bladder function disorders and paresthesia ( 73 ). The exact etiology of TM has not been established yet. Nevertheless, different types of vaccines were formerly linked to the appearance of TM including hepatitis B vaccine, MMRV and others ( 74 ). Most of the cases documented occurred between several days to several months from vaccination however, longer durations were also recorded.

BNT162b2 (BioNTech/Pfizer) and Transverse Myelitis

Actually, little evidence with no obvious association exists regarding the appearance of TM after the administration of BNT162b2 COVID-19 vaccine. Case reports include a 75-year-old Japanese patient who presented with TM 3 days following the first dose of the BNT162b2 vaccine ( 75 ). The authors could not elucidate a clear association between the vaccine and the clinical presentation and concluded that more epidemiological studies are needed. Furthermore, among more than 700,000 individuals who received the first dose of the BNT162b2 vaccine in Mexico, only 2 cases of TM were documented, equal to a rate of 0.28 per 100,000 cases ( 76 ).

mRNA-1273 (Moderna) and Transverse Myelitis

Concerning the mRNA-1273 COVID-19 vaccine, case reports of ADEM ( 77 ), neuromyelitis optica ( 78 ), and acute TM ( 79 ) were reported. Ismail et al. ( 80 ) reviewed central nervous system (CNS) demyelination disorders among recipients of various COVID-19 vaccinations. A total of 32 cases were registered. Among the cases, higher rates of women (68.8%) than men and a median age of 44 years were noticed. Moreover, most of the cases (71.8%) occurred following the first dose whereas more than a half of the cases had a previous history of immune-mediated diseases (53.1%). As for TM in particular, 6 cases occurred after receiving the mRNA-1273 vaccine. According to the same study, the other vaccines possibly correlating with TM were as follows: 11 cases following BNT162b2 vaccine, 8 after ChAdOx1 vaccine, 5 after Sinovac/Sinopharm vaccines, and one after each of the Sputnik and the Janssen/Johnson & Johnson vaccines.

ChAdOx1 (AstraZeneca) and Transverse Myelitis

During the phase 3 clinical trial of the ChAdOx1 vaccine, 3 participants were diagnosed with TM ( 81 ). As a result, the trial was temporarily paused enabling further investigations. Two of the cases were determined to be unrelated to the vaccine, the first one had pre-existing, undiagnosed multiple sclerosis; and the other was in fact in the control group with his symptoms appearing over 68 days post vaccination ( 82 ). As experts concluded the phenomenon was unlikely to be related to the vaccination the trial resumed however, the incidents raised global concerns regarding the safety profile of the ChAdOx1 vaccine. The longitudinally extensive TM (LETM) is a rare subtype of TM, in which the damage extends over 3 or more vertebrae ( 83 ). Reports regarding the LETM subtype started to emerge following the worldwide distribution of the COVID-19 vaccines ( 84 – 88 ). All cases occurred in patients under 60 years of age who presented with neurological symptoms starting within 3 weeks of receiving the first dose of the ChAdOx1 vaccine. As the COVID-19 infection was previously implicated in inducing acute TM ( 89 ), all reported cases tested negative on standard PCR testing. Furthermore, all cases completely recovered after treatment with either high dose corticosteroids or plasma exchange and were subsequently discharged.

Bell’s Palsy

Bell’s palsy, or facial nerve palsy, is the partial (paresis) or total (paralysis) loss of function of the facial nerve ( 90 ). The etiology of Bell’s palsy is most commonly idiopathic but may occur due to several factors such as viral infections (herpes viruses), ischemia, inflammatory and immune-mediated diseases ( 91 ). Bell’s palsy is divided into central facial palsy and peripheral facial palsy ( 92 ). Central palsy is characterized by contralateral sensory disturbances, as well as dry mouth. In turn, peripheral palsy is characterized by ipsilateral paralysis of the eyelid and forehead muscles.

In a case-control study from Switzerland which aimed to assess the correlation between the inactivated intranasal influenza vaccines and Bell’s palsy ( 93 ), a significantly increased risk of Bell’s palsy was found among vaccinated people. The risk of developing Bell’s palsy was 19 times higher than the control group. As a result, the vaccine was withdrawn from clinical use.

BNT162b2 (BioNTech/Pfizer) and Bell’s Palsy

The safety database of the BNT162b2 vaccine revealed a slight increase in the cases of Bell’s palsy in vaccinated individuals ( 94 ). There were 4 cases of Bell’s palsy in the vaccine group compared to none in the placebo group. As the rate was as expected in the general population, no causal relationship was established. Reviewing the reported safety data concluded that mRNA-based vaccines might be associated with Bell’s palsy ( 95 ). Accordingly, several case reports highlighted a similar association, including a healthy 37-year-old male who had Bell’s palsy several days following the first BNT162b2 vaccine dose ( 96 ). In addition, different population-based studies which investigated the adverse effects of the BNT162b2 vaccine reported an increase in the frequency of Bell’s palsy as well. For instance, in a nationwide study from Israel which analyzed more than 800 thousand people, the incidence of Bell’s palsy was higher in the vaccinated group compared to the control group, but without significant results ( 97 ). Similarly, Shibli and colleagues ( 98 ) researched a database of Bell’s palsy cases occurring within 21-days after the first dose and 30-days after the second dose of the vaccine in comparison to the expected cases based on a database from 2019. The authors found a slightly increased incidence of Bell’s palsy following the first dose, mainly among females aged 65 and older, with an estimated attributable risk of 4.46 per 100,000 vaccinated individuals. The study suggested an association between the vaccine and an increased risk of Bell’s palsy but with a small impact on public health. In contrast, Shasha et al. ( 53 ) reported no association between COVID-19 vaccines, including BNT162b2, and Bell’s palsy among more than 400 thousand vaccinated people compared to the control group.

mRNA-1273 (Moderna) and Bell’s Palsy

Three cases of Bell’s palsy were reported following vaccination with mRNA-1273 vaccine ( 99 – 101 ). The symptoms appeared 12 hours to 2 days after the vaccine was administered. Two cases occurred after the first dose, while one appeared following the second dose. In one patient, a prior episode of Bell’s palsy was recorded whereas the other 2 persons were healthy young people aged 35-36-year-old. Moreover, Sato et al. ( 102 ) showed that the rate of Bell’s palsy after both types of the mRNA COVID-19 vaccines (BNT162b2 and mRNA-1273) are lower or equivalent to the rate of Bell’s palsy after influenza vaccines. Additionally, a study from Singapore including 1.4 million subjects who received COVID-19 vaccination, 86.7% by BNT162b2 vaccine and 13.3% by mRNA-1273 vaccine, 11 patients were referred to hospital with Bell’s palsy and 27 patients had cranial mononeuropathy ( 103 ). According to data from the WHO pharmacovigilance database, no close association between BNT162b2 and mRNA-1273 COVID-19 vaccines and facial paralysis could be found ( 104 ).

ChAdOx1 (AstraZeneca) and Bell’s Palsy

While Bell’s palsy is a lower motor neuron disease manifesting as a unilateral facial paralysis ( 105 ), GBS may also present with facial paralysis, and most cases linked to the ChAdOx1 vaccine presented in this form ( 59 – 67 ). Therefore, it is difficult to distinguish between the two conditions. One study attempted to examine the risk of co-occurrence of Bell’s palsy and GBS following COVID-19 vaccinations and found an increased risk of co-occurrence following the ChAdOx1 vaccination ( 106 ). However, as previously mentioned, the study could not distinguish between the two conditions. Furthermore, a review of published literature failed to identify case reports of isolated Bell’s palsy linked to the ChAdOx1 vaccination. Therefore, it is impossible to draw conclusions of a possible link.

Encephalitis

Postvaccinal encephalitis was described in regard to COVID-19 vaccines. In a case series of three patients who presented with symptoms suspected of encephalitis in a range of 7-11 days of receiving the ChAdOx1 vaccine was previously documented ( 107 ). The symptoms included gait disturbance, aphasia, headaches, and seizures. As the criteria for autoimmune encephalitis was fulfilled in the three cases, treatment with systemic corticosteroids led to clinical improvement. Other causes of encephalitis including infectious agents were ruled out. All cases were mild and resolved without sequelae. Due to its rarity, the authors highlighted that the benefits of the vaccine outweigh the risks. It is noteworthy to mention hereby, that herpes simplex encephalitis was reported following ChAdOx1 vaccine ( 108 ) however, this can be regarded as an infection-related AEFI rather than autoimmune induced.

In addition, a case report of a Japanese lady admitted who developed diplopia the next day following the first dose of the BNT162b2 vaccine administration was described in the literature ( 109 ). The symptoms aggravated after the second dose of the vaccine and the patients was finally diagnosed with encephalitis based on brain MRI findings. The patient responded well and totally recovered after treatment with steroids was initiated. The authors could not prove any causal relationship in their case.

In regard to the mRNA-1273 COVID vaccine, a case report of a patient diagnosed with acute encephalitis, myoclonus and Sweet syndrome was described after receiving the first dose of the mRNA-1273 vaccine ( 110 ). The symptoms resolved following glucocorticoids treatment.

Myocarditis

Myocarditis is defined as the presence of inflammatory cellular infiltrate in the myocardium alongside tissue necrosis, which is not caused by coronary heart disease, and diagnosed by a combination of histological, immunological and immunohistochemical criteria ( 111 ). Based on etiologic factors, myocarditis can be divided into 2 subgroups: infectious, due to bacterial, viral, fungal, or parasitic infections; or non-infectious causes like autoimmunity, drugs, or vaccines ( 112 ). However, viral infections are seemingly the most common cause of myocarditis ( 113 ). Myocarditis can present with a range of symptoms from non-specific complaints such as fever and mild dyspnea, to fulminant hemodynamic imbalances and sudden death ( 114 ). In fact, myocarditis is considered as a common cause of sudden cardiac death ( 115 ). In a review article which included 1230 patients who initially had unexplained cardiomyopathy, 9% of the patients were eventually diagnosed with myocarditis ( 116 ). A link between vaccines and myocarditis was repeatedly reported in medical literature. For instance, in a review data of 35,188 individuals from the Vaccine Adverse Event Reporting System (VAERS), 8 cases of myocarditis were registered in individuals below 18 years of age, and 12 cases in elderly persons ( 117 ). However, it should be mentioned that VAERS presents some limitations, including the fact that it is a passive reporting system, and, therefore, information collected could be inaccurate, incomplete, coincidental, or unverifiable, warranting thorough epidemiological surveys to confirm such findings.

In another study designed to determine the incidence of cardiac symptoms and subclinical myocarditis/pericarditis after smallpox and trivalent influenza vaccine, out of 1081 individuals who received the smallpox vaccine, 4 Caucasian males were diagnosed with probable myocarditis and 1 female with suspected pericarditis. The study did not find any possible or probable cases of myocarditis/pericarditis following trivalent influenza vaccine however, some patients developed new cardiac symptoms like chest pain, dyspnea, and palpitations ( 118 ). One study that focused on myopericarditis following smallpox virus vaccination in the US military found that the incidence of myopericarditis was 7.5 times higher in soldiers who received the vaccine compared to the expected rate ( 119 ). Influenza vaccines as well were associated with cases of myocarditis ( 120 – 122 ). While myocarditis is well documented in regard to COVID-19 ( 123 ), the correlation with myocarditis and various types of COVID-19 vaccines is illustrated hereby.

BNT162b2 (BioNTech/Pfizer) and Myocarditis

Myocarditis was documented by several case reports in individuals after receiving the COVID-19 vaccines ( 124 – 126 ). Montgomery et al. ( 127 ) investigated the association between myocarditis and the mRNA COVID-19 vaccines in healthy military members of the US army. After approximately 2.8 million mRNA vaccine doses, 23 persons were diagnosed with myocarditis within 4 days of vaccination. Seven out of the 23 cases (30%) received the BNT162b2 vaccine and most developed the symptoms following the second dose. The study concluded that further evaluation of this rare adverse effect is warranted. Furthermore, several large-scale studies were conducted to evaluate this association in Israel. For instance, Barda and colleagues ( 128 ) evaluated 884,828 people vaccinated with BNT162b2, based on data from the largest health care organization in Israel. While the BNT162b2 vaccine was not associated with most of the side effects searched, the vaccine was strongly associated with increased risk of myocarditis. The risk was calculated as 2.7 events per 100,000 people, with highest risk among young men with a median age of 25. Another study conducted by Witberg et al. ( 129 ) searched specifically regarding the diagnosis of myocarditis among individuals in the largest health care organization in Israel that received at least one dose of the BNT162b2 vaccine. Among more than 2.5 million vaccinated members who were 16 years of age or older, the estimated incidence of myocarditis was 2.13 cases per 100,000 people. The highest incidence was documented in young male patients aged 16-29, the majority with mild to moderate disease. In addition, Mevorach and colleagues ( 130 ) followed the diagnosis of myocarditis in Israel after approximately 5.1 million people were vaccinated with two doses of the mRNA COVID-19 vaccines. A total of 283 persons developed symptoms of myocarditis, 142 (50%) occurred after the BNT162b2 vaccine, whereas 136 were eventually diagnosed with probable or definitive myocarditis. Most of the cases were mild and the highest incidence was recorded following the second dose and in young male patients aged 16-19 years. In comparison with unvaccinated people, the rate ratio 30 days after the second dose was 2.35 (95% CI, 1.10 to 5.02). The authors concluded that despite the low incidence, it increased after the BNT162b2 vaccine, all were mild in severity. Following the extension of the FDA authorization of the emergency use of the BNT162b2 vaccine to include children aged 12-16 in May 2021, Dionne et al. ( 131 ) presented a case series of 15 children with myocarditis after receiving the BNT162b2 vaccine. Similarly, myocarditis developed mainly in boys and after the second dose, all with a mild course.

mRNA-1273 (Moderna) and Myocarditis

Myocarditis is a rare post-mRNA-vaccine sequela, and was seen mostly among young, vaccinated males. Up to June 2021, around 1226 reports of myocarditis have been reported in the US after more than 296 million doses of mRNA COVID-19 vaccines administered ( 132 ). Symptoms usually began 3 days after vaccination with more than 75% of the cases happened after receiving the second dose of mRNA vaccine. The median age was 26 years of age with at least 56% of the affected people being younger than 30 years old. About 76% of the cases were found in men. Experts estimated the rate of post-vaccination myocarditis among young males aged 12-29 as 40.6 cases per million second doses of mRNA COVID-19 vaccines and 2.4 per million second doses among males aged > 30. On the other hand, a million vaccinations among young males aged 12-29 could prevent 560 hospitalizations, 138 intensive care unit (ICU) admissions, and six deaths associated with the COVID-19 infection. In turn, according to the UK MHRA, toward the end of November 2021, a total of 103 reports of myocarditis after the use of mRNA-1273 were registered. A rate of 37 suspected myocarditis cases per million doses were calculated for the mRNA-1273 COVID-19 vaccine ( 133 ). No death cases were registered. Additionally, in a study conducted by Diaz et al. ( 134 ) the mean monthly number of the cases of myocarditis during the vaccine period was found significantly higher than that before the vaccine was available (27.3 vs.16.9). The study included about 2 million subjects who received at least one dose of COVID-19 vaccine. Despite the fact that more people received the BNT162b2 vaccine than the mRNA-1273 vaccine (52.6% vs 44.4%), 55% of the cases occurred after receiving mRNA-1273 (11/20). Cases were mild, few required hospital admission, and discharged after a median of 2 days. No deaths or readmissions were reported. As mentioned earlier regarding 2.8 million doses of mRNA COVID-19 vaccines in healthy members of the US army, 23 cases of myocarditis were reported, 16 cases among persons who received the mRNA-1273 COVID-19 vaccine ( 127 ). The majority occurred after the second dose. In terms of possible pathogenetic explanation of the appearance of myocarditis, Bozkurt and colleagues ( 135 ) suggested the involvement of various mechanisms such as molecular mimicry between the viral spike protein and self-antigens. In addition, triggering of pre-existing dysregulated immune pathways in certain individuals, activation of immunologic pathways and dysregulated cytokine release, were also proposed. Regarding the higher rates of myocarditis among males, the difference in the immune response of sex hormones and underdiagnosed cases of myocarditis among women were suggested.

ChAdOx1 (AstraZeneca) and Myocarditis

While myocarditis has been extensively reported following mRNA COVID-19 vaccines, very few reports exist regarding the development of myocarditis in relation to the ChAdOx1 vaccine ( 136 ). In the latter, the patient was given a diagnosis of myopericarditis with pleuritis. The European Medicines Agency (EMA) estimated that 38 cases of myocarditis and 47 cases of pericarditis were attributed to ChAdOx1 vaccine out of 40 million vaccine doses administered in Europe until May 2021 ( 137 ). The agency ensured a monthly review of new cases but refuted the need for an accelerated investigation. In turn, the UK Health Security Agency dismissed a connection between the vaccine and reported cases, claiming that considering the extensive distribution of the ChAdOx1 vaccine in the UK, the cases are more likely attributed to the background incidence rate ( 138 ).

Vaccine-Induced Immune Thrombotic Thrombocytopenia/Vaccine-Induced Prothrombotic Immune Thrombotic Thrombocytopenia

Since the appearance of the vaccine-induced immune thrombotic thrombocytopenia (VITT)/vaccine-induced prothrombotic immune thrombotic thrombocytopenia (VIPITT) was mainly attributable to the adenoviral vector vaccines, the ChAdOx1 (AstraZeneca) and the Ad26.COV2.S (Janssen/Johnson & Johnson) vaccines, the current section starts with the AstraZeneca vaccine, then the mRNA vaccines follow.

ChAdOx1 (AstraZeneca) and VITT/VIPITT

In early 2020, as the newly authorized COVID-19 vaccinations were being distributed worldwide, cases of thrombotic events as well as thrombocytopenia following vaccination with the ChAdOx1 vaccine began to emerge ( 139 – 141 ). While the EMA and the WHO issued statements declaring the reported incidence was not enough to deduce causation and cautioning against premature pausing of vaccination programs ( 142 , 143 ), numerous European countries decided to halt the use of the vaccine pending further investigations. Subsequently, larger studies demonstrated slightly increased rates of venous thromboembolic events among the adenoviral vector vaccine recipients ( 144 , 145 ). A thorough investigation by the Pharmacovigilance Risk Assessment Committee (PRAC) introduced for the first time the notion of VITT/VIPITT ( 146 ), as a rare adverse reaction to the vaccine. VITT/VIPITT is defined as presence of venous or arterial thrombosis, thrombocytopenia, and autoantibodies (anti-PF4–polyanion or anti-PF4–heparin antibodies) within 5–30 days of vaccination with either AstraZeneca or Janssen/Johnson & Johnson COVID-19 vaccines ( 147 ). In fact, VITT/VIPITT shares many similarities with heparin-induced thrombocytopenia (HIT) as both disorders are facilitated by platelet factor 4 (PF4) autoantibodies leading to platelet activation and consumption ( 147 ). The trigger for the autoantibody formation is poorly understood, as the cases reported had no previous exposure to heparin ( 148 ), and the autoantibodies were not found to cross-react with the viral spike proteins indicating a previous infection ( 139 ). Furthermore, epidemiological data illustrated that over 85% of VITT/VIPITT cases occurred in women under 60 years of age, despite higher rates of vaccination among the elderly ( 146 ). Therefore, the findings supported the assumption that VITT/VIPITT is most likely to be an autoimmune phenomenon. While its exact pathogenesis has not been established yet, a recent study demonstrated that ChAdOx1 COVID-19 vaccine induces higher rates of inflammation and platelet activation compared to other COVID-19 vaccines ( 149 ). The VITT/VIPITT autoantibodies consequently bind to PF4 ( 150 ), a chemokine secreted from activated platelets, in a site that corresponds to the heparin-binding site. In turn, immune complexes formation induces FcγRIIA receptor mediated platelet activation leading to widespread thrombosis with secondary thrombocytopenia due to platelet consumption ( Figure 1 ) ( 148 , 150 ). As the ChAdOx1 vaccination has proven to be effective in prevention of COVID-19 infection ( 151 , 152 ), and due to the rarity of serious adverse events such as VITT/VIPITT, it is important to view this phenomenon in the proper clinical context.

An external file that holds a picture, illustration, etc.
Object name is fimmu-13-872683-g001.jpg

The pathogenesis of vaccine-induced immune thrombotic thrombocytopenia (VITT). Vector-based COVID-19 vaccines, particularly ChAdOx1 COVID-19 vaccine was shown to induce platelet activation alongside platelet factor 4 (PF4) autoantibodies formation. The activated platelets secret greater amount of PF4 which binds to autoantibodies forming the PF4-IgG complex. As a result, FcγRIIA receptor mediated platelet activation leads to widespread thrombosis and secondary thrombocytopenia.

BNT162b2 (BioNTech/Pfizer) and VITT/VIPITT

Since the mass vaccination campaign began with the BNT162b2 vaccine, several case reports have been published regarding the combination of thrombotic events together with thrombocytopenia, in a form of cerebral venous sinus thrombosis, VITT/VIPITT, and acquired thrombotic thrombocytopenic purpura (TTP) several days following vaccination with the BNT162b2 vaccine ( 153 – 162 ). In a large-scale study based on national data made up of 29,121,633 vaccinated individuals with the first dose of BNT162b2 vaccine ( 163 ), the association between VITT/VIPITT occurring as post-vaccine and post-infection was investigated. Among the enrollees, 9,513,625 were vaccinated with the BNT162b2 vaccine, and 1,758,095 were in the post-infection group as they were previously infected with COVID-19. An increased risk of arterial thromboembolism, cerebral venous sinus thrombosis, and ischemic stroke after the BNT162b2 vaccine was found however, an even higher risk of these associations was documented following COVID-19 infection. The study concluded an increased risk of hematological and vascular events for short-time intervals following the BNT162b2 vaccine. Still, most of these events were substantially higher and more prolonged after COVID-19 infection than after vaccination in the same population. In contrast, in another large-scale study using a national prospective cohort in Scotland with regard to hematologic and vascular events following vaccination with various vaccine types, no positive correlations were found between BNT162b2 and thrombocytopenic, thromboembolic, and hemorrhagic events ( 164 ).

mRNA-1273 (Moderna) and VITT7VIPITT

Cases of ITP and TTP after mRNA-1273 COVID-19 vaccine have been documented since the early vaccination campaign ( 165 – 168 ). A flare of familial thrombocytopenia has been reported as well ( 169 ). In addition, 13 cases of thrombocytopenia following 16 million doses of mRNA-1273 were found in the USA, suggesting a rate of 0.8 cases per million ( 170 ). Lee et al. ( 171 ) discussed 20 cases of thrombocytopenia among 20 million people who were vaccinated by at least one dose of the BNT162b2 or the mRNA1273 vaccines. A total of 17 cases considered new-onset ITP, and 11 out of the 20 cases received mRNA-1273. Furthermore, in a study based on the WHO Vigibase which includes 361 million COVID-19 vaccinated people, 2161 thrombotic events were noted, 325 of the cases appeared after the mRNA-1273 vaccine ( 172 ). Among the 325 thrombotic events, only 8 had associated thrombocytopenia. Regarding VITT/VIPITT, a German study assessed the rate of cerebral sinus and venous thrombosis (CVT) within 1 month of the first dose of the BNT162b2, ChAdOx1 and mRNA‐1273 COVID-19 vaccines and the frequency of VITT/VIPITT as the causing mechanism ( 173 ). The authors identified 45 cases of CVT, none of the cases occurred after the administration of mRNA-1273. Similar findings were shown by Krzywicka et al. ( 174 ) as out of 213 European patients with CVT, only one patient received the mRNA-1273 vaccine. According to the same study, out of 107 patients who had CVT alongside thrombocytopenia, no patient received the mRNA-1273 vaccine. In addition, only 5 possible cases of CVT were reported in Europe among 4 million subjects who received the mRNA-1273 vaccine ( 175 ). In summary, the risk of thrombocytopenia and thrombotic events in mRNA-1273 vaccinated people appears to be very low. In fact, the incidence of CVT among people vaccinated with the mRNA COVID-19 vaccines, both the BNT162b2 and mRNA-1273, was shown to be lower than that among people infected with COVID-19 (4.1 vs 39 per million) ( 176 ).

Other Autoimmune Side Effects

Immune thrombocytopenia.

ITP is a well-known autoimmune hematological condition characterized by a substantial reduction in peripheral platelet count to less than <100,000/microL due to platelet destruction by antiplatelet antibodies ( 177 ). Patients generally asymptomatic, may have minor mucocutaneous bleeding, and not uncommonly progress to life-threatening hemorrhages in severe cases ( 178 ). ITP was reported in correlation to COVID-19, COVID-19 vaccines, as well as other vaccines ( 179 , 180 ).

Minimal Change Disease

Minimal change disease (MCD) is a histologically based renal pathology which is the leading cause of idiopathic nephrotic syndrome in adults and children ( 181 ). As cases of MCD were described after vaccination in the past ( 182 – 184 ), the reporting of cases following COVID-19 vaccines were not unforeseen.

BNT162b2 (BioNTech/Pfizer) and MCD

Several case reports have been published regarding new-onset or relapse of MCD following vaccination with the BNT162b2 vaccine. A 50-year-old healthy male presented with nephrotic syndrome and acute kidney injury four days following the first dose of the BNT162b2 vaccine ( 185 ). The diagnosis was confirmed by kidney biopsy. Renal function returned to normal within few days following treatment with corticosteroids. Following this report, similar cases in older people were presented ( 186 , 187 ). The symptoms appeared seven days following the first dose of the vaccine. MCD was confirmed by kidney biopsy as well. In addition, a relapse of MCD was also described in a patient diagnosed with MCD 20 years prior to vaccination, developed proteinuria after vaccination, which resolved within 2 weeks following corticosteroids and cyclosporine treatment ( 188 ).

mRNA-1273 (Moderna) and MCD

Though autoimmune renal AEFIs of the mRNA-1273 COVID-19 vaccine were reported, MCD was not common among them. For instance, Thappy et al. ( 189 ) presented a case of 43-year-old man who developed symptoms of minimal change disease 7 days after receiving the first dose of the mRNA-1273 vaccine. The biopsy confirmed concomitant MCD and IgA nephropathy. The patient responded well to oral steroid treatment. In addition, biopsy proven IgA nephropathy, both as new onset and flare, was previously described following the mRNA-1273 COVID-19 vaccine, manifesting as hematuria ( 190 , 191 ). Interestingly, the symptoms appeared 1-2 days after the second dose of the vaccine.

ChAdOx1 (AstraZeneca) and MCD

Several cases of MCD were described in the context of the ChAdOx1 vaccine ( 192 – 194 ). All these cases presented with a clinical picture of nephrotic syndrome that started up to 15 days after the first dose of the vaccine. The fairly short period from vaccination to presentation could be attributed to cytokine release by activated T-cells, as the ChAdOx1 vaccine has been shown to induce a robust T-cell response in most individuals ( 195 ). However, as the reported cases were limited, a definite conclusion could not be drawn. The number of cases of MCD following the vaccine was small, thus a strong association could not be inferred.

As for vasculitis, two cases of antineutrophil cytoplasmic antibody (ANCA) associated vasculitis after the second dose of the mRNA-1273 COVID-19 vaccine were reported. As a result of the new onset ANCA vasculitis, one patient became dependent on dialysis ( 196 ), while in the other case renal improvement was achieved after plasma exchange, pulse steroid and cyclophosphamide therapy ( 197 ). Additional case of ANCA vasculitis which manifested as renal failure together with pulmonary hemorrhage 3 weeks after the first dose of mRNA-1273 was also described ( 198 ).

Miscellaneous

Autoimmune hepatitis was reported as well among individuals vaccinated with various types of COVID-19 vaccines ( 199 – 202 ). In a multicenter study conducted in several countries during the early implementation of the program of vaccination against the COVID-19, Watad and colleagues ( 203 ) investigated the association between new-onset or flares of immune-mediated diseases 28-days following mRNA COVID-19 vaccination. The authors detected 27 cases; 17 (63%) had flares of their illnesses whereas 10 (37%) cases had a new-onset disease. In total, 23 (85%) patients received the BNT162b2 vaccine, 2 (7.5%) received the mRNA-1273 vaccine, and 2 (7.5%) received the ChAdOx1 vaccine. Taking into consideration the great proportion of people vaccinated, the authors concluded that immune-mediated diseases flares or new-onset temporally associated with COVID-19 vaccination are rare. Furthermore, Ishay et al. ( 204 ) presented a case series of patients with new-onset or flares of autoimmune AEFIs after the BNT162b2 vaccine. Eight patients presented with either symmetric polyarthritis, panuveitis, pericarditis, temporal arteritis-like disease, fever of unknown origin (FUO), oligoarthritis, and myocarditis following either the first dose (62.5%) or the second dose (37.5%) of the vaccine. Based on their findings, the authors concluded that immune AEFIs might occur following vaccination however, they usually follow a mild course.

A brief summary of the studies addressing autoimmune AEFIs of the most utilized COVID-19 vaccines, including the study population and the conclusion of the works, is presented in Table 1 .

Table 1

A brief summary of studies addressing immune and autoimmune side effects of COVID-19 vaccines.

Side effectVaccineAuthorsStudy populationConclusions
Pfizer (BNT162b2)Trimboli et al. ( )1 case of GBS after receiving the second dose of BNT162b2 COVID-19 vaccineAuthors believe that the clinical and laboratory findings including the lack of overt trigger are consistent with a causal association between GBS and Pfizer anti-SARS-CoV-2 vaccine
Bouattour et al. ( )1 case of GBS after 1 dose of BNT162b2 vaccineA patient who developed GBS 7 days after receiving the first dose of Pfizer-BioNTech COVID-19 vaccine
Shapiro Ben David et al. ( )Retrospective cohort study in the second largest health maintenance organization in Israel, diagnosis code for GBS after receiving at least 1 vaccine doseIn this cohort study, which included 702 patients, only 1 needed short medical care for relapse of previous syndrome, which represents a minimal risk.
Garcia-Grimshaw et al. ( )Analysis of cohort of 3,890,250 Hispanic/Latinx recipients of the BNT162b2 mRNA vaccine (613,780 of for incident GBS occurring within 30 days from vaccinationAmong recipients of the BNT162b2 mRNA vaccine, GBS may occur at the expected community-based rate
Shasha et al. ( )Individuals ≥16 years vaccinated with at least one dose of BNT162b2No association was found between vaccination, Bell’s palsy, herpes zoster or GBS. This study adds reassuring data regarding the safety of the BNT162b2 vaccine.
Garcia-Grimshaw et al. ( )Prospective observational cohort of 700,000 patients from a database of all systemic and neurologic adverse events following immunizationNon-serious events occurred in less than 1% of recipients, while serious ones occurred in only 33 (0.005%) recipients, suggesting that the vaccine is not only effective but also safe
Waheed et al. ( )1 case of GBS presenting after 1 vaccineAn 82 y/o female who presented 2 weeks after vaccination with difficulty in walking
Nishiguchi et al. ( )1 case of MFS presenting after 1 vaccineAuthors report the first case of COVID-19 vaccination-associated MFS. However, it is difficult to deny that this result may be a coincidence in time, and therefore, no cause-and-effect relationship can be concluded at this time.
Razok et al. ( )1 case of GBS presenting after 2 vaccineThe patient presentd with acute flaccid paralysis (AFP) after receiving the COVID-19 vaccine.
Miyaue et al. ( )1 case of LETM presenting after 1 vaccineThis case meets the criteria for a “probable” AEFI, considering the following features: (i) the onset of neurological symptoms occurred within a week after vaccination; (ii) the patient had no previous neurological symptoms after other vaccines, nor symptoms suggestive of prior infection; and (iii) no other cause of LETM was identified on a thorough diagnostic evaluation.
Colella et al. ( )1 case of Bell’s palsy after 1 vaccineAlthough a causal relationship cannot be established for most rare adverse events, the timing and mode of onset of the palsy strongly suggests that it was related to BNT162b2 vaccine injection.
Shibli et al. ( )Population based study in Israel comparing expected cases of Bell’s palsy with number of cases after 1 and 2 vaccine doseThe overall observed rate of Bell’s palsy after vaccination was higher than the expected rates
Kobayashi et al ( )1 case of encephalitis after 1 dose, exacerbated after 2 dose.No evidence of causal relationship was found.
Moderna (mRNA-1273)Masuccio et al. ( )1 case of GBS after 2 vaccineAccording to clinical features, a subacute GBS might be reasonably hypothesized after the administration of COVID-19 mRNA-1273 vaccine second dose, with about 6 weeks elapsing between the vaccination and the symptoms onset.
Kania et al. ( )1 case of acute disseminated encephalomyelitis after 1 vaccinationThe patient manifested a typical radiological pattern for ADEM with extensive, diffuse demyelinating lesions in the brain and along all cervical and thoracic spinal cord
Fujikawa et al. ( )1 case of neuromyelitis optica spectrum disorder presenting after 1 vaccinationConsidering the temporal association between administration of the vaccine, onset of patient’s symptoms, and previous reports of post-vaccination NMOSD, patient’s NMOSD was triggered by the SARS-CoV-2 mRNA-1273 vaccine.
Gao et al. ( )1 case of LETM after 1 vaccinationThe temporal relationship between vaccination and ATM in the case was clinically reasonable (48 h post-vaccination)
Cellina et al. ( )1 case of Bell’s palsy after 1 vaccinationThe patient complained of symptoms at 12 h from the injection. The timing of Bell’s palsy onset after mRNA vaccine administration varies
Iftikhar et al. ( )1 case of Bell’s palsy after 2 vaccinationThis case highlights the importance of vaccine history in patients presenting to the emergency department with Bell’s palsy. COVID-19 mRNA vaccines can be considered as an additional possible risk factor in the etiology of Bell’s palsy.
Martin-Villares et al. ( )1 case of Bell’s palsy after 1 vaccinationEvidence of a temporal association between the vaccine administration and the facial nerve palsy is clear: Bell´s palsy appeared 2 days after the administration of the mRNA COVID-19 vaccine
Torrealba-Acosta et al. ( )1 case of encephalitis and Sweet syndrome after 1 doseThough temporal relation was described, causality could not be proven
AstraZeneca (ChAdOx1)Min et al. ( )2 case presentations + review of 12 published casesThe two patients shared many clinical features: pure sensory manifestations, short-latency from vaccination to onset, progression duration, and no serum antibodies against gangliosides. Sensory GBS was considered the most probable diagnosis.
Oo et al. ( )4 case presentations + review of 15 published casesThese four cases can lend further weight to the likely causal link between COVID-19 vaccine AZ and GBS
McKean et al. ( )1 case of GBS following the 1st vaccinationThis is the first reported case of GBS which was temporally related to the Vaxzevria vaccine in Malta.
Introna et al. ( )1 case of GBS following the 1st vaccinationA case of GBS presenting with papilledema as atypical onset
Allen et al. ( )4 cases of GBS following 1st vaccinationThere was an interval of 11 to 22 days between vaccination and symptom onset.
Notghi et al. ( )1 case of LETM after 1st vaccination58-year-old man admitted to hospital 10 days after his first AstraZeneca COVID-19 vaccination with progressive neurological symptoms and signs, and investigations and imaging consistent with LETM
Pagenkopf et al. ( )1 case of LETM after 1st vaccinationThe case of LETM presented here shows a close temporal association to COVID-19 vaccination, as symptoms occurred within 11 days post injection of first dose AZD1222, AstraZeneca
Helmchen et al. ( )1 case of LETM in a patient with MS after 1st vaccinationThe case suggests that the vector-based COVID-19 vaccine should not be used in RRMS if mRNA vaccines are available.
Voysey et al. ( )Evaluation of 4 controlled trials in 3 countries (11636 patients)ChAdOx1 nCoV-19 has an acceptable safety profile and has been found to be efficacious against symptomatic COVID-19 in this interim analysis of ongoing clinical trials.
Hsiao et al. ( )1 case of acute transverse myelitis after 1st vaccinationAlthough they rarely occur, the association of the COVID-19 vaccine and the disease, along with other neurological complications, should not be ignored
Malhotra et al. ( )1 case of acute transverse myelitis after 1st vaccinationConsidering an incidence of 1–4 cases per million per year, 6 an event of myelitis occurring after more than 50 million vaccine doses appears fairly acceptable
Bonifacio et al. ( )5 cases of bilateral facial weakness after 1 vaccinationThe incidence of five cases of the very uncommon BFP variant of GBS occurring within 2 weeks of Vaxzevria is further suggestive of an etiological link.
Hasan et al. ( )1 case of paresthesia and progressive weakness presenting after 1 caseNo direct link could be ascertained
Kanabar et al. ( )2 patients with GBS presenting after 1 vaccinationBoth patients described in this report had bilateral facial weakness at presentation
Maramattom et al. ( )7 patients who presented with GBS after 1 vaccinationPatients were in their 5 to 7 decades of life and predominantly female. All patients progressed to areflexic quadriplegia, and six of the seven cases required mechanical ventilation for respiratory failure. All seven cases had bilateral facial paresis, which usually occurs in fewer than 20% of unselected GBS cases
Tan et al. ( )1 case of LETM after 1 vaccinationAlthough TM following vaccination is rare, the temporal causality of LETM, in this case, is undeniable
Zuhorn et al. ( )3 cases of encephalitis, one after the 1 dose, others not mentionedThe complication of autoimmune encephalitis after ChAdOx1 nCoV-19 vaccination appears to be very rare. Clearly, the benefit of vaccination outweigh the risks
Multiple vaccine typesKaulen et al. ( )21 consecutive cases of neurological autoimmunity, which occurred 3–23 days following SARS‐CoV‐2 vaccinationsA large series of neurological autoimmunity in temporal association with various SARS‐CoV‐2 vaccines (BNT162b2, ChAdOx1 and mRNA‐1273) is reported
Koh et al. ( )A prospective study at 7 acute hospitals in Singapore of hospitalized patients who were referred for neurological complaints and had COVID-19 mRNA vaccinesOver a 4-month period during which approximately 1.4 million people received the COVID-19 mRNA vaccines, authors recorded a spectrum of neurological disorders in only 457 hospitalized patients
Loo et al. ( )A retrospective study examining all persons presenting with acute-onset polyradiculoneuropathy from January 1, 2021, to June 30, 2021 who were admitted to UK hospitalsMost cases identified in the study (87.5%) occurred after the AstraZeneca vaccine
Renoud et al. ( )133,883 cases of adverse drug reactions reported with mRNA COVID-19 vaccines in the World Health Organization pharmacovigilance databaseWhen compared with other viral vaccines, mRNA COVID-19 vaccines did not display a signal of facial paralysis
Ismail et al. ( )Review of 32 cases of CNS demyelination after all vaccine typesCNS demyelination was reported following all types of authorized COVID-19 vaccines (no protein-based vaccine was authorized at the time of writing). Neurological symptoms appeared within the first 1–2 weeks in most cases. Females comprised the majority of cases. Furthermore, more than half of the cases had history of probable or definite autoimmune diseases
Ozonoff et al. ( )Literature review of Bell’s palsy after all types of COVID vaccinesThe observed incidence of Bell’s palsy after mRNA vaccines is between 3·5-times and 7-times higher than would be expected in the general population.
Sato et al. ( )Analysis of Bell’s palsy cases databases after mRNA vaccinesThe incidence of facial nerve palsy as a non-serious AEFI may be lower than, or equivalent to, that for influenza vaccines.
Patone et al. ( )Case series studies investigating hospital admissions from neurological complications after 1 dose of AstraZeneca or Pfizer vaccinesAuthors found an increased risk of hospital admission for GBS (15–21 days and 22–28 days), Bell’s palsy (15–21 days) and myasthenic disorders (15–21 days) in those who received the ChAdOx1nCoV-19 vaccine. Second, an increased risk of hospital admission for hemorrhagic stroke (1–7 days and 15–21 days) was observed in those who received the BNT162b2 vaccine
Pfizer (BNT162b2)Barda et al. ( )884,828 people on a nation-wide settingThe vaccine was associated with an excess risk of myocarditis (1 to 5 events per 100,000 persons)
Montgomery et al. ( )23 male patients within the US Military Health System who experienced myocarditis after COVID-19 vaccination between January and April 2021.The consistent pattern of clinical presentation, rapid recovery, and absence of evidence of other causes support the diagnosis of hypersensitivity myocarditis
Abu Mouch et al. ( )6 cases of myocarditis, which occurred shortly after BNT162b2 vaccinationFive patients presented shortly after the second vaccine dose and one patient presented 16 days after receiving his first vaccine dose
Mevorach et al. ( )Retrospectively review data obtained from December 20, 2020, to May 31, 2021, regarding all cases of myocarditis in IsraelThe incidence of myocarditis, although low, increased after the receipt of the BNT162b2 vaccine, particularly after the second dose among young male recipients. The clinical presentation of myocarditis after vaccination was usually mild.
Larson et al. ( )8 patients 2-4 days post mRNA-based vaccineThe temporal association between receiving an mRNA-based COVID-19 vaccine and the development of myocarditis is notable, potentially supporting the hypothesis that myocarditis could be an mRNA vaccine–related adverse reaction
Dionne et al. ( )Case series of children younger than 19 years hospitalized with myocarditis within 30 days of BNT162b2 vaccine.Myocarditis was diagnosed in children after COVID-19 vaccination, most commonly in boys after the second dose
Witberg et al. ( )Nationwide Israeli cohort through Health care service database evaluating myocarditis cases after Pfizer vaccinesThe estimated incidence of myocarditis was 2.13 cases per 100,000 persons; the highest incidence was among male patients between the ages of 16 and 29 years. Most cases of myocarditis were mild or moderate in severity.
Moderna (mRNA-1273)Gargano et al. ( )296 million doses of administered mRNA (Pfizer and Moderna) COVID-19 vaccines up to June 11,2021 in the US.Myocarditis reporting rates were 40.6 cases per million second doses of mRNA COVID-19 vaccines administered to males aged 12−29 years and 2.4 per million second doses administered to males aged ≥30 years
UK Medicines & Healthcare products Regulatory Agency ( )1.5 million recipients of the first doses and approximately 1.3 million recipients of the second doses of mRNA-1273 in the UK.The expected benefits of the vaccines in preventing COVID-19 and serious complications associated with COVID-19 far outweigh any currently known side effects in the majority of patients.
AstraZeneca (ChAdOx1)Hung et al. ( )1 case of myopericarditis with pleuritisSymptoms occurred 7 days post-vaccination, and the patient was hospitalized for 12 days with a total recovery. Due to a negative result for other etiologies, the possibility of vaccine-related myopericarditis with bilateral pleural effusion cannot be totally excluded.
Multiple vaccine typesVidula et al. ( )Two healthy young patients with clinically suspected myocarditis after receiving an mRNA-based COVID-19 vaccineWhile endomyocardial biopsy was not performed, both patients met the diagnostic criteria for clinically suspected myocarditis. The temporal association of the receipt of the vaccine and absence of other plausible causes suggest the vaccine as the likely precipitant
Diaz et al. ( )2,000,287 individuals receiving at least 1 COVID-19 vaccination in the USThis study shows that myocarditis after vaccination is primary seen in younger male individuals a few days after the second vaccination. Pericarditis may be more common than myocarditis among older patients.
Pfizer (BNT162b2)Maayan et al. ( )4 patients from two academic medical centers who developed TTP were identified from mid- February to mid- March 2021A disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13) activity should be evaluated in patients with history of aTTP before and after any vaccination, especially the SARS‐CoV‐2 vaccination
de Bruijn et al. ( )1 case report of TTP after first BNT162b2 vaccineThis is the first case report of iTTP after mRNA-based COVID-19 vaccination in a previously TTP-naïve patient.
Akiyama et al. ( )1 case of ITP after 1 vaccinationAn extremely rare case of secondary ITP presumed to have occurred after BNT162b2 vaccination
Dias et al. ( )2 cases of thromboembolism after 1 vaccinationIn both patients, there was no evidence of thrombocytopenia or antiplatelet antibodies, and alternative causes for cerebral venous thrombosis were found. As such, despite the temporal relation of both cases to vaccine administration, these types of cerebral venous thrombosis do not seem to be pathophysiological different from cerebral venous thrombosis not associated to SARS-CoV-2 vaccination
Ganzel et al. ( )1 case of ITP after 1 vaccinationMay have a temporal relationship with administration of the Pfizer-BioNTech COVID-19
vaccine
King et al. ( )1 case of ITP after 2 vaccinationITP should be considered a severe AE of the BNT162b2 mRNA COVID-19 vaccine.
Matsumura et al. ( )2 cases of ITP after 1 vaccinationWhether or not ITP is triggered by the vaccination or not is difficult to identify
Rodríguez et al. ( )1 case of VITT after 1 vaccinationThis case meets the Brighton Collaboration case definition of VITT, with thrombocytopenia and thrombosis without prior heparin exposure
Waqar et al. ( )1 case of TTP after 2 dose of vaccinationFurther studies are, however, needed to verify possible associations between microangiopathic, thrombocytopenic thrombotic disorders and the administration of vaccines against COVID-19
Yoshida et al. ( )1 case of TTP after 1 vaccinationThe first case of acquired TTP in Japan may have been associated with the first dose of the BNT162b2 mRNA COVID-19 vaccine
Moderna (mRNA-1273)Hines et al. ( )1 case of ITP after 1 vaccinationThe temporal relationship of her vaccination with thrombocytopenia and abnormal liver enzymes points towards the Moderna mRNA-1273 SARS-CoV-2 vaccine as the most likely inciting factor
Karabulut et al. ( )1 case of TTP in a patient with known ITP and TTP after 1 vaccinationThe close temporal association between vaccine administration, recent COVID-19, and relapse of remitted TTP raises concern for an enhanced immune reaction to COVID-19 vaccine in the setting of recent COVID-19 and underlying autoimmune disease
Malayala et al. ( )1 case of thrombocytopenia after 1 vaccinationAuthors attribute this thrombocytopenia and purpuric rash as the side effects of the mRNA-1273 vaccine
Su et al. ( )1 case of VITT in a patient with pancreatic cancer after 1 vaccinationThis case study was the first to report a cancer patient who was diagnosed with VITT after mRNA-1273 vaccination
Toom et al. ( )Patient with ITP who presented with flareup after 1 vaccinationThe temporal sequence of the events suggests an exacerbation of patient’s chronic thrombocytopenia related to the receipt of the mRNA‐1273 Covid‐19 vaccine
AstraZeneca (ChAdOx1)Greinacher et al. ( )11 patients in Germany and Austria with thrombocytopenia and clottingVaccination with ChAdOx1 nCov-19 can result in the rare development of immune thrombotic thrombocytopenia mediated by platelet-activating antibodies against PF4, which clinically mimics autoimmune heparin-induced thrombocytopenia
Schultz et al. ( )5 patients in Norway (healthcare workers) with thrombocytopenia and clottingFive cases occurred in a population of more than 130,000 vaccinated persons, they represent a rare vaccine-related variant of spontaneous heparin-induced thrombocytopenia
Pottegård et al. ( )282572 patients in Norway and Denmark who experienced clotting events after vaccinationExcess rate of venous thromboembolism, including cerebral venous thrombosis, among recipients of the Oxford-AstraZeneca covid-19 vaccine ChAdOx1-S within 28 days of the first dose
Perry et al. ( )99 patients from 43 hospitals in the UK with clotting and thrombocytopeniaCerebral venous thrombosis appears to be more severe in the context of VITT
Scully et al. ( )23 patients who presented with thrombosis and thrombocytopenia after 1 vaccinationTesting for antibodies to platelet factor 4 (PF4) was positive in 22 patients (with 1 equivocal result) and negative in 1 patient
Multiple vaccine typesSchulz et al. ( )45 CVT cases occurring after 7,126,434 first vaccine doses of all types- using official statistics of 9 German states.The findings point toward a higher risk for CVT after ChAdOx1 vaccination, especially for women
Krzywicka et al. ( )213 European patients with CVT after any vaccinationCerebral venous sinus thrombosis occurring after ChAdOx1 nCov-19 vaccination has a clinical profile distinct from CVST unrelated to vaccination. Only CVST after ChAdOx1 nCov-19 vaccination was associated with thrombocytopenia
Welsh et al. ( )Case-series study of thrombocytopenia after mRNA vaccines using Vaccine Adverse Event Reporting System (VAERS)The number of thrombocytopenia cases reported to the Vaccine Adverse Event Reporting System (VAERS) does not suggest a safety concern attributable to mRNA COVID-19 vaccines at this time
Cines et al. ( )4 million subjects which received any vaccine type in EuropeCases of immune thrombocytopenia and bleeding without thrombosis that were induced or revealed after exposure to the messenger RNA (mRNA)–based vaccines produced by Moderna (mRNA-1273) and Pfizer–BioNTech (BNT162b2). The study has now highlighted three independent descriptions of 39 persons with a newly described syndrome characterized by thrombosis and thrombocytopenia that developed 5 to 24 days after initial vaccination with ChAdOx1 nCoV-19 (AstraZeneca)
Lee et al. ( )20 million people who have received at least one dose of Pfizer or Moderna vaccines. in the USAThe possibility that the Pfizer and Moderna vaccines have the potential to trigger ITP (including clinically undiagnosed cases) cannot be excluded, albeit very rarely. Distinguishing vaccine‐induced ITP from coincidental ITP presenting soon after vaccination is impossible at this time.
Smadja et al. ( )361 million vaccinated people from the whole world with any vaccine typeThe authors suggest that thrombotic events, including CVT, might occur in association with all three vaccines, but this hypothesis requires further investigations
Torjesen et al. ( )Using a US electronic health records, comparing incidence of cerebral venous thrombosis in patients two weeks after a COVID-19 diagnosis with that in patients two weeks after COVDI-19 vaccination in all vaccine typesSARS-CoV-2 infection is associated with more risk for CVT than COVID-19 mRNA vaccines
Hippisley-Cox et al. ( )Using a UK national data on covid-19 vaccination (AstraZeneca or Pfizer) and hospital admissions due to thrombocytopenia, venous thromboembolism, and arterial thromboembolismIncreased risks of hematological and vascular events that led to hospital admission or death were observed for short time intervals after first doses of the ChAdOx1 nCoV-19 and BNT162b2 mRNA vaccines. The risks of most of these events were substantially higher and more prolonged after SARS-CoV-2 infection than after vaccination in the same population.
Simpson et al. ( )National prospective cohort estimating hematological and vascular adverse events after 1 vaccination with either AstraZeneca or PfizerA first dose of ChAdOx1 was found to be associated with small increased risks of ITP, with suggestive evidence of an increased risk of arterial thromboembolic and hemorrhagic events
Pfizer (BNT162b2)Avci et al. ( )1 case report after 1 BNT162b2 vaccineAlthough the exact cause of autoimmune reactions is unknown, an abnormal immune response and bystander activation induced by molecular mimicry is considered a potential mechanism, especially in susceptible individuals
Moderna (mRNA-1273)Zin Tun et al. ( )1 case report of autoimmune hepatitis with Moderna vaccine+ review of other casesThis case illustrates immune-mediated hepatitis secondary to the Moderna vaccine, which on inadvertent re-exposure led to worsening liver injury with deranged synthetic function
AstraZeneca (ChAdOx1)Rela et al. ( )2 cases of AIH following 1 vaccinationThere were no clear clinical or biochemical features apart from a chronological association to differentiate patients’ vaccine-related AIH from idiopathic AIH.
Clayton-Chubb et al. ( )1 case of AIH following 1 vaccinationThis case supports the notion of COVID-19 vaccine-triggered autoimmune phenomena irrespective of the vaccine’s mechanism of action, though this is the first report of an adenovirus-based vaccine precipitating AIH
Pfizer (BNT162b2)Lebedev et al. ( )1 case report after 1 BNT162b2 vaccineThe association between the vaccination and MCD is at this time temporal and by exclusion, and by no means firmly established
D’Agati et al. ( )1 case report after 1 BNT162b2 vaccineThe strong temporal association with vaccination suggests a rapid T cell–mediated immune response to viral mRNA as a possible trigger for podocytopathy
Maas et al. ( )1 case report after 1 BNT162b2 vaccineThis case can provide support for a potential association between the BNT162b2 vaccine and onset of MCD
Komaba et al. ( )1 case report after 1 BNT162b2 vaccineWhether SARS-CoV-2 vaccines could trigger a relapse of MCD or other forms of nephrotic syndrome is currently unclear.
AstraZeneca (ChAdOx1)Leclerc et al. ( )1 case of AKI due to MCD after 1st vaccinationThis report suggests a potential relationship between MCD and the Oxford-AstraZeneca COVID-19 vaccine
Morlidge et al. ( )2 cases of previous MCD patients relapsing after 1st vaccinationAt 2 days after vaccination, one would assume the vaccine triggered a more generalized cytokine-mediated response. Others have postulated that symptoms after 4 days represent a rapid T cell–mediated response to viral mRNA
Anupama et al. ( )1 case of nephrotic syndrome after 1st vaccinationThe temporal profile of nephrotic syndrome after the coronavirus disease 2019 vaccination and absence of any other precipitating factors points toward the vaccine as a possible trigger
Pfizer (BNT162b2)Ishay et al. ( )8 patients presenting with or flares of existing autoimmune conditionsAuthors observed that while immune phenomena may occur following vaccination, they usually follow a mild course and require modest therapy
Watad et al. ( )27 cases of immune-mediated diseases flares or new disease onset within 28-days of SARS-CoV-2 vaccinationDespite the high population exposure in the regions served by these centers, IMDs flares or onset temporally-associated with SARS-CoV-2 vaccination appear rare. Most are moderate in severity and responsive to therapy although some severe flares occurred.

Immune and autoimmune AEFIs after COVID-19 vaccines are rare and mostly non-life-threatening. Their rate follows 1 out of 3 possibilities, either it is very low, or similar to the occurrence rate in the general population, or even much lower in comparison to the COVID-19 infection itself. Of AEFIs mentioned, Bell’s palsy and myocarditis seemingly have the greatest risk when it comes to the mRNA-based COVID-19 vaccines however, in addition to their rarity, the disease course is mild and full recovery is the rule. In turn, GBS and VITT/VIPITT were found to be associated mainly with the adenovirus vector based COVID-19 vaccines, still with a low rate. Addressing this sort of AEFIs, their severity, and long-term effects of the COVID-19 vaccines while fighting against vaccine hesitancy is of great importance. Doubtlessly, such efforts will decrease public concern, increase vaccination coverage rate, and guide physicians towards the rapid identification of AEFIs, reassuring patients, and applying appropriate treatment in a timely manner.

Author Contributions

NM: Supervision, Writing- Review and Editing. NL: Writing - Original Draft, Software. AO: Writing - Original Draft. RS: Writing - Original Draft. AA: Writing - Original Draft. MA: Writing - Original Draft. MZ: Writing - Original Draft. NB: Conceptualization, Writing- Review and Editing. All authors contributed to the article and approved the subitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

Literature review of HPV vaccine delivery strategies: considerations for school- and non-school based immunization program

Affiliations.

  • 1 Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA. Electronic address: [email protected].
  • 2 Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA.
  • PMID: 24295804
  • DOI: 10.1016/j.vaccine.2013.11.070

School-based vaccination is becoming a more widely considered method of delivering HPV immunizations to an adolescent population; however, many countries do not have experience with delivering adolescent vaccines or school-based programs. This literature review will summarize the experiences from countries implementing non-health facility-based and health facility-based vaccination programs and assess HPV vaccine coverage. In October 2012, a systematic search in PubMed for studies related to the evaluation of national/regional, pilot, or demonstration HPV immunization programs that worked within existing health system yielded nine articles, representing seventeen countries. School-based programs achieved high HPV vaccination coverage rates in 9 to 13-year-old girls across the different studies and geographic locations, suggesting non-health facility-based programs are possible for HPV vaccine introduction. Grade-based, compared to age-based, eligibility criteria may be easier to implement in school settings. More studies are needed to explore the methods to standardize estimates for HPV vaccine coverage so that programs can be appropriately evaluated.

Keywords: HPV vaccine; School-based vaccination; Vaccine delivery strategies.

Copyright © 2013 Elsevier Ltd. All rights reserved.

PubMed Disclaimer

Similar articles

  • Challenges, lessons learned and results following the implementation of a human papilloma virus school vaccination program in South Australia. Watson M, Shaw D, Molchanoff L, McInnes C. Watson M, et al. Aust N Z J Public Health. 2009 Aug;33(4):365-70. doi: 10.1111/j.1753-6405.2009.00409.x. Aust N Z J Public Health. 2009. PMID: 19689598
  • Human papillomavirus (HPV) vaccine coverage achievements in low and middle-income countries 2007-2016. Gallagher KE, Howard N, Kabakama S, Mounier-Jack S, Burchett HED, LaMontagne DS, Watson-Jones D. Gallagher KE, et al. Papillomavirus Res. 2017 Dec;4:72-78. doi: 10.1016/j.pvr.2017.09.001. Epub 2017 Oct 3. Papillomavirus Res. 2017. PMID: 29179873 Free PMC article.
  • Coverage from Ontario, Canada's school-based HPV vaccine program: the first three years. Wilson SE, Harris T, Sethi P, Fediurek J, Macdonald L, Deeks SL. Wilson SE, et al. Vaccine. 2013 Jan 21;31(5):757-62. doi: 10.1016/j.vaccine.2012.11.090. Epub 2012 Dec 13. Vaccine. 2013. PMID: 23246265
  • Human papillomavirus vaccine introduction--the first five years. Markowitz LE, Tsu V, Deeks SL, Cubie H, Wang SA, Vicari AS, Brotherton JM. Markowitz LE, et al. Vaccine. 2012 Nov 20;30 Suppl 5:F139-48. doi: 10.1016/j.vaccine.2012.05.039. Vaccine. 2012. PMID: 23199957 Review.
  • Implementation of human papillomavirus immunization in the developing world. Kane MA, Serrano B, de Sanjosé S, Wittet S. Kane MA, et al. Vaccine. 2012 Nov 20;30 Suppl 5:F192-200. doi: 10.1016/j.vaccine.2012.06.075. Vaccine. 2012. PMID: 23199963 Review.
  • Sociodemographic determinants of HPV vaccine awareness, uptake, and intention among parents of adolescents in France 2021-22. Rivera AF, Dussault JM, Oudin Doglioni D, Chyderiotis S, Sicsic J, Barret AS, Raude J, Gauchet A, Gagneux-Brunon A, Bruel S, Michel M, Le Duc-Banaszuk AS, Thilly N, Mueller JE. Rivera AF, et al. Hum Vaccin Immunother. 2024 Dec 31;20(1):2381300. doi: 10.1080/21645515.2024.2381300. Epub 2024 Aug 6. Hum Vaccin Immunother. 2024. PMID: 39105306 Free PMC article.
  • Understanding Barriers to Human Papillomavirus Vaccination among Parents of 9-10-Year-Old Adolescents: A Qualitative Analysis. Agana-Norman DFG, Martinez Martinez M, Shanmugasundaram M, Berenson AB. Agana-Norman DFG, et al. Vaccines (Basel). 2024 Feb 27;12(3):245. doi: 10.3390/vaccines12030245. Vaccines (Basel). 2024. PMID: 38543879 Free PMC article.
  • A systematic literature review of human papillomavirus vaccination strategies in delivery systems within national and regional immunization programs. Felsher M, Shumet M, Velicu C, Chen YT, Nowicka K, Marzec M, Skowronek G, Pieniążek I. Felsher M, et al. Hum Vaccin Immunother. 2024 Dec 31;20(1):2319426. doi: 10.1080/21645515.2024.2319426. Epub 2024 Feb 27. Hum Vaccin Immunother. 2024. PMID: 38410931 Free PMC article.
  • Between now and later: a mixed methods study of HPV vaccination delay among Chinese caregivers in urban Chengdu, China. Yim VW, Wang Q, Li Y, Qin C, Tang W, Tang S, Jit M, Smith JS, Larson HJ, Tucker JD, Li J, Lin L, Wu D. Yim VW, et al. BMC Public Health. 2024 Jan 15;24(1):183. doi: 10.1186/s12889-024-17697-6. BMC Public Health. 2024. PMID: 38225563 Free PMC article.
  • A comprehensive narrative review of challenges and facilitators in the implementation of various HPV vaccination program worldwide. Aggarwal S, Agarwal P, Gupta N. Aggarwal S, et al. Cancer Med. 2024 Feb;13(3):e6862. doi: 10.1002/cam4.6862. Epub 2024 Jan 11. Cancer Med. 2024. PMID: 38213086 Free PMC article. Review.

Publication types

  • Search in MeSH

Related information

  • Cited in Books

LinkOut - more resources

Full text sources.

  • Elsevier Science

Other Literature Sources

  • scite Smart Citations
  • MedlinePlus Health Information
  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

IMAGES

  1. Literature Review Guidelines

    literature review on immunization

  2. Executive Summary

    literature review on immunization

  3. Immunization Recommendations for Pediatric Patients with CKD, Nephrotic

    literature review on immunization

  4. ≫ Childhood Immunization Free Essay Sample on Samploon.com

    literature review on immunization

  5. Executive Summary

    literature review on immunization

  6. IMMUNIZATION AND THE HEALTH CARE SYSTEM

    literature review on immunization

COMMENTS

  1. Pediatric Inpatient Immunizations: A Literature Review

    CONTEXT:. Timely vaccine uptake in children remains suboptimal. Eliminating missed opportunities is key to increasing childhood immunization rates, and hospitalization offers another potential setting to vaccinate.OBJECTIVE:. To better understand pediatric inpatient immunization programs, including vaccination rates of inpatients, parental and provider attitudes, barriers to vaccine delivery ...

  2. Pediatric Inpatient Immunizations: A Literature Review

    Data extraction: Abstracts were screened for relevance, articles were read, and themes were identified. Results: Children who are hospitalized have been shown to have lower immunization rates compared with the general population, with 27% to 84% of pediatric inpatients due or overdue for vaccines nationally when verified with official records.

  3. Interventions to Improve Immunization Coverage Among Children and

    BACKGROUND AND OBJECTIVES. Vaccinations are recognized as a feasible, cost-effective public health intervention for reducing the burden and associated mortality of many infectious diseases. The purpose of this study was to evaluate the effectiveness of potential interventions to improve the uptake of vaccines among children and adolescents.METHODS. We performed a literature search until ...

  4. Factors affecting childhood immunization: Thematic analysis of parents

    Themes were then identified using thematic analysis. A total of 44 studies met the review criteria and were summarized and categorized into 4 themes: barriers to immunization, parental knowledge, attitude and behavior (KAB), health system factors and HCWs' KAB. This review found that immunization decision-making is a complex process.

  5. Review of Scientific Findings

    The literature published in the past 10 years was chosen to fill the gap since the 2002 IOM review and because several changes to the immunization schedule have been made since 2000 (e.g., addition of the pneumococcal and rotavirus vaccines).

  6. A systematic review and meta-analysis of strategies to promote ...

    Although immunization can dramatically curb the mortality and morbidity associated with vaccine-preventable diseases, vaccination uptake remains suboptimal in many areas of the world. Here, in ...

  7. Vaccine uptake and barriers to vaccination among at-risk adult

    This suboptimal vaccine uptake facilitates the continued burden of VPDs and therefore reinforces the importance of life-course immunization. This literature review identified several key barriers to vaccination, including concerns regarding perceived vaccine safety and effectiveness, cost of vaccination, and lack of insurance and provider ...

  8. Influences and policies that affect immunisation coverage-a summary

    Here we summarise a recent literature review that was conducted to identify and examine key factors that influence immunisation coverage, timeliness of vaccinations and the core drivers that affect vaccine uptake. We conclude that well designed systems, taking a holistic approach with multiple components, gain broader community acceptance and ...

  9. Full article: Factors affecting childhood immunization: Thematic

    Themes were then identified using thematic analysis. A total of 44 studies met the review criteria and were summarized and categorized into 4 themes: barriers to immunization, parental knowledge, attitude and behavior (KAB), health system factors and HCWs' KAB. This review found that immunization decision-making is a complex process.

  10. Advancing sustainable development goals through immunization: a

    Methods: We conduct a literature review through a keyword search strategy complemented with handpicking and snowballing from earlier reviews. After title and abstract screening, we conducted a qualitative analysis of key insights and categorized them according to showing the impact of immunization on SDGs, sustainability challenges, and model ...

  11. A systematic literature review to clarify the concept of vaccine

    Abstract. Vaccine hesitancy (VH) is considered a top-10 global health threat. The concept of VH has been described and applied inconsistently. This systematic review aims to clarify VH by ...

  12. Advancing sustainable development goals through immunization: a

    The challenges that came out of the literature review correspond with the Immunization Agenda 2030's strategic priorities and core values. Under-addressed challenges include public-private partnerships, the role of sustainable innovation, cross-sectoral collaboration, and service coordination.

  13. PDF Systematic literature review of the impact and effectiveness of

    Systematic literature review of the impact and effectiveness of monovalent meningococcal C conjugated vaccines when used in routine immunization programs Myint Tin Tin Htar1*, Sally Jackson2, Paul Balmer3, Lidia Cristina Serra3, Andrew Vyse4, Mary Slack5, Margarita Riera-Montes2, David L. Swerdlow3 and Jamie Findlow1 Abstract

  14. Factors influencing childhood immunisation uptake in Africa: a

    The majority of the literature cited in this review is observational in nature and therefore, this study is lacking evidence for causation. Furthermore, many studies relied on survey data which may increase the risk of nonresponse bias or selection bias. ... Juni M. Adherence among parents on under-five childhood immunization schedule: a review ...

  15. Vaccine safety and efficacy: A literature review

    Vaccine safety and efficacy: A literature review. To introduce vaccine safety and efficacy, the two main public concerns for vaccine use. This is a review of the literature including but not limited to scientific publications and government documents that are related to vaccine safety and efficacy. The publication dates range from 1984 to 2020.

  16. COVID-19 Vaccination and the Rate of Immune and Autoimmune ...

    COVID-19 Vaccination and the Rate of Immune and Autoimmune Adverse Events Following Immunization: Insights From a Narrative Literature Review Front Immunol. 2022 Jul 5:13:872683. doi: 10.3389 ... In this narrative review, we illustrate the different vaccine types in current use, their associated immune and autoimmune AEFIs, with a focus on the ...

  17. Too little but not too late: Results of a literature review to improve

    This review builds on two similar reviews of immunization service strategies in developing countries which were published between 2004 and 2005, one of published literature and the other of gray literature [4, 5]. Although similar to this review, additional papers not identified by the previous reviews have been included as our approach was ...

  18. Comprehensive literature review on COVID-19 vaccines and role of SARS

    This literature review aims to describe the physiology of the vaccines and how the spike protein is used as a target to elicit an antibody-dependent immune response in humans. Furthermore, the overview, dosing strategies, efficacy, and side effects will be discussed for the notable vaccines: BioNTech/Pfizer, Moderna, AstraZeneca, Janssen ...

  19. PDF A literature review and proposed learning agenda on Immunisation

    Acronyms. as the HDA, Health Development Army)MenA Meningococcal A (vaccine)AcknowledgementsThis literature review is a product of the Immunisation-Nutrition Integration (INI) Partnership between Gavi, the Vaccine Alliance (Gavi) and the Eleanor Crook Foundation (ECF), which ai. ed to advance integrated immunisation-nutrition programming ...

  20. Addressing Parental Vaccine Hesitancy towards Childhood Vaccines in the

    Frew et al. conducted a systematic literature review of 34 studies that used parental reminder and recall interventions based on the use of postcards, letters, the telephone, or a combination to improve early childhood immunization uptake. Researchers found that the majority of reminder and recall systems (79%) increased the likelihood of ...

  21. Factors Affecting Completion of Childhood Immunization in North West

    Immunization is reported to prevent an estimated 2 to 3 million deaths each year worldwide (WHO, 2009). The year 2014 marked the 40th anniversary of the WHO's Expanded Program on Immunization (EPI), which was established to ensure equitable access to routine immunization (RI) services (CDC, 2015). The WHO (2009) has stated

  22. Types of Vaccines, Adverse Events Following Immunization, and COVID-19

    Literature review of Bell's palsy after all types of COVID vaccines: The observed incidence of Bell's palsy after mRNA vaccines is between 3·5-times and 7-times higher than would be expected in the general population. Sato et al. Analysis of Bell's palsy cases databases after mRNA vaccines

  23. Literature review of HPV vaccine delivery strategies ...

    This literature review will summarize the experiences from co … School-based vaccination is becoming a more widely considered method of delivering HPV immunizations to an adolescent population; however, many countries do not have experience with delivering adolescent vaccines or school-based programs. This literature review will summarize the ...