1,839
Views
5
CrossRef citations to date
0
Altmetric
Review

A systematic review of immunogenicity, clinical efficacy and safety of human papillomavirus vaccines in people living with the human immunodeficiency virus

ORCID Icon, , , &
Pages 426-435 | Received 13 May 2019, Accepted 09 Aug 2019, Published online: 20 Sep 2019

ABSTRACT

The human papillomavirus (HPV) is the most prevalent sexually transmitted infection worldwide. People living with the human immunodeficiency virus (HIV) are at high risk of HPV infection. This systematic review evaluates the immunogenicity, clinical efficacy, and safety of prophylactic HPV vaccines in people living with HIV. We registered the protocol for this review in the International Prospective Register of Systematic Reviews (CRD42018109898) and prepared the review following the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). Five randomized trials with 1042 participants are included in this review. One trial with 120 participants compared the bivalent HPV vaccine to placebo, three trials with 830 participants compared the quadrivalent vaccine to placebo, and another trial with 92 participants compared the quadrivalent to the bivalent vaccine. There was low to moderate certainty evidence suggesting that seroconversion was higher among participants in the vaccine arms compared to the placebo arms for both vaccines. In one study with very low certainty evidence, participants who received the bivalent vaccine had higher anti-HPV-18 geometric mean titers (GMTs) compared to those who received the quadrivalent vaccine, despite little difference in anti-HPV-16 GMTs between the two vaccines. There were no differences in the incident and persistent HPV infections in both groups. None of the studies reported data on the incidence of precancerous lesions, or cancer. There were no reports of serious adverse events following vaccination in any of the trials. None of the included studies assessed the effects of HPV vaccines in adolescents living with HIV. Very limited evidence suggests lower immunogenicity of HPV vaccines in HIV positive compared to HIV-negative people. Finally, the long-term effect of the HPV vaccine in the incidence of cervical precancerous lesions and cervical cancer needs to be monitored. There is an urgent need for more high-quality randomized controlled trials that can address these gaps.

Introduction

Human papillomavirus (HPV) infection is the most common sexually transmitted infection worldwide.Citation1 It is estimated that 75% of sexually active people are infected with HPV during their lifetime. Although most HPV infections are transient and asymptomatic,Citation2 persistent infection with high-risk HPV types may result in diseases.Citation3 Persistent HPV infection causes more than 600 000 cancers worldwide every year, including cervical, anal, vulvar and vaginal, penile, and certain oropharyngeal cancers.Citation4 It is also associated with other skin and mucosal lesions such as warts and benign papillomas.Citation5 Most of HPV associated morbidity and mortality is due to cervical cancer,Citation6 the fourth most common cancer in women worldwide, with an estimated 569,847 cases and 311,365 deaths in 2018.Citation7 To date, more than 200 HPV types have been identified and classified into two groups: high-risk and low-risk types.Citation8 High-risk HPV types, including HPV-16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58 and −59 are associated with cancers in humans,Citation9 while low-risk HPV types, including HPV-6, 11, 40, 42, 43, 44, 54, 61 and −72 cause benign diseases such as genital warts.Citation10 Among these HPV types, majority of HPV-related clinical diseases are associated with HPV-16, 18, 6 and −11. HPV types 16 and 18 cause approximately 70% of cervical cancer, HPV-6 and HPV-11 are responsible for approximately 90% of genital warts in both men and women.Citation11

People infected with human immunodeficiency virus (HIV) are at high risk of HPV infection and developing HPV-associated cancers.Citation12 HPV infections are also more persistent in people living with HIV, which increases their risk of developing HPV-related cancers.Citation13 This is because the state of immunosuppression induced by HIV infection impairs the immune systems’ ability to clear HPV infection.Citation14 HPV-associated cancers occur at higher rates in HIV-positive people compared with the general population.Citation15

Vaccination is one of the most effective public health interventions for combating infectious diseases.Citation16 It is estimated that vaccines save approximately 2–3 million lives worldwide each year.Citation17 Currently, there are three prophylactic HPV vaccines used across the world: Cervarix, a bivalent HPV vaccine that targets HPV-16 and −18; Gardasil, a quadrivalent HPV vaccine that targets HPV-6, 11, 16, and −18; and Gardasil 9, a nonavalent HPV vaccine that targets HPV-6, 11, 16, 18, 31, 33, 45, 52, and −58.Citation18 All three vaccines are composed of non-infectious L1 protein subunits assembled into virus-like particles. They prevent HPV infections caused by targeted types by eliciting the production of neutralizing antibodies that bind to the viral particles and block their entrance into host cells.Citation19-Citation21 These vaccines have shown a high degree of safety, immunogenicity, and efficacy in HIV-negative individuals.Citation22,Citation23 However, little is known about their effects on people living with HIV. In this review, we evaluate the immunogenicity, clinical efficacy, and safety of prophylactic HPV vaccines in people living with HIV.

Methods

We registered the protocol for this review in the International Prospective Register of Systematic Reviews (PROSPERO),Citation24 and prepared the review following the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA).Citation25

Criteria for considering studies for this review

We included randomized controlled trials conducted among HIV-positive people irrespective of their setting, age, sex, HIV stage, and antiretroviral therapy status. Eligible trials compared prophylactic HPV vaccines to placebo or any other vaccine, irrespective of the number of doses administered, vaccination schedule used, and the site of vaccine administration. Finally, eligible studies should have reported a least one of our outcomes of interest. Our primary outcomes included immunogenicity (measured using percentage of participants who seroconverted or mean antibody levels) and adverse events observed after HPV vaccine administration. Our secondary outcomes included incident and persistent infection with both vaccine and non-vaccine HPV types, cervical intraepithelial neoplasia, and invasive cervical cancer.

Search and selection of studies

We developed a comprehensive search strategy for searching electronic databases and other resources. On 10 October 2018, we searched the Cochrane Central Register of Controlled Trials (CENTRAL), PubMed, EMBASE, WHO International Clinical Trials Registry Platform, clinicaltrials.gov, and abstract databases of the International AIDS Society and the Conference on Retroviruses and opportunistic infections, for articles indexed from 2000 to the date of the search; with no language restrictions. We have provided the search strategy for one database, PubMed, in . We also conducted hand searches of the reference lists of included studies and related reviews. We exported all studies retrieved from the electronic searches into EndNote for deduplication and screening. Two review authors (EM and AW) independently screened the titles and abstracts to identify potentially eligible studies. Disagreements between the two authors were resolved by discussion and consensus. We obtained the full-texts of all potentially eligible studies. Two authors (EM and AW) independently screened the full texts and identified included studies, resolving discrepancies through discussion and consensus. Excluded studies are described in the table of excluded studies alongside their reasons for exclusion.

Table 1. PubMed search strategy.

Data extraction and management

Two review authors (EM and AW) independently extracted data from each selected study using a structured and standardized data extraction form. Extracted data included study details (geographical locations, number of participants), intervention details (number of participants, type of vaccine, number of doses), comparator details (number of participants, type of comparator used), outcome details and funding sources. Differences between the two review authors were resolved by discussion and consensus.

Assessment of risk of bias in included studies

Two review authors (EM and AW) independently assessed the risk of bias in each included study using the Cochrane risk of bias tool for randomized controlled trialsCitation26 The domains assessed include: random sequence generation, allocation concealment, blinding of participants, blinding of outcome assessors, completeness of outcome data, completeness of outcome reporting, and other sources of bias. Disagreements between the two authors were resolved by discussion and consensus. We had planned to asses for publication bias using a funnel plot, but this was not done due to the few number of included studies.

Data analysis

Data were entered into Review Manager 5.3 (RevMan 5.3) software and checked for accuracy. We calculated risk ratios with the 95% confidence intervals for dichotomous data such as seroconversion, and the mean difference for continuous data such as antibody levels. When the authors reported means and 95% confidence intervals, we estimated the standard deviations using the sample size, and the upper and lower limits of the confidence intervals. Due to important difference in the study populations, interventions and outcome measures, we did not conduct a meta-analysis of the included studies but rather narratively synthesized the evidence. Finally, we assessed the quality of the evidence using the GRADE approach.Citation27

Results

Results of the search

The search yielded 504 records. After removing 89 duplicates, 415 titles and abstracts were screened and 389 of them were not relevant. The full texts of 26 potential eligible publications were reviewed. Seventeen publications reporting data on five randomized trials were included.Citation4,Citation28-Citation43 The search and selection of studies for this review are described in .

Figure 1. PRISMA flow diagram showing the study search and selection process.

Figure 1. PRISMA flow diagram showing the study search and selection process.

Description of studies

Study design

All the included studies were randomized trails. The characteristics of the five included studiesCitation4,Citation28,Citation31,Citation34,Citation39 are summarized in .

Table 2. Characteristics of included studies.

Population

The studies were conducted among people living with HIV including: women aged 18–25 years,Citation28 adult men and women aged ≥18 years,Citation4 men who have sex with men (MSM) aged ≥18 years,Citation31 MSM and women aged ≥27 years,Citation39 and male and female children aged 7–12 years.Citation34 The study size ranged between 92 and 575 participants. Two trials were conducted in the United States of America (USA)Citation34,Citation39 and the other three were carried out in South Africa,Citation28 DenmarkCitation4, and SpainCitation31 between 2010 and 2018.

Intervention and comparator

Three trials compared bivalent HPV vaccine to placebo.Citation31,Citation34,Citation39 One compared quadrivalent HPV vaccine to bivalent vaccine,Citation4 and the other one compared bivalent vaccine to placebo.Citation28 In the study that compared the bivalent vaccine to placebo in asymptomatic HIV-positive women, the efficacy and safety of the vaccine were also assessed in a third arm comprising of HIV-negative women. In all five studies, three doses of the vaccines and placebos were administered in the respective intervention and comparator arms. In three trials, intervention vaccines were given at day 0, month 2 and month 6.Citation31,Citation34,Citation39 In the other two trials, intervention vaccines were given at day 0, month 1 and month 2Citation28 and at day 0, month 1.5 and month 6.Citation4

Outcome measures

Primary outcomes

All five studies reported on immunogenicity and adverse events. The immunogenicity outcomes of these five included studies are summarized in . HPV specific antibodies were measured using enzyme-linked immunosorbent assay (ELISA),Citation28,Citation31 Luminex immunoassay,Citation34,Citation39 and pseudovirion-based neutralization assay (PBNA).Citation4

Table 3. The immunogenicity outcome of included studies.

Secondary outcomes

One study reported on incidence and persistence of HPV infection.Citation4 None of the included studies reported on cervical intraepithelial neoplasia, and invasive cervical cancer as the duration of the trials too short for the development of cervical intraepithelial neoplasia and invasive cervical cancer.

Excluded studies

Nine studiesCitation8,Citation13,Citation22,Citation44-Citation49 were excluded for reasons described in the characteristics of excluded studies ().

Table 4. Characteristics of excluded studies.

Risk of bias in included studies

The risk of bias in the included studies is summarized in . We assessed for selection bias in the included studies. All five studies described the methods used to generate the randomization sequence adequately and were judged as having low risk of bias. Three trials were at low risk of bias related to allocation concealment.Citation4,Citation28,Citation31 The other two trials were judged to have unclear risk of bias because the authors did not provide sufficient information regarding the methods used to conceal allocation in the intervention and comparison groups.Citation34,Citation39 Three trials were at low risk of performance bias because participants and personnel were blinded.Citation31,Citation34,Citation39 The other two trials did not describe the blinding of participants and personnel clearly, thus we assessed them as having unclear risk of performance bias.Citation4,Citation28 One trial was considered to be at low risk of detection bias because the outcome assessors were blinded.Citation31 The other four trials did not describe the blinding of outcome assessors and we considered them to be having unclear risk of detection bias.Citation4,Citation28,Citation34,Citation39 We judged all the included trials to be at low risk of bias in relation to the completeness of outcome data. One trial was judged to be at low risk of reporting bias,Citation28 while the other four trials were judged to have an unclear risk of bias.Citation4,Citation31,Citation34,Citation39

Table 5. Risk of bias summary.

Effects of HPV vaccines

Comparison of bivalent HPV vaccine to placebo

Seroconversion

One trial that compared bivalent HPV vaccine to placebo reported on seroconversion.Citation28 The study was conducted in Cape Town, South Africa among HIV-positive and HIV-negative women aged 18–25 years. At baseline, 85.4% and 73.0% were seropositive for HPV 16 while 64.3% and 56.8% were seropositive HPV-18 in the vaccine and placebo groups that included HIV-positive women, respectively. A third arm, consisting of HIV-negative women who also received the vaccine had baseline seroconversion rates of 63.6% and 50.0% for HPV 16 and 18, respectively. All HIV positive and negative participants who received the vaccine seroconverted for HPV-16 and HPV-18 1 month after the second dose and remained seropositive for both antigens 6 months after the third dose. Anti-HPV 16 and 18 antibody GMTs did not increase in the placebo group. However, anti-HPV-16 and 18 antibody GMTs increased in the vaccine groups. There were significant differences in anti-HPV-16 GMTs between HIV positive and HIV-negative participants at 1 month (MD −4610.60 (95% CI: −6791.06; −2430.14)) and at 6 months (MD-2045.50 (95% CI: −2868.51; −1222.49)) after the third dose, with higher GMTs in HIV-negative women. There were equally differences in the anti-HPV-18 antibody GMTs between the HIV positive and HIV-negative participants at 1 month (MD −1757.20 (95% CI: −3268.07; −246.33)) and at 6 months (MD −678.20 (95% CI: −1182.29;-174.11)) after the third dose.

Adverse events

HIV positive and negative women who received the bivalent vaccine had a higher incidence of solicited local and general adverse events than HIV-positive women who received placebo, with most of them being mild and moderate, resolving spontaneously.Citation28 There was severe pain and swelling in 1.9% and 0.6% of the doses given to HIV-positive women who got the HPV vaccine and in 1.2% and 5.9% of the doses given to HIV-negative women who got the vaccine. No severe local adverse events were reported in the placebo group. Unsolicited adverse events were reported by 86.9% of HIV-positive women in the vaccine group and 86.7% of HIV-negative women in the vaccine group and 78.0% of HIV-positive women in the placebo group. The most commonly reported unsolicited adverse events were headache (19.7% and 23.7%, vaccine and placebo group in HIV-positive women and 13.3% in HIV-negative women in the vaccine arm, respectively) and upper respiratory tract infection (16.4%, and 16.9% in vaccine and placebo group, and 23.3% in HIV-negative women in the vaccine arm respectively, respectively). Medically significant adverse events were recorded by 11.1% and 9.6% of HIV-positive women in the vaccine group and placebo group, and 16.7% in HIV-negative women in the vaccine arm, respectively. There were six women with serious adverse events, none of them vaccine-related.

Comparison of quadrivalent HPV vaccine to placebo

Seroconversion

Although all three trials that compared quadrivalent HPV vaccine to placebo reported on this outcome, there was heterogeneity between these trials in the reporting of the outcome measures, hence the reported studies could not be meta-analyzed.Citation31,Citation34,Citation39 Hidalgo 2017Citation31 assessed seroconversion in 129 HIV-positive Spanish men who have sex with men. Although baseline anti-HPV antibodies are unknown, there were significantly more people who seroconverted in the vaccine group compared to the placebo group 1 month after the third dose (RR 2.51) (95% CI: 1.68; 3.75). Levin 2010Citation34 evaluated seroconversion in HIV-positive children aged 7 to 12 years. All vaccinated participants seroconverted to HPV-6, HPV-11, and HPV-16 antigens, except for HPV-18, where only 96% seroconverted 4 weeks after the third dose. In the placebo group, there was no participant who seroconverted to HPV-6, HPV-11, and HPV 18, except for HPV-16 where only 4% seroconverted. The antibody GMTs were significantly higher in the intervention group compared to the placebo group for HPV-6 (MD 548.00) (95% CI: 398.14; 697.86), HPV-11 (MD 1367.00) (95% CI: 1088.83; 1645.17), and HPV-16 (MD 5225.00) (95% CI: 3966.35; 6483.65) respectively. Wilkin 2018Citation39 assessed seroconversion in HIV-positive adults aged 27 years or older. One month after the third dose, seroconversion in the vaccine group was higher than in the placebo group; 98.9%, 100%, 99.6%, and 97.4% for HPV-6, HPV-11, HPV-16, and HPV-18, respectively, compared to corresponding 64%, 45%, 47%, and 31% at baseline. At baseline, seroconversion in the placebo group was 61%, 38%, 47%, and 33% for HPV-6, HPV-11, HPV-16, and HPV-18, respectively, and it did not change appreciably 1 month after the third dose although the authors do not report the exact values.

Adverse events

All three trials that compared quadrivalent vaccine to placebo reported adverse events.Citation31,Citation34,Citation39 However, there was heterogeneity in the reporting of the adverse events between these trials. Due to this heterogeneity, the reported data findings could not be meta-analyzed. Hidalgo 2017Citation31 reported few adverse events, with these adverse events higher in the placebo group compared to vaccine group after the first (RR 0.62) (95% CI: 0.49;0.79), second (RR 0.91) (95% CI: 0.83;0.99), and third doses (RR 0.74) (95% CI: 0.61; 0.89), with injection-site pain being the most common adverse event. However, there were no grade 3 and grade 4 vaccine-related adverse events reported. There were also no serious adverse events related to the vaccine administration observed. Levin 2010Citation34 also found that adverse events were generally few in both the placebo group and vaccine group. There was no significant differences in grade 1 (RR 2.60) (95% CI: 0.85, 8.02), grade 2 (RR 0.91) (95% CI: 0.50, 1.64), grade 3 (RR 0.78) (95% CI: 0.16, 3.82), and grade 4 (RR 1.60) (95% CI: 0.08, 32.40) adverse events between the vaccine and placebo groups. Injection-site reaction was the most common reported adverse event. Wilkin 2018Citation39 reported that there were no grade 3, grade 4 serious adverse events related to the vaccination.

Comparison of quadrivalent HPV vaccine to bivalent HPV vaccine

Seroconversion

One trial involving 92 participants, comparing quadrivalent HPV vaccine to bivalent HPV vaccine reported on this outcome.Citation4 The trial was conducted in Denmark among HIV-positive adults. One month and 6 months after the third dose, the bivalent vaccine group had higher anti-HPV-18 GMTs compared to quadrivalent vaccine group. The absolute values of the antibody GMTs are not reported. However, the ratio of the anti-HPV-18 antibodies in the bivalent group compared to the quadrivalent group was 4.31 and 4.15 1 month and 6 months after the third dose, respectively. During the same period, there were no significant differences in anti-HPV-16 GMTs found between these two vaccines.

Adverse events

No serious adverse events were reported in this trial; both vaccines were well tolerated. However, injection-related adverse events were observed. Injection-site pain was more common in the bivalent vaccine group than in the quadrivalent vaccine group after the first (RR 0.39) (95%CI: 0.25; 0.60) and second doses (RR 0.40) (95% CI: 0.21, 0.77). Injection-site swelling was also more common in the bivalent vaccine group than in the quadrivalent vaccine group after the second dose (RR 0.22) (95% CI: 0.05; 0.95).

Discussion

Summary of main results

When the effects of HPV vaccines were compared to placebo in HIV-positive people, seroconversion rates in the vaccinated groups were higher compared to the placebo group. GMTs reported in two trials were also higher in the vaccinated group compared to the placebo group. In one trial that compared bivalent to quadrivalent vaccine, anti-HPV-18 GMTs were higher in the bivalent group compared with the quadrivalent group. However, there were no significant differences in anti-HPV-GMTs between these two vaccine groups. With regards to the safety of HPV vaccines in HIV-positive people, there were no serious vaccine-related adverse events reported in these five trials. The vaccines were generally safe and well tolerated. Injection-site reaction was the most common adverse event of HPV vaccines reported. In four trials where HPV vaccines were compared to placebo, seroconversion rates in the vaccinated groups were higher compared to the placebo group. Of these four trials, GMTs were reported only in two trials and they were higher in the vaccinated group compared to the placebo group. In one trial that compared bivalent to quadrivalent vaccine, anti-HPV-18 GMTs were in the bivalent group compared with the quadrivalent group. However, there were no significant differences in anti-HPV-GMTs between these two vaccine groups.

Overall completeness and applicability of evidence

Despite our comprehensive search, we found only five trials which met our inclusion criteria. Four out of five trials were conducted in high-income countries with a low burden of HIV/AIDS. Only one trial was conducted in South Africa, an upper middle-income country with a high burden of HIV/AIDS. Although all participants were HIV positive, they were generally healthy patients. Although all included studies administered three doses of the HPV vaccines, they differed in vaccination schedules, time taken to determine immunogenicity endpoint, and methods used to measure and interpret immunogenicity outcome. None of the included trials assessed the effect of the nonavalent HPV vaccine, one of the three recommended HPV vaccines, highlighting a gap in the evidence in this area. In addition, none of the included studies was conducted against HIV-positive adolescents, most affected group by HIV and HPV infection. We had intended to assess other outcomes such as histologically confirmed high-grade cervical intraepithelial neoplasia (CIN2, CIN3, and adenocarcinoma in situ (AIS)), and invasive cervical cancer. However, none of the included studies reported on these.

Quality of the evidence

We used the GRADE approach to assess the certainty of the evidence on the effects of HPV vaccines. The evidence on the effects of the bivalent HPV vaccine compared to placebo in people living with HIV was based on one study conducted amongst women 18–25 years old, including 120 participants. We graded the certainty of the evidence around the immunogenicity as moderate due to imprecision. The certainty of the evidence on the effects of the bivalent HPV vaccine in HIV-positive women compared to HIV-negative women was graded as low because we downgraded by two for important imprecision arising from the small study sample and the study design. The evidence around the effect of the quadrivalent HPV vaccine compared to placebo in people living with HIV was based on three studies with a highly diverse population, including MSM, Children 7–12 years and adults >27 years. We rated the certainty of evidence on the immunogenicity as moderate in adults >27 due to risk of bias, moderate in MSM due to imprecision and low in children due to imprecision and risk of bias. The evidence around the effect of the quadrivalent HPV vaccine compared to the bivalent HPV vaccine in people living with HIV was based on one study in HIV-positive adults with only 92 participants. We rated the certainty of evidence on the immunogenicity as very low due to important imprecision.

Potential biases in the review process

We minimized potential biases in the review process by adhering to the Cochrane guidelines.Citation26 We conducted comprehensive searches of both peer-reviewed and gray literature, without limiting the searches to a specific language. Two review authors independently assessed study eligibility, extracted data, and assessed the risk of bias in each included study. We are not aware of any biases in the review process.

Agreements and disagreements with other studies or reviews

We found that people living with HIV who were vaccinated with HPV vaccines had high rates of seroconversion compared to the ones who received placebo. These findings are consistent with the findings of an unpublished report of HPV vaccines in HIV infected males and females produced by Cochrane Response.Citation50 However, the latter included only four studies published up to 2016.Citation4,Citation13,Citation28,Citation34 We excluded one of the four studies from our review because it is not a randomized controlled trial.Citation13 Our review includes an additional two studies published in 2017 and 2018.Citation31,Citation39 Our findings are also in agreement with the findings previously reported in non-randomized control and excluded studies.Citation8,Citation13,Citation45-Citation47,Citation49 The similar findings of high seroconversion rates were also reported in HIV-positive adolescents boys and girls vaccinated with quadrivalent HPV vaccine.Citation51 Our review also found that HIV-positive people vaccinated with the bivalent HPV vaccine had higher anti-HPV-18 GMTs compared to those who were vaccinated with the quadrivalent HPV vaccine. With regards to anti-HPV-16, the GMTs were similar in both groups. These findings are also in consistency with the findings of an unpublished report of HPV vaccines in HIV infected males and females produced by Cochrane Response.Citation50

Authors’ conclusions

Implication for research

None of the included studies assessed the immunogenicity and safety of HPV vaccines in adolescents. There were also no included studies that assessed the effects of the nonavalent HPV vaccine in people living with HIV. There is, therefore an urgent need for more high-quality randomized controlled trials that can address these gaps. There is also limited evidence suggesting that although the HPV vaccine is immunogenic in HIV-positive people, anti-HPV GMTs are lower in HIV positive compared to HIV-negative people. This could have implications on the number of doses to be administered in this population in order to optimize the benefits of the vaccines and warrants further research.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Author’s contributions

EJM and CSW conceived the study. EJM drafted the study protocol. EJM and ABW screened the titles, abstracts and full texts, and extracted data. ABW, PWM, and CSW provided content and methodological expertise. All the authors read, amended, and approved the final version of the study protocol for submission. CSW is the guarantor for this review.

Acknowledgments

The authors would like to thank Joy Oliver at Cochrane South Africa for assisting with the search strategy.

Additional information

Funding

This review was supported by the South African Medical Research Council and the National Research Foundation of South Africa (Grant Number: 106035).

References

  • Loke AY, Kwan ML, Wong YT, Wong AKY. The uptake of human papillomavirus vaccination and its associated factors among adolescents: a systematic review. J Prim Care Commun Health. 2017;8(4):349–62. doi:10.1177/2150131917742299.
  • Lacombe-Duncan A, Newman PA, Baiden P. Human papillomavirus vaccine acceptability and decision-making among adolescent boys and parents: a meta-ethnography of qualitative studies. Vaccine. 2018;36(19):2545–58. doi:10.1016/j.vaccine.2018.02.079.
  • Bloem P, Ogbuanu I. Vaccination to prevent human papillomavirus infections: from promise to practice. PLoS Med. 2017;14(6):e1002325. doi:10.1371/journal.pmed.1002230.
  • Toft L, Storgaard M, Muller M, Sehr P, Bonde J, Tolstrup M, Ostergaard L, Sogaard OS. Comparison of the immunogenicity and reactogenicity of cervarix and gardasil human papillomavirus vaccines in HIV-infected adults: a randomized, double-blind clinical trial. J Infect Dis. 2014;209(8):1165–73. doi:10.1093/infdis/jit657.
  • Doorbar J, Egawa N, Griffin H, Kranjec C, Murakami I. Human papillomavirus molecular biology and disease association. Rev Med Virol. 2015;25(Suppl 1):2–23. doi:10.1002/rmv.1822.
  • Munoz N, Bosch FX, de Sanjose S, Herrero R, Castellsague X, Shah KV, Snijders PJ, Meijer CJ. International agency for research on cancer multicenter cervical cancer study, G., Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med. 2003;348(6):518–27. doi:10.1056/NEJMoa021641.
  • Cancer IAFRO Global cancer observatory. [accessed 2019 Jul 19]. https://gco.iarc.fr/.
  • Fontes A, Andreoli MA, Villa LL, Assone T, Gaester K, Fonseca LAM, Duarte AJ, Casseb J. High specific immune response to a bivalent anti-HPV vaccine in HIV-1-infected men in Sao Paulo, Brazil. Papillomavirus Res. 2016;2:17–20. doi:10.1016/j.pvr.2016.01.001.
  • de Sanjosé S, Brotons M, Pavón MA. The natural history of human papillomavirus infection. Best Pract Res Clin Obstet Gynaecol. 2017;47:2–13. doi:10.1016/j.bpobgyn.2017.08.015.
  • Egawa N, Doorbar J. The low-risk papillomaviruses. Virus Res. 2017;231:119–27. doi:10.1016/j.virusres.2016.12.017.
  • Bhatia N, Lynde C, Vender R, Bourcier M. Understanding genital warts: epidemiology, pathogenesis, and burden of disease of human papillomavirus. J Cutan Med Surg. 2013;17:S47–54.
  • Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet. 2007;370(9581):59–67. doi:10.1016/S0140-6736(07)61050-2.
  • Giacomet V, Penagini F, Trabattoni D, Vigano A, Rainone V, Bernazzani G, Bonardi CM, Clerici M, Bedogni G, Zuccotti GV. Safety and immunogenicity of a quadrivalent human papillomavirus vaccine in HIV-infected and HIV-negative adolescents and young adults. Vaccine. 2014;32(43):5657–61. doi:10.1016/j.vaccine.2014.08.011.
  • Palefsky J. Human papillomavirus-related disease in people with HIV. Curr Opin HIV AIDS. 2009;4(1):52–56. doi:10.1097/COH.0b013e32831a7246.
  • Toft L, Tolstrup M, Storgaard M, Ostergaard L, Sogaard OS. Vaccination against oncogenic human papillomavirus infection in HIV-infected populations: review of current status and future perspectives. Sex Health. 2014;11(6):511–23. doi:10.1071/SH14015.
  • Perlman S, Wamai RG, Bain PA, Welty T, Welty E, Ogembo JG. Knowledge and awareness of HPV vaccine and acceptability to vaccinate in sub-Saharan Africa: a systematic review. PLoS One. 2014;9(3):e90912. doi:10.1371/journal.pone.0090912.
  • Dlamini SK, Madhi SA, Muloiwa R, von Gottberg A, Moosa M-YS, Meiring ST, Wiysonge CS, Hefer E, Mulaudzi MB, Nuttall J. Guidelines for the vaccination of HIV-infected adolescents and adults in South Africa. South Afr J HIV Med. 2018;19(1):1–8. doi:10.4102/sajhivmed.v19i1.839.
  • Chabeda A, Yanez RJR, Lamprecht R, Meyers AE, Rybicki EP, Hitzeroth II. Therapeutic vaccines for high-risk HPV-associated diseases. Papillomavirus Res. 2018;5:46–58. doi:10.1016/j.pvr.2017.12.006.
  • Harper DM, Franco EL, Wheeler C, Ferris DG, Jenkins D, Schuind A, Zahaf T, Innis B, Naud P, De Carvalho NS, et al. Efficacy of a bivalent L1 virus-like particle vaccine in prevention of infection with human papillomavirus types 16 and 18 in young women: a randomised controlled trial. Lancet. 2004;364(9447):1757–65. doi:10.1016/S0140-6736(04)17398-4.
  • Joura EA, Giuliano AR, Iversen OE, Bouchard C, Mao C, Mehlsen J, Moreira ED Jr., Ngan Y, Petersen LK, Lazcano-Ponce E, et al. A 9-valent HPV vaccine against infection and intraepithelial neoplasia in women. N Engl J Med. 2015;372(8):711–23. doi:10.1056/NEJMoa1405044.
  • Villa LL, Costa RL, Petta CA, Andrade RP, Ault KA, Giuliano AR, Wheeler CM, Koutsky LA, Malm C, Lehtinen M, et al. Prophylactic quadrivalent human papillomavirus (types 6, 11, 16, and 18) L1 virus-like particle vaccine in young women: a randomised double-blind placebo-controlled multicentre phase II efficacy trial. Lancet Oncol. 2005;6(5):271–78. doi:10.1016/S1470-2045(05)70101-7.
  • McClymont E, Lee M, Raboud J, Coutlée F, Walmsley S, Lipsky N, Loutfy M, Trottier S, Smaill F, Klein M. The efficacy of the quadrivalent human papillomavirus vaccine in girls and women living with human immunodeficiency virus. Clin Infect Dis. 2019;68(5):788–94. doi:10.1093/cid/ciy575.
  • Arbyn M, Xu L, Simoens C, Martin-Hirsch PP. Prophylactic vaccination against human papillomaviruses to prevent cervical cancer and its precursors. Cochrane Database Syst Rev. 2018;5:CD009069.
  • Mavundza EMP, Wiyeh A, Wiysonge C A systematic review of the immunogenicity and safety of human papillomavirus vaccines in people living with HIV. [accessed 2019 May 09]. http://www.crd.york.ac.uk/PROSPERO/display_record.php?ID=CRD42018109898.
  • Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, Clarke M, Devereaux PJ, Kleijnen J, Moher D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Med. 2009;6(7):e1000100. doi:10.1371/journal.pmed.1000100.
  • Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0. The Cochrane Collaboration; 2011. (updated March 2011).
  • Balshem H, Helfand M, Schunemann HJ, Oxman AD, Kunz R, Brozek J, Vist GE, Falck-Ytter Y, Meerpohl J, Norris S, et al. GRADE guidelines: 3. Rating the quality of evidence. J Clin Epidemiol. 2011;64(4):401–06. doi:10.1016/j.jclinepi.2010.07.015.
  • Denny L, Hendricks B, Gordon C, Thomas F, Hezareh M, Dobbelaere K, Durand C, Herve C, Descamps D. Safety and immunogenicity of the HPV-16/18 AS04-adjuvanted vaccine in HIV-positive women in South Africa: a partially-blind randomised placebo-controlled study. Vaccine. 2013;31(48):5745–53. doi:10.1016/j.vaccine.2013.09.032.
  • Denny LA, Hendricks B, Gordon C, Hezareh M, Dobbelaere K, David MP, Thomas F. Safety and immunogenicity of the bi-valent vaccine in HIV-positive women in South Africa. Int J Gynecol Cancer. 2012;22:E590.
  • Denny L, Hendricks B, Gordon C, Hervé C, Thomas F, Hezareh M, Dobbelaere K, Durand C, Struyf F. Safety and immunogenicity of the HPV-16/18 as04-adjuvanted vaccine in HIV-positive women in South Africa up to 12 months after vaccination. Int J Gynecol Obstet. 2012;119:S323–S324. doi:10.1016/S0020-7292(12)60610-9.
  • Hidalgo-Tenorio C, Ramirez-Taboada J, Gil-Anguita C, Esquivias J, Omar-Mohamed-Balgahata M, SamPedro A, Lopez-Ruz M, Pasquau J. Safety and immunogenicity of the quadrivalent human papillomavirus (qHPV) vaccine in HIV-positive Spanish men who have sex with men (MSM). AIDS Res Ther. 2017;14:34. doi:10.1186/s12981-017-0160-0.
  • Weinberg A, Song LY, Saah A, Brown M, Moscicki AB, Meyer WA 3rd, Bryan J, Levin MJ. Humoral, mucosal, and cell-mediated immunity against vaccine and nonvaccine genotypes after administration of quadrivalent human papillomavirus vaccine to HIV-infected children. J Infect Dis. 2012;206(8):1309–18. doi:10.1093/infdis/jis489.
  • Weinberg A, Huang S, Moscicki AB, Saah A, Levin MJ. Persistence of memory B-cell and T-cell responses to the quadrivalent HPV vaccine in HIV-infected children. AIDS. 2018;32(7):851–60. doi:10.1097/QAD.0000000000001773.
  • Levin MJ, Moscicki AB, Song LY, Fenton T, Meyer WA 3rd, Read JS, Handelsman EL, Nowak B, Sattler CA, Saah A, et al. Safety and immunogenicity of a quadrivalent human papillomavirus (types 6, 11, 16, and 18) vaccine in HIV-infected children 7 to 12 years old. J Acquir Immune Defic Syndr. 2010;55(2):197–204. doi:10.1097/QAI.0b013e3181de8d26.
  • Levin MJ, Huang S, Moscicki AB, Song LY, Read JS, Meyer WA, Saah AJ, Richardson K, Weinberg A. Four-year persistence of type-specific immunity after quadrivalent human papillomavirus vaccination in HIV-infected children: effect of a fourth dose of vaccine. Vaccine. 2017;35(13):1712–20. doi:10.1016/j.vaccine.2017.02.021.
  • Toft L, Tolstrup M, Müller M, Sehr P, Bonde J, Storgaard M, Østergaard L, Søgaard OS. Comparison of the immunogenicity of Cervarix® and Gardasil® human papillomavirus vaccines for oncogenic non-vaccine serotypes HPV-31, HPV-33, and HPV-45 in HIV-infected adults. Hum Vaccines Immunother. 2014;10(5):1147–54. doi:10.4161/hv.27925.
  • Toft L, Storgaard M, Müller M, Sehr P, Bonde J, Tolstrup M, Østergaard L, Søgaard OS. Immunogenicity and reactogenicity of cervarix ® versus gardasil® in HIV-infected adults: an RCT. Top Antivir Med. 2014;22:174–75.
  • Faust H, Toft L, Sehr P, Muller M, Bonde J, Forslund O, Ostergaard L, Tolstrup M, Dillner J. Human Papillomavirus neutralizing and cross-reactive antibodies induced in HIV-positive subjects after vaccination with quadrivalent and bivalent HPV vaccines. Vaccine. 2016;34(13):1559–65. doi:10.1016/j.vaccine.2016.02.019.
  • Wilkin TJ, Chen H, Cespedes MS, Leon-Cruz JT, Godfrey C, Chiao EY, Bastow B, Webster-Cyriaque J, Feng Q, Dragavon J, et al. A randomized, placebo-controlled trial of the quadrivalent human papillomavirus vaccine in human immunodeficiency virus-infected adults aged 27 years or older: AIDS clinical trials group protocol A5298. Clin Infect Dis. 2018;67(9):1339–46. doi:10.1093/cid/ciy274.
  • Wilkin TJ, Chen H, Cespedes M, Paczuski P, Godfrey C, Chiao E, Luque A, Webster-Cyriaque JY, Bastow B, Cranston R. ACTG A5298: a phase 3 trial of the quadrivalent hpv vaccine in older HIV+ adults. Top Antivir Med. 2016;24:65–66.
  • Cranston RD, Cespedes MS, Paczuski P, Yang M, Coombs RW, Dragavon J, Saah A, Godfrey C, Webster-Cyriaque JY, Chiao EY, et al. High baseline anal human papillomavirus and abnormal anal cytology in a phase 3 trial of the quadrivalent human papillomavirus vaccine in human immunodeficiency virus-infected individuals older than 26 years: ACTG 5298. Sex Transm Dis. 2018;45(4):266–71. doi:10.1097/OLQ.0000000000000745.
  • Cranston R, Yang M, Paczuski P, Cespedes M, Chiao E, Webster-Cyriaque J, Godfrey C, Wilkin T. Baseline data of a phase 3 trial of the quadrivalent hpv vaccine in HIV+ males and females: ACTG 5298. Top Antivir Med. 2014;22:364.
  • Zurek Munk-Madsen M, Toft L, Kube T, Richter R, Ostergaard L, Søgaard OS, Tolstrup M, Kaufmann AM. Cellular immunogenicity of human papillomavirus vaccines Cervarix and Gardasil in adults with HIV infection. Hum Vaccin Immunother. 2018;14(4):909–16. doi:10.1080/21645515.2017.1407896.
  • Anderson JS, Hoy J, Hillman R, Barnden M, Eu B, McKenzie A, Gittleson C. A randomized, placebo-controlled, dose-escalation study to determine the safety, tolerability, and immunogenicity of an HPV-16 therapeutic vaccine in HIV-positive participants with oncogenic HPV infection of the anus. J Acquir Immune Defic Syndr. 2009;52(3):371–81. doi:10.1097/QAI.0b013e3181b7354c.
  • Kahn JA, Xu J, Kapogiannis BG, Rudy B, Gonin R, Liu N, Wilson CM, Worrell C, Squires KE. Immunogenicity and safety of the human papillomavirus 6, 11, 16, 18 vaccine in HIV-infected young women. Clin Infect Dis. 2013;57(5):735–44. doi:10.1093/cid/cit319.
  • Kojic EM, Kang M, Cespedes MS, Umbleja T, Godfrey C, Allen RT, Firnhaber C, Grinsztejn B, Palefsky JM, Webster-Cyriaque JY, et al. Immunogenicity and safety of the quadrivalent human papillomavirus vaccine in HIV-1-infected women. Clin Infect Dis. 2014;59(1):127–35. doi:10.1093/cid/ciu238.
  • Money DM, Moses E, Blitz S, Vandriel SM, Lipsky N, Walmsley SL, Loutfy M, Trottier S, Smaill F, Yudin MH, et al. HIV viral suppression results in higher antibody responses in HIV-positive women vaccinated with the quadrivalent human papillomavirus vaccine. Vaccine. 2016;34(40):4799–806. doi:10.1016/j.vaccine.2016.08.016.
  • Palefsky JM, Berry JM, Jay N, Krogstad M, Da Costa M, Darragh TM, Lee JY. A trial of SGN-00101 (HspE7) to treat high-grade anal intraepithelial neoplasia in HIV-positive individuals. AIDS. 2006;20(8):1151–55. doi:10.1097/01.aids.0000226955.02719.26.
  • Wilkin T, Lee JY, Lensing SY, Stier EA, Goldstone SE, Berry JM, Jay N, Aboulafia D, Cohn DL, Einstein MH, et al. Safety and immunogenicity of the quadrivalent human papillomavirus vaccine in HIV-1-infected men. J Infect Dis. 2010;202(8):1246–53. doi:10.1086/656320.
  • Response C Randomized controlled trials of human papillomavirus vaccines: systematic reviews. [accessed 2019 May 09]. https://www.who.int/immunization/sage/meetings/2016/october/04_Clinical_trials_of_HPV_vaccines.pdf.
  • Mugo NR, Eckert L, Magaret AS, Cheng A, Mwaniki L, Ngure K, Celum C, Baeten JM, Galloway DA, Wamalwa D, et al. Quadrivalent HPV vaccine in HIV-1-infected early adolescent girls and boys in Kenya: month 7 and 12 post vaccine immunogenicity and correlation with immune status. Vaccine. 2018;36(46):7025–32. doi:10.1016/j.vaccine.2018.09.059.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.