Skip to main content
Erschienen in: Current HIV/AIDS Reports 6/2023

Open Access 05.12.2023

Awareness of Heightened Sexual and Behavioral Vulnerability as a Trigger for PrEP Resumption Among Adolescent Girls and Young Women in East and Southern Africa

verfasst von: Krishnaveni Reddy, Thesla Palanee-Phillips, Renee Heffron

Erschienen in: Current HIV/AIDS Reports | Ausgabe 6/2023

Abstract

Purpose of Review

East and Southern Africa are the epicenter of the HIV epidemic. High HIV incidence rates among adolescent girls and young women (AGYW) remain stable over the last decade despite access to daily oral PrEP. Some settings have experienced high PrEP uptake among AGYW; however, discontinuation has been high. This review sought to understand drivers of PrEP discontinuation in this population in order to identify potential mechanisms to facilitate PrEP restart and optimize PrEP use.

Recent Findings

Drivers of PrEP discontinuation included low perceived HIV acquisition risk, PrEP-associated side effects, pill burden, family/sexual partner disapproval, lack of/intermittent sexual activity, PrEP use stigma, fear of intimate partner violence, misinformation about long-term PrEP use, and limited/inconsistent access to PrEP.

Summary

The most frequently reported driver of PrEP discontinuation was low perceived HIV acquisition risk. This indicates that innovative interventions to help AGYW recognize their HIV risk and make informed decisions about PrEP use are urgently needed.
Hinweise

Publisher's Note

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

Introduction

East and Southern Africa (ESA) remain the epicenter of the HIV epidemic with 20.7 million people living with HIV (representing 55% of the number of people living with HIV worldwide even though this region accounts for < 8% of the world’s population) [13]. ESA includes 15 of the top 28 countries with HIV infections globally, 8 of which (Botswana, Eswatini, Lesotho, Mozambique, Namibia, South Africa, Zambia, and Zimbabwe) have some of the world’s highest HIV prevalence rates, ranging from 11% to over 26% among adults [1]. Cisgender heterosexual women comprise one of the most affected populations in this region with adult women comprising 3 in 5 new HIV infections in the region in 2020 and adolescent girls being 2.6 times more likely to acquire HIV than their male peers [1].
These high rates of new HIV infection have continued to occur despite the substantial investment that has been made in ESA over the past decade to scale-up education and access to HIV testing, implement universal testing and treatment of HIV, and roll out oral pre-exposure prophylaxis (PrEP) as an efficacious user-controlled HIV prevention strategy [4, 5]. Daily oral PrEP consisting of tenofovir disoproxil fumarate/emtricitabine (TDF/FTC) in a single fixed-dose combination pill has been the flagship PrEP regimen [6]. It was approved by the US Food and Drug Administration (FDA) for HIV prevention by uninfected individuals in July 2012 [7] and was recommended by the World Health Organization (WHO) to be offered to people at substantial risk of HIV infection as part of comprehensive prevention in September 2015 [8]. South Africa became the first country in Africa to approve oral PrEP with its drug regulatory authority providing approval in November 2015 [9] and followed closely by Kenya in December 2015 [10]. Subsequently, drug-regulatory authorities in ten other African countries have approved a formal indication for TDF-based formulations as HIV PrEP, and national policies in nine countries in Africa have incorporated PrEP as part of a prevention strategy [11].
Since 2012, additional PrEP products have advanced significantly through stages of product development and testing. Beginning in July 2020, regulatory authorities, including the European Medicines Agency (EMA) and the World Health Organization, issued approvals for the dapivirine vaginal ring, a monthly-replaced silicone matrix ring containing the novel antiretroviral dapivirine, as an additional prevention option for adult cisgender women at substantial risk of HIV infection [12, 13]. Reviews and approvals by national bodies in South Africa, Zimbabwe, Kenya, and others have taken place since 2021 [14, 15]. Unlike oral PrEP though, the dapivirine vaginal ring is only approved for women ≥ 18 years at this time. In 2020, the first efficacy data for a 2-monthly cabotegravir injectable suspension relative to daily oral TDF/FTC were announced with superiority demonstrated across populations [16, 17]. Regulatory approvals in the USA followed in late 2021 for use as HIV PrEP in at-risk adults and adolescents weighing at least 35 kgs [18]. Zimbabwe was the first country in Africa to approve injectable cabotegravir as PrEP [19] followed by South Africa and others [20]. Development and testing of other novel longer-acting agents are in progress, including some that aim to bundle prevention of pregnancy or other sexually transmitted infections (STIs), alongside their planned HIV prevention benefit [6].
As the HIV prevention field advances to develop new products and increase HIV prevention coverage, understanding drivers of discontinuation is paramount to guide product developers and local providers to be more attentive to end-user preferences and barriers to use of available effective modalities and to identify mechanisms that may prompt PrEP restart and optimize PrEP use.

PrEP Use Among Adolescent Girls and Young Women (AGYW) in ESA

AGYW are disproportionately impacted by the HIV epidemic, having several behavioral, biological, and socioeconomic characteristics that cause them to be vulnerable to contracting HIV and contribute to their high HIV incidence rates [21]. These include but are not limited to the following: early sexual debut, engaging in age-disparate relationships that are associated with inconsistent condom use and transactional sex, financial insecurity and gender inequalities that limit their agency to negotiate sex and condom use in order to maintain relationship security and avoid violence from their sexual partners, and high prevalence of other STIs which predispose them to a higher risk of HIV acquisition [2224]. They are also biologically more susceptible to HIV than young men due to the comparatively larger surface area of the cervix and vaginal mucosa and differences in the mucosal immunology [25].
Since 2015 when oral PrEP was approved in these regions [911], several oral PrEP demonstration projects and research studies have targeted AGYW (Table 1). Synthesis of this work in research clinics and STI/HIV clinics shows that AGYW have been willing to start using PrEP with an initial prescription and bottle of pills (estimates ranging from 55 to 100% with highest uptake seeming to occur in programs offering PrEP initiation in safe space settings). One exception is the public family planning clinic setting in Kenya that hosted PrIYA where only 22% of 1271 women screened initiated PrEP [26]. In that program, a substantial proportion of women had partners of unknown HIV status and felt they needed to consult their male partners before they could consider PrEP; however, uptake among women whose partner was known to have HIV was 94% demonstrating that women with known exposure recognized the benefits of PrEP and were willing to use it. When asked about factors motivating uptake of oral PrEP, AGYW responses have included the following: increased autonomy over their sexual health independent of sexual partners’ knowledge or approval, desire for HIV protection from sexual partner/s with multiple concurrent partnerships, unknown HIV status, and/or low condom use (either to avoid hostile condom negotiations, to please a long-term partner, or due to peer pressure), to protect themselves from HIV when engaging in transactional sex or sex under the influence of alcohol or when exposed to sexual violence in their communities, and to reduce HIV-related anxiety [27, 28].
Table 1
AGYW oral PrEP uptake and persistence in oral PrEP clinical trials and demonstration projects in ESA
Clinical trials/demo projects
Location
Duration
Participant age (years)
Number enrolled
Initial oral PrEP uptake
Persistence
Adherence by drug level measurement (TFV-DP levels via DBS)
1 month
3 months
6 months
12 months
HPTN 082/HERS [33]
South Africa (Cape Town, Johannesburg), Zimbabwe (Harare)
Oct 2016–Oct 2018
16–25
451
Harare, 148
Cape Town, 141
Johannesburg, 162
(Clinical trial sites)
95%
12 months: 55%
N/A
84%
57%
31%
MPYA [34, 35]
Kenya (Thika, Kisumu)
Dec 2016–Mar 2020
18–24
348 (Adolescent friendly research clinics)
100%
 
Electronically monitored adherence declined from 65% (month 0–1) to 15% (months 22–24). Electronically monitored adherence data were available for 150 (85%) of 177 TFV-DP samples with 67% concordance seen between the 2 measures
DREAMS PrEP Program [30]
Kenya (Kisumu, Homa Bay)
Mar 2017–Dec 2017
15–24 years
1259
Kisumu, 572
Homa Bay, 687
(Safe space settings)
100%
1 month: 57%
2 months: 46%
3 months: 37%
Not done
Not done
Not done
Not done
POWER [27, 36]
Kenya (Kisumu), South Africa (Cape Town Johannesburg)
Jun 2017–Sep 2020
16–25
2550
Kisumu, 1000
(Family planning clinics)
Cape Town, 787
(Youth and primary healthcare clinics)
Johannesburg, 763)
(Youth and primary healthcare clinics)
94%
31% returned for the first refill,
20% persisted without ≥ 15 day gap in refills,
14% stopped and restarted after a gap of ≥ 15 days
Among 1156 participants eligible for 3 and 6 month visits, 193 (16.7%) specimens were selected and indicated ~ 47% took an average of ≥ 4 doses/week
DREAMS, Namibian Ministry of Health and Social Services [31]
Namibia (Khomas)
Oct 2017–Sep 2019
15–24
1994
372 Facility Model (Public health facilities)
302 Community Concierge Model (Safe space settings) and
1320 Hybrid community-clinic model (Initiation at safe spaces and referral to public facility for refills/follow-up)
100%
Facility Model
1 month: 36.8%
3 months: 26.7%
Community Concierge Model
1 month: 41.2%
3 months: 34.9%
Hybrid community-clinic model
1 month: 6.2%
3 months: 4.8%
Not done
Not done
Not done
Not done
PrIYA [26, 40]
Kenya (Kisumu)
Nov 2017–June 2018
15 to 45 (PrEP offered during antenatal/breastfeeding period)
1271 (Family Planning Clinics)
22%
Month 1: 41%,
Month 3: 24%,
Month 6: 15%
Not done
Not done
Not done
Not done
TB HIV Care [32]
South Africa
2018–2020
15–35 years or older
28,100 (Integrated health services clinics)
55%
1 month: 38%
Not done
Not done
Not done
Not done
Behaviors necessary for sustained HIV protection, such as persistence and adherence, are however frequently cited challenges for AGYW [29, 30]. Overall, PrEP persistence decreased over time across PrEP programs and studies of AGYW (Table 1). Data from the DREAMS PrEP Program in Kenya, one of the largest real world demonstration projects, reported a median PrEP persistence time of 56 days among AGYW who initiated PrEP with the proportions of AGYW who persisted in the PrEP program at 1, 2, and 3 month(s) after PrEP initiation being 57%, 46%, and 37% respectively [30]. Further to this, data collected from DREAMS PrEP service delivery in Namibia’s Khomas Region (372 (18.7%) AGYW through a facility model, 302 (15.1%) through a community model, and 1320 (66.2%) through a hybrid model) showed PrEP persistence at 1 and 3 months to be 36.8% and 26.7% in the facility model, 41.2% and 34.9% in the community model, and 6.2% and 4.8% in the hybrid model [31]. The TB HIV Care (THC) PrEP program initiated 28100 AGYW on PrEP between 2018 and 2020. The AGYW included accessed health services from THC (e.g., HIV testing, sexual and reproductive health services) and were at greater risk of HIV acquisition than AGYW more broadly, with possibility of age-disparate relationships, multiple partnerships, transactional sex, or unstable home environments and/or school attendance. Results from this program indicated that about 38% of participants remained on PrEP at 1 month. PrEP stop and restart were common with early missed visits and inconsistent, but ongoing use [32].
In studies in research clinics, persistence appeared somewhat better than public clinic settings. In the HPTN 082/HERS study in South Africa and Zimbabwe, oral PrEP uptake among young women aged 16–25 years was 95% (N = 427), and 55% had uninterrupted PrEP refills through 12 months. Of those with dried blood spots (DBS) samples to measure oral PrEP adherence, 84% had detectable tenofovir-diphosphate (TFV-DP) levels at month 3 that declined as the study progressed (57% at month 6 and 31% at month 12) [33]. The MPYA study in Kenya, determining the impact of SMS reminders on PrEP adherence, enrolled 348 AGYW aged 18–24. Adherence was measured with pharmacy refill and real-time electronic monitoring, plus tenofovir diphosphate levels in 15% of participants. Pharmacy refills steadily declined from 100% (month 0–1) to 54% (months 22–24) and average electronically monitored adherence similarly declined from 65% (month 0–1) to 15% (months 22–24) with moderately high concordance with TFV-DP levels (67%) [34, 35]. In the POWER study in Kenya and South Africa set in diverse settings (research clinic, mobile units, public family planning), 2397 AGYW (94%) initiated PrEP and only 749 (31%) had a refill at 1 month. Of AGYW who reached 6 months or more of post-PrEP initiation follow-up, there was considerable lack of persistence with 128/646 (20%) persisting with PrEP for 6 months and great variation by site (8% to 38%). Interestingly, 14% of participants with a gap in PrEP use restarted PrEP at some point during follow-up. Among a small subsample (16.7%), intracellular TFV-DP levels indicated that 19.0% had taken PrEP daily, 28.2% an average of 4–6 doses per week, 5.1% an average of 2–3 doses per week, 19.5% an average of 1 dose per week, and 28.2% had TFV-DP levels below the limit of quantification [36]. Additionally, these outcomes demonstrate that low persistence and low adherence to oral PrEP are common occurrences for AGYW which has led to descriptions of AGYW PrEP use as a journey ranging from awareness, initiation, and early use through to persistence, including PrEP pauses, restarts, and discontinuation as their need for PrEP fluctuates with changes in risk behaviors (seasons of risk) [27]. This journey indicates a need for prevention-effective adherence where users take PrEP when they are at risk of HIV acquisition then discontinue when they are no longer exposed to HIV [37, 38]. This is similar to experience in the contraceptive field where women cycle through different patterns of contraceptive use based on partnerships and sexual behavior [39]. There are limited data available on how AGYW’s needs for PrEP change over time and how they recognize when to stop and restart PrEP. It is therefore critical to understand the drivers and correlates of PrEP non-use so we may find ways to better inform efforts to trigger PrEP re-start when it is indicated. With this information, healthcare providers can support end-users to re-start PrEP when there are strong reasons to use PrEP and the protective impact of PrEP can be maximized to ultimately reduce HIV incidence in this young population.

Reasons for PrEP Non-use/Discontinuation Among AGYW in ESA

Recent literature provides insight into reasons for oral PrEP discontinuation and non-use, including low self-perceived HIV vulnerability or risk of HIV acquisition, side effects related with PrEP use, pill burden/fatigue, disapproval from family and sexual partners, lack of sexual activity, stigma associated with using PrEP, fear of intimate partner violence (IPV), misinformation about drug resistance related to long term oral PrEP use, accidental disclosure, and limited/loss of access [27, 4145] (Table 2).
Table 2
Reasons for AGYW oral PrEP non-use/discontinuation
Study
Countries
Population
Age range (years)
Reasons for PrEP non-use or discontinuation
POWER Qualitative [27]
Kenya (Kisumu), South Africa (Cape Town, Johannesburg)
AGYW (104 in-depth interviews (IDI), 6 focus group discussions (FGD, n = 33)
16–25
• Poor perception of HIV vulnerability
• Perceived side-effects
• Pill-taking burden
• PrEP stigma
• Disapproval from family and sexual partners
MPYA [46]
Kenya (Thika, Kisumu)
AGYW (50 IDI)
18–24
• Reduction in risky sexual behaviors and lowered risk perception
• PrEP fatigue
PrIYA Qualitative [41]
Kenya (Kisumu)
AGYW (93 IDI)
15–24
• Forgetfulness
• Difficulty concealing pill taking or swallowing PrEP pills due to size
• Limited access
• Side effects
• Misinformation from male partners that instilled fear about drug resistance if using PrEP on a long-term basis
• Male partners confiscating PrEP pills
• Male partners subsequently testing HIV-negative
• Lack of sexual activity /Time apart from male partners
DREAMS [43]
Kenya (Kisumu)
AGYW (549)
15–24
• Lack of perceived risk
• Relocation
SEARCH (Qualitative) [42]
Rural communities in Kenya and Uganda
Young adults (4 male FGDs, n = 32 and 4 female FGDs, n = 56; 23 IDI)
15–24
• Early side effects of PrEP use
• Ending of relationships
• Unsupportive partners or needing to hide PrEP use from partners
• Being stigmatized by friends
• No longer feeling at risk of HIV because of learning their partners’ HIV status
• Other life events, such as travel outside the community
HPTN 082/HERS (Qualitative) [44]
South Africa (Cape Town, Johannesburg), Zimbabwe (Harare)
67 AGYW (serial IDIs at 2 time points)
16–25
• HIV stigma where PrEP is mistaken for HIV treatment
• Sexual stigma where PrEP was thought to promote sexual promiscuity
• Feelings of embarrassment/humiliation about pill bottles being seen or pills heard rattling and resultant teasing
REACH/MTN-034/REACH [45]
South Africa, Zimbabwe, and Uganda
25 AGYW (serial IDIs at 3 time points)
16–21
• Side effects
• Large size of the pill making it hard to swallow
• Dislike for the taste of the pill
• Burden of remembering to take the pill daily
• Accidental disclosure through someone discovering the pill bottles, hearing the sound of pills rattling, or seeing the participant taking their pills
• Social stigma
• Community rumors
• School or work schedules conflicting with planned time to take the pills
As adolescents exist in a broader context of family, networks and society, these reasons for discontinuation can be imposed over the socioecological framework, a model that emphasizes multiple levels of influence and considers the complex interplay between individual, relationship (social, sexual, family, network), community, and environment [47] (Fig. 1).
From this review, the majority of reasons for discontinuation were related to individual and relationship factors with low perceived risk of HIV acquisition (including poor perception of HIV vulnerability and partner negative HIV status), pill burden, and side effects being frequently cited across many of the studies. Pill burden and side effects are objective challenges that are to be expected given that AGYW are usually healthy and not accustomed to taking medication for long periods or dealing with side effects for a preventive indication. Additionally, as many AGYW live separately from their partners, frequency of sex and potential HIV exposure may be intermittent and daily oral pills (and resulting side effects) for an intermittent risk may be too burdensome. These challenges can be minimized with effective counseling and management of side effects by providers and users. Perception of HIV vulnerability, however, is an individual’s subjective appraisal of the likelihood of being exposed to or acquiring HIV and addressing this factor has potential to assist AGYW recognize periods of heightened HIV acquisition vulnerability in their lives and trigger PrEP restart with sufficient time to ensure protection when necessary.

Triggering PrEP Restart Among AGYW in ESA

During adolescence, AGYW develop levels of maturity with their growing personal autonomy and are most likely the best advocates and custodians of their sexual health if they are provided with the right tools and the support they need to make informed decisions [48]. As such, AGYW may be empowered to predict and raise self-awareness of their own periods of HIV vulnerability and make decisions to consider using PrEP in ways that match their patterns of possible exposure to HIV. This should include guidance on how to practically assess their actual sexual health vulnerability versus their perceived vulnerability which would then serve as an external cue that they could be exposed to or acquire HIV and should therefore re-start PrEP.
Numerous quantitative HIV risk assessment tools have been developed and are used to identify young women who have demographic, clinical, and behavioral factors that may place them at heightened HIV vulnerability [4953]. These tools serve several purposes. In community settings, they help counsel women about their periods of HIV vulnerability while in clinical trial settings, they improve recruitment efficiency by targeting enrolment of women who are more vulnerable to HIV. They also assist in the prioritization of these populations for scale-up of new HIV prevention interventions in public health and policy settings through ongoing programs, clinics, and primary care providers [50, 52]. These risk assessment tools could also potentially be used to trigger PrEP restart among AGYW. An analysis of individual-level risk assessment tools used in PrEP demonstration and implementation projects [53] (including 8 studies in ESA with AGYW participants) confirmed that individuals usually have inaccurate and often low perceptions of their own vulnerability to HIV and that accurate vulnerability perception increases with repeated risk assessments through opportunities to reflect on and self-assess personal vulnerability. The majority of tools in the analysis were provider-led and there was a recommendation for the creation of more opportunities for discrete self-administered or combined (self and provider-led) risk assessments (online or otherwise) that are more client/participant centered and put the tool in the PrEP user’s hand for them to internalize and reflect on their own vulnerability for accurate self-awareness. For AGYW, these risk assessment tools should be designed with AGYW input so that it meets their unique needs [47]. One suggestion pertaining to language use when communicating about risk was that the terminology of “being at risk for HIV” be reframed to “vulnerable to HIV” as it may be construed as stigmatizing and act as a barrier to oral PrEP use. Per the article cited, the terminology “vulnerable to HIV” bears less of a negative connotation and includes many things that can make people vulnerable to HIV, including their environment while “risk” tends to blame behavior (as in “taking risks”), with active decisions to engage in unsafe sex, such as sexual violence or unplanned condom-less sex [54]. Further to this, there is also an initiative to reframe and refocus how HIV prevention services for women are developed with a recommendation to move from “HIV risk” terminology to a more empowering “reasons for HIV prevention” based method. Reasons for HIV prevention include personal choices, power, autonomy, and privacy and suggest positive and empowering motivations rather than the negative “risk” and “vulnerability” terms and may help women engage rather than disconnect with their potential HIV prevention needs [55].
In addition to HIV risk assessment tools, the presence of curable STIs such as Chlamydia trachomatis, Neisseria gonorrhoeae and Trichomonas vaginalis also have potential to be used as a marker of heightened HIV vulnerability as there is clear evidence that these STIs increase the risk of HIV transmission [56, 57] and signal personal exposure to a pathogen, most often through condomless sex. AGYW in ESA are subject to high STI prevalence and incidence rates (Table 3). A meta-analysis published in 2018 of over 37,000 women across 18 HIV prevention studies and three primary region/population groups (South Africa community based, Southern/Eastern Africa community-based, and Eastern Africa higher-risk) revealed high prevalence rates of Chlamydia trachomatis (estimates ranging from 2.7 to 15.1%), Neisseria gonorrhoeae (1.7 to 8.2%), and Trichomonas vaginalis (6.7–12.7%) among women aged 15–24 years regardless of region [58]. Individual studies since the review have demonstrated continued high rates. In the Kenya Girls Study, STI incidence was 11% for Chlamydia trachomatis, 1.3% for Neisseria gonorrhoeae, and 0.8% for Trichomonas vaginalis among AGYW aged 16–20 years [59]. Among participants ≤ 24 years enrolled in the ECHO trial conducted in Eswatini, Kenya, South Africa and Zambia sites, 22% and 20% had Chlamydia trachomatis at baseline and final visits and 5% and 6% had Neisseria gonorrhoeae at baseline and final visits respectively indicating STI persistence or high rates of reinfection, even in clinical trial settings with provision of treatment [60]. In the HPTN082/HERS study, STI incidence was 27.8% per year for Chlamydia trachomatis, 11.4% for Neisseria gonorrhoeae, and 6.7% for Trichomonas vaginalis [61]. The recent MTN-034/REACH study also observed alarmingly high STI prevalence and incidence among younger AGYW (aged 16–21 years) in South Africa, Uganda, and Zimbabwe with 35% testing positive for any STI and 7% having > 1 STI. In this study, 90% of AGYW diagnosed with an STI were asymptomatic [62] and STI incidence was higher among AGYW who were diagnosed with an STI at baseline, despite receiving treatment.
Table 3
Summary of STI prevalence and incidence rates in East and Southern African regions
Study
Countries
Duration
No. enrolled
Age (years)
Chlamydia
Gonorrhea
Trichomoniasis
Prevalence
Incidence
Prevalence
Incidence
Prevalence
Incidence
Meta-analysis across 18 HIV prevention studies [58]
South Africa, Southern/Eastern Africa
1993–2011
37,000
15–24
2.7 to 15.1% (summary estimate)
-
1.7 to 8.2% (summary estimate)
-
6.7–12.7% (summary estimate)
-
Kenya Girls Study [59]
Kenya (Thika)
2014–2016
400
16–20
11%
-
1.3%
-
0.8%
-
ECHO trial [60]
Eswatini, Kenya, South Africa, and Zambia
Dec 2015 to Oct 2018
4967
 ≤ 24
22%
20%
5%
6%
-
-
HPTN 082/HERS [61]
South Africa (Cape Town, Johannesburg), Zimbabwe (Harare)
Oct 2016 to Oct 2018
451
16–25
30%
27.8%
7.8%%
11.4%
6.2%
6.7%
MTN-034/REACH [62, 63]
South Africa, Uganda, and Zimbabwe
Jan 2019 to Sep 2021
247
16–21
29%
49.1%
8.5%
21.3%
4.9%
18.8%
The most widely used approach to address STIs in resource limited public healthcare settings like ESA, where laboratory diagnosis is not readily available or accessible, is syndromic management [64]. While less expensive, syndromic management has poor diagnostic accuracy compared to conventional laboratory testing resulting in many STI cases going undetected. More recent STI data show a growing incline in STI prevalence and incidence rates and highlight an urgent need for diagnostic STI management for AGYW. This will ensure targeted and timely STI treatment, inform AGYW perception for HIV and other STI vulnerability, and facilitate data-informed counseling approaches around the HIV and STI syndemics as well as their prevention, including through the use of PrEP. Anecdotal information as well as published qualitative data [65, 66] suggest that AGYW in ESA consider targeted STI testing a benefit over the syndromic management approach and are keen to participate in screening efforts in order to be aware of their STI status. To this end, a variety of point-of-care STI diagnostic tests are being developed or are available for Chlamydia, Gonorrhea, and Trichomoniasis testing with varying assay performance, costs, and resulting time [67]. These point-of-care STI tests have potential to allow individuals to be tested and treated for STIs within the same day thus reducing burden on the individual and the clinic and may be incorporated together with PrEP and contraception services to reduce stigma associated with standalone facilities. Nevertheless, persistent barriers to testing for STIs include fear of positive test result outcomes, stigmatization by parents, family members, public clinic staff and the community, and uncomfortable or embarrassing methods of specimen collection [65]. Self-testing for HIV has been a trail blazer among concepts to support and optimize frequency of testing—and an extension of self-testing to sample self-collection and eventually self-testing for STIs is promising. Uptake, and by proxy, acceptability, of HIV self-testing with either finger-prick blood-based or saliva-based kits among adolescents has been high in initiatives in ESA and has been described as having the potential to revolutionize discrete HIV testing among young people [6870]. It empowers them to choose the location and timing of the test and control disclosure around their results thus potentially reducing the stigma associated with provider-based testing and offering the convenience of testing external to a public or less private setting. Young people are also attracted by the innovative new technology and have expressed appreciation for the decision-making autonomy and control it gave them at a time of life when they were becoming more independent from their parents and more sexually active [69]. Extending the potential benefits associated with HIV self-testing to other STI would appear the next valuable step in STI management and HIV prevention. While STI self-tests are not yet available, there is potential for AGYW to self-collect vaginal swabs for STI testing based on recent studies showing high rates of acceptability and feasibility of this method due to its privacy, convenience, and time saving nature [71, 72].Self-testing options for common and manageable STIs, such as Chlamydia trachomatis, Neisseria gonorrhoeae, and Trichomonas vaginalis where AGYW perform the test themselves, would appear the next logical innovation needed to facilitate discrete and independent assessment of risk and to trigger PrEP restart.

Conclusion

AGYW are at the epicenter of the HIV epidemic in ESA and experience evolving challenges with PrEP use and continuation. We found that while reasons for PrEP discontinuation are varied in this population, a common barrier to PrEP persistence was low perceived risk of HIV acquisition. This barrier could be addressed by encouraging accurate self-awareness and recognition of HIV vulnerability, including with behavioral assessment tools and STI self-testing. Innovative and AGYW-tailored interventions, such as self-administered sexual risk assessments and discrete STI testing with effective STI management, are urgently needed. Through these approaches, AGYW can be empowered to make informed decisions about PrEP use that align with their patterns of possible exposure to HIV. Further research to identify and develop these innovations is needed to maximize the impact that available PrEP options can have on HIV incidence in AGYW from ESA.

Declarations

Ethical Approval

This article does not contain any studies with human or animal subjects performed by any of the authors.

Competing Interests

K. Reddy, Prof. Renee Heffron and Prof. Thesla Palanee-Phillips declare that they have no conflict of interest.
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/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Parker E, Judge MA, Macete E, Nhampossa T, Dorward J, Langa DC, et al. HIV infection in Eastern and Southern Africa: highest burden, largest challenges, greatest potential. South Afr J HIV Med. 2021;22(1):1237.PubMedCrossRef Parker E, Judge MA, Macete E, Nhampossa T, Dorward J, Langa DC, et al. HIV infection in Eastern and Southern Africa: highest burden, largest challenges, greatest potential. South Afr J HIV Med. 2021;22(1):1237.PubMedCrossRef
4.
Zurück zum Zitat Heffron R, Ngure K, Odoyo J, Bulya N, Tindimwebwa E, Hong T, et al. Pre-exposure prophylaxis for HIV-negative persons with partners living with HIV: uptake, use, and effectiveness in an open-label demonstration project in East Africa. Gates Open Res. 2017;1:3.PubMedCrossRef Heffron R, Ngure K, Odoyo J, Bulya N, Tindimwebwa E, Hong T, et al. Pre-exposure prophylaxis for HIV-negative persons with partners living with HIV: uptake, use, and effectiveness in an open-label demonstration project in East Africa. Gates Open Res. 2017;1:3.PubMedCrossRef
5.
Zurück zum Zitat Baeten JM, Haberer JE, Liu AY, Sista N. Preexposure prophylaxis for HIV prevention: where have we been and where are we going? J Acquir Immune Defic Syndr. 2013;63 Suppl 2(0 2):S122-9.CrossRef Baeten JM, Haberer JE, Liu AY, Sista N. Preexposure prophylaxis for HIV prevention: where have we been and where are we going? J Acquir Immune Defic Syndr. 2013;63 Suppl 2(0 2):S122-9.CrossRef
6.
Zurück zum Zitat Cambou MC, Landovitz RJ. Challenges and opportunities for preexposure prophylaxis. Top Antivir Med. 2021;29(4):399–406.PubMedPubMedCentral Cambou MC, Landovitz RJ. Challenges and opportunities for preexposure prophylaxis. Top Antivir Med. 2021;29(4):399–406.PubMedPubMedCentral
10.
Zurück zum Zitat Masyuko S, Mukui I, Njathi O, Kimani M, Oluoch P, Wamicwe J, et al. Pre-exposure prophylaxis rollout in a national public sector program: the Kenyan case study. Sex Health. 2018;15(6):578–86.PubMedPubMedCentralCrossRef Masyuko S, Mukui I, Njathi O, Kimani M, Oluoch P, Wamicwe J, et al. Pre-exposure prophylaxis rollout in a national public sector program: the Kenyan case study. Sex Health. 2018;15(6):578–86.PubMedPubMedCentralCrossRef
11.
16.
Zurück zum Zitat Landovitz RJ, Donnell D, Clement ME, Hanscom B, Cottle L, Coelho L, et al. Cabotegravir for HIV prevention in cisgender men and transgender women. N Engl J Med. 2021;385(7):595–608.PubMedPubMedCentralCrossRef Landovitz RJ, Donnell D, Clement ME, Hanscom B, Cottle L, Coelho L, et al. Cabotegravir for HIV prevention in cisgender men and transgender women. N Engl J Med. 2021;385(7):595–608.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Machado DM, de Sant’ Anna Carvalho AM, Riera R. Adolescent pre-exposure prophylaxis for HIV prevention: current perspectives. Adolesc Health Med Ther. 2017;8:137–48.PubMedPubMedCentral Machado DM, de Sant’ Anna Carvalho AM, Riera R. Adolescent pre-exposure prophylaxis for HIV prevention: current perspectives. Adolesc Health Med Ther. 2017;8:137–48.PubMedPubMedCentral
23.
Zurück zum Zitat Palanee-Phillips T, Rees HV, Heller KB, Ahmed K, Batting J, Beesham I, et al. High HIV incidence among young women in South Africa: data from a large prospective study. PLoS one. 2022;17(6):e0269317.PubMedPubMedCentralCrossRef Palanee-Phillips T, Rees HV, Heller KB, Ahmed K, Batting J, Beesham I, et al. High HIV incidence among young women in South Africa: data from a large prospective study. PLoS one. 2022;17(6):e0269317.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Lewis L, Kharsany ABM, Humphries H, Maughan-Brown B, Beckett S, Govender K, et al. HIV incidence and associated risk factors in adolescent girls and young women in South Africa: a population-based cohort study. PLoS one. 2022;17(12):e0279289.PubMedPubMedCentralCrossRef Lewis L, Kharsany ABM, Humphries H, Maughan-Brown B, Beckett S, Govender K, et al. HIV incidence and associated risk factors in adolescent girls and young women in South Africa: a population-based cohort study. PLoS one. 2022;17(12):e0279289.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Yi TJ, Shannon B, Prodger J, McKinnon L, Kaul R. Genital immunology and HIV susceptibility in young women. Am J Reprod Immunol (New York, NY : 1989). 2013;69 Suppl 1:74–9.CrossRef Yi TJ, Shannon B, Prodger J, McKinnon L, Kaul R. Genital immunology and HIV susceptibility in young women. Am J Reprod Immunol (New York, NY : 1989). 2013;69 Suppl 1:74–9.CrossRef
26.
Zurück zum Zitat Mugwanya KK, Pintye J, Kinuthia J, Abuna F, Lagat H, Begnel ER, et al. Integrating preexposure prophylaxis delivery in routine family planning clinics: a feasibility programmatic evaluation in Kenya. PLoS Med. 2019;16(9):e1002885.PubMedPubMedCentralCrossRef Mugwanya KK, Pintye J, Kinuthia J, Abuna F, Lagat H, Begnel ER, et al. Integrating preexposure prophylaxis delivery in routine family planning clinics: a feasibility programmatic evaluation in Kenya. PLoS Med. 2019;16(9):e1002885.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Rousseau E, Katz AWK, O’Rourke S, Bekker LG, Delany-Moretlwe S, Bukusi E, et al. Adolescent girls and young women’s PrEP-user journey during an implementation science study in South Africa and Kenya. PLoS one. 2021;16(10):e0258542.PubMedPubMedCentralCrossRef Rousseau E, Katz AWK, O’Rourke S, Bekker LG, Delany-Moretlwe S, Bukusi E, et al. Adolescent girls and young women’s PrEP-user journey during an implementation science study in South Africa and Kenya. PLoS one. 2021;16(10):e0258542.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Govender E, Mansoor L, MacQueen K, Abdool KQ. Secrecy, empowerment and protection: positioning PrEP in KwaZulu-Natal, South Africa. Cult Health Sex. 2017;19(11):1268–85.PubMedCrossRef Govender E, Mansoor L, MacQueen K, Abdool KQ. Secrecy, empowerment and protection: positioning PrEP in KwaZulu-Natal, South Africa. Cult Health Sex. 2017;19(11):1268–85.PubMedCrossRef
29.
Zurück zum Zitat Dunbar MS, Kripke K, Haberer J, Castor D, Dalal S, Mukoma W, et al. Understanding and measuring uptake and coverage of oral pre-exposure prophylaxis delivery among adolescent girls and young women in sub-Saharan Africa. Sex Health. 2018;15(6):513–21.PubMedPubMedCentralCrossRef Dunbar MS, Kripke K, Haberer J, Castor D, Dalal S, Mukoma W, et al. Understanding and measuring uptake and coverage of oral pre-exposure prophylaxis delivery among adolescent girls and young women in sub-Saharan Africa. Sex Health. 2018;15(6):513–21.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat de Dieu TJ, Zangeneh SZ, Appelmans E, Pasalar S, Mori K, Peng L, et al. Persistence of oral pre-exposure prophylaxis (PrEP) among adolescent girls and young women initiating PrEP for HIV prevention in Kenya. AIDS Care. 2021;33(6):712–20.CrossRef de Dieu TJ, Zangeneh SZ, Appelmans E, Pasalar S, Mori K, Peng L, et al. Persistence of oral pre-exposure prophylaxis (PrEP) among adolescent girls and young women initiating PrEP for HIV prevention in Kenya. AIDS Care. 2021;33(6):712–20.CrossRef
31.
Zurück zum Zitat Barnabee G, O’Bryan G, Ndeikemona L, Billah I, Silas L, Morgan KL, et al. Improving HIV pre-exposure prophylaxis persistence among adolescent girls and young women: insights from a mixed-methods evaluation of community, hybrid, and facility service delivery models in Namibia. Front Reprod Health. 2022;4:1048702.PubMedPubMedCentralCrossRef Barnabee G, O’Bryan G, Ndeikemona L, Billah I, Silas L, Morgan KL, et al. Improving HIV pre-exposure prophylaxis persistence among adolescent girls and young women: insights from a mixed-methods evaluation of community, hybrid, and facility service delivery models in Namibia. Front Reprod Health. 2022;4:1048702.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Rao A, Lesko C, Mhlophe H, Rucinski K, McIngana M, Pretorius A, et al. Longitudinal patterns of initiation, persistence, and cycling on preexposure prophylaxis among female sex workers and adolescent girls and young women in South Africa. AIDS. 2023;37(6):977–86.PubMedCrossRef Rao A, Lesko C, Mhlophe H, Rucinski K, McIngana M, Pretorius A, et al. Longitudinal patterns of initiation, persistence, and cycling on preexposure prophylaxis among female sex workers and adolescent girls and young women in South Africa. AIDS. 2023;37(6):977–86.PubMedCrossRef
33.
Zurück zum Zitat Celum C, Hosek S, Tsholwana M, Kassim S, Mukaka S, Dye BJ, et al. PrEP uptake, persistence, adherence, and effect of retrospective drug level feedback on PrEP adherence among young women in southern Africa: results from HPTN 082, a randomized controlled trial. PLoS Med. 2021;18(6):e1003670.PubMedPubMedCentralCrossRef Celum C, Hosek S, Tsholwana M, Kassim S, Mukaka S, Dye BJ, et al. PrEP uptake, persistence, adherence, and effect of retrospective drug level feedback on PrEP adherence among young women in southern Africa: results from HPTN 082, a randomized controlled trial. PLoS Med. 2021;18(6):e1003670.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Haberer JE, Bukusi EA, Mugo NR, Pyra M, Kiptinness C, Oware K, et al. Effect of SMS reminders on PrEP adherence in young Kenyan women (MPYA study): a randomised controlled trial. Lancet HIV. 2021;8(3):e130–7.PubMedPubMedCentralCrossRef Haberer JE, Bukusi EA, Mugo NR, Pyra M, Kiptinness C, Oware K, et al. Effect of SMS reminders on PrEP adherence in young Kenyan women (MPYA study): a randomised controlled trial. Lancet HIV. 2021;8(3):e130–7.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Haberer JE, Mugo N, Bukusi EA, Ngure K, Kiptinness C, Oware K, et al. Understanding pre-exposure prophylaxis adherence in young women in Kenya. J Acquir Immune Defic Syndr. 2022;89(3):251–60.PubMedCrossRef Haberer JE, Mugo N, Bukusi EA, Ngure K, Kiptinness C, Oware K, et al. Understanding pre-exposure prophylaxis adherence in young women in Kenya. J Acquir Immune Defic Syndr. 2022;89(3):251–60.PubMedCrossRef
36.
Zurück zum Zitat Celum CL, Bukusi EA, Bekker LG, Delany-Moretlwe S, Kidoguchi L, Omollo V, et al. PrEP use and HIV seroconversion rates in adolescent girls and young women from Kenya and South Africa: the POWER demonstration project. J Int AIDS Soc. 2022;25(7):e25962.PubMedPubMedCentralCrossRef Celum CL, Bukusi EA, Bekker LG, Delany-Moretlwe S, Kidoguchi L, Omollo V, et al. PrEP use and HIV seroconversion rates in adolescent girls and young women from Kenya and South Africa: the POWER demonstration project. J Int AIDS Soc. 2022;25(7):e25962.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Haberer JE, Bangsberg DR, Baeten JM, Curran K, Koechlin F, Amico KR, et al. Defining success with HIV pre-exposure prophylaxis: a prevention-effective adherence paradigm. AIDS. 2015;29(11):1277–85.PubMedCrossRef Haberer JE, Bangsberg DR, Baeten JM, Curran K, Koechlin F, Amico KR, et al. Defining success with HIV pre-exposure prophylaxis: a prevention-effective adherence paradigm. AIDS. 2015;29(11):1277–85.PubMedCrossRef
38.
Zurück zum Zitat Gilbert HN, Wyatt MA, Pisarski EE, Muwonge TR, Heffron R, Katabira ET, et al. PrEP discontinuation and prevention-effective adherence: experiences of PrEP users in Ugandan HIV serodiscordant couples. J Acquir Immune Defic Syndr. 2019;82(3):265–74.PubMedPubMedCentralCrossRef Gilbert HN, Wyatt MA, Pisarski EE, Muwonge TR, Heffron R, Katabira ET, et al. PrEP discontinuation and prevention-effective adherence: experiences of PrEP users in Ugandan HIV serodiscordant couples. J Acquir Immune Defic Syndr. 2019;82(3):265–74.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Myers JE, Sepkowitz KA. A pill for HIV prevention: déjà vu all over again? Clin Infect Dis : Off Publ Infect Dis Soc Am. 2013;56(11):1604–12.CrossRef Myers JE, Sepkowitz KA. A pill for HIV prevention: déjà vu all over again? Clin Infect Dis : Off Publ Infect Dis Soc Am. 2013;56(11):1604–12.CrossRef
40.
Zurück zum Zitat Rutstein SE, Smith DK, Dalal S, Baggaley RC, Cohen MS. Initiation, discontinuation, and restarting HIV pre-exposure prophylaxis: ongoing implementation strategies. Lancet HIV. 2020;7(10):e721–30.PubMedPubMedCentralCrossRef Rutstein SE, Smith DK, Dalal S, Baggaley RC, Cohen MS. Initiation, discontinuation, and restarting HIV pre-exposure prophylaxis: ongoing implementation strategies. Lancet HIV. 2020;7(10):e721–30.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Pintye J, O’Malley G, Kinuthia J, Abuna F, Escudero JN, Mugambi M, et al. Influences on early discontinuation and persistence of daily oral PrEP use among Kenyan adolescent girls and young women: a qualitative evaluation from a PrEP implementation program. J Acquir Immune Defic Syndr. 2021;86(4):e83–9.PubMedPubMedCentralCrossRef Pintye J, O’Malley G, Kinuthia J, Abuna F, Escudero JN, Mugambi M, et al. Influences on early discontinuation and persistence of daily oral PrEP use among Kenyan adolescent girls and young women: a qualitative evaluation from a PrEP implementation program. J Acquir Immune Defic Syndr. 2021;86(4):e83–9.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Camlin CS, Koss CA, Getahun M, Owino L, Itiakorit H, Akatukwasa C, et al. Understanding demand for PrEP and early experiences of PrEP use among young adults in rural Kenya and Uganda: a qualitative study. AIDS Behav. 2020;24(7):2149–62.PubMedPubMedCentralCrossRef Camlin CS, Koss CA, Getahun M, Owino L, Itiakorit H, Akatukwasa C, et al. Understanding demand for PrEP and early experiences of PrEP use among young adults in rural Kenya and Uganda: a qualitative study. AIDS Behav. 2020;24(7):2149–62.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Ohiomoba RO, Owuor PM, Orero W, Were I, Sawo F, Ezema A, et al. Pre-exposure prophylaxis (PrEP) initiation and retention among young Kenyan Women. AIDS Behav. 2022;26(7):2376–86.PubMedCrossRef Ohiomoba RO, Owuor PM, Orero W, Were I, Sawo F, Ezema A, et al. Pre-exposure prophylaxis (PrEP) initiation and retention among young Kenyan Women. AIDS Behav. 2022;26(7):2376–86.PubMedCrossRef
44.
Zurück zum Zitat Velloza J, Khoza N, Scorgie F, Chitukuta M, Mutero P, Mutiti K, et al. The influence of HIV-related stigma on PrEP disclosure and adherence among adolescent girls and young women in HPTN 082: a qualitative study. J Int AIDS Soc. 2020;23(3):e25463.PubMedPubMedCentralCrossRef Velloza J, Khoza N, Scorgie F, Chitukuta M, Mutero P, Mutiti K, et al. The influence of HIV-related stigma on PrEP disclosure and adherence among adolescent girls and young women in HPTN 082: a qualitative study. J Int AIDS Soc. 2020;23(3):e25463.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Mary Kate Shapley-Quinn ST, Destry Jensen, Thelma Tauya, Lydia Mampuru Juliane Etima, Doreen Kemigisha, Millicent Atujuna, Lydia Soto-Torres, Sherri Johnson, Nombeko Mpongo, Nomsa Mhlanga, Kenneth Ngure, Ariane van der Straten, editor Adolescent girls and young women overcoming vaginal and oral PrEP use challenges: a longitudinal qualitative study from a crossover trial of two HIV prevention products in Uganda, Zimbabwe, and South Africa. AIDS Impact 2023 12–14 June 2023; Stockholm, Sweden. Mary Kate Shapley-Quinn ST, Destry Jensen, Thelma Tauya, Lydia Mampuru Juliane Etima, Doreen Kemigisha, Millicent Atujuna, Lydia Soto-Torres, Sherri Johnson, Nombeko Mpongo, Nomsa Mhlanga, Kenneth Ngure, Ariane van der Straten, editor Adolescent girls and young women overcoming vaginal and oral PrEP use challenges: a longitudinal qualitative study from a crossover trial of two HIV prevention products in Uganda, Zimbabwe, and South Africa. AIDS Impact 2023 12–14 June 2023; Stockholm, Sweden.
46.
Zurück zum Zitat Ngure K, Thuo N, Ogello V, Kiptinness C, Kamolloh K, Burns BFO, et al. Dynamic perceived HIV risk and sexual behaviors among young women enrolled in a PrEP trial in Kenya: a qualitative study. Front Reprod Health. 2021;3:637869.PubMedCentralCrossRef Ngure K, Thuo N, Ogello V, Kiptinness C, Kamolloh K, Burns BFO, et al. Dynamic perceived HIV risk and sexual behaviors among young women enrolled in a PrEP trial in Kenya: a qualitative study. Front Reprod Health. 2021;3:637869.PubMedCentralCrossRef
47.
Zurück zum Zitat Pettifor A, Stoner M, Pike C, Bekker L-G. Adolescent lives matter: preventing HIV in adolescents. Curr Opin HIV AIDS. 2018;13:1.CrossRef Pettifor A, Stoner M, Pike C, Bekker L-G. Adolescent lives matter: preventing HIV in adolescents. Curr Opin HIV AIDS. 2018;13:1.CrossRef
48.
Zurück zum Zitat Archary M, Pettifor AE, Toska E. Adolescents and young people at the centre: global perspectives and approaches to transform HIV testing, treatment and care. J Int AIDS Soc. 2020;23(S5):e25581.PubMedPubMedCentralCrossRef Archary M, Pettifor AE, Toska E. Adolescents and young people at the centre: global perspectives and approaches to transform HIV testing, treatment and care. J Int AIDS Soc. 2020;23(S5):e25581.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Ayton SG, Pavlicova M, Tamir H, Abdool KQ. Development of a prognostic tool exploring female adolescent risk for HIV prevention and PrEP in rural South Africa, a generalised epidemic setting. Sex Transm Infect. 2020;96(1):47–54.PubMedCrossRef Ayton SG, Pavlicova M, Tamir H, Abdool KQ. Development of a prognostic tool exploring female adolescent risk for HIV prevention and PrEP in rural South Africa, a generalised epidemic setting. Sex Transm Infect. 2020;96(1):47–54.PubMedCrossRef
50.
Zurück zum Zitat Balkus JE, Brown E, Palanee T, Nair G, Gafoor Z, Zhang J, et al. An empiric HIV risk scoring tool to predict HIV-1 acquisition in African women. J Acquir Immune Defic Syndr (1999). 2016;72(3):333–43.CrossRef Balkus JE, Brown E, Palanee T, Nair G, Gafoor Z, Zhang J, et al. An empiric HIV risk scoring tool to predict HIV-1 acquisition in African women. J Acquir Immune Defic Syndr (1999). 2016;72(3):333–43.CrossRef
51.
Zurück zum Zitat Balkus JE, Brown ER, Palanee-Phillips T, MatovuKiweewa F, Mgodi N, Naidoo L, et al. Performance of a validated risk score to predict HIV-1 acquisition among African women participating in a trial of the dapivirine vaginal ring. J Acquir Immune Defic Syndr. 2018;77(1):e8–10.PubMedPubMedCentralCrossRef Balkus JE, Brown ER, Palanee-Phillips T, MatovuKiweewa F, Mgodi N, Naidoo L, et al. Performance of a validated risk score to predict HIV-1 acquisition among African women participating in a trial of the dapivirine vaginal ring. J Acquir Immune Defic Syndr. 2018;77(1):e8–10.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Pintye J, Drake AL, Kinuthia J, Unger JA, Matemo D, Heffron RA, et al. A risk assessment tool for identifying pregnant and postpartum women who may benefit from preexposure prophylaxis. Clin Infect Dis. 2017;64(6):751–8.PubMed Pintye J, Drake AL, Kinuthia J, Unger JA, Matemo D, Heffron RA, et al. A risk assessment tool for identifying pregnant and postpartum women who may benefit from preexposure prophylaxis. Clin Infect Dis. 2017;64(6):751–8.PubMed
56.
Zurück zum Zitat Cameron DW, Simonsen JN, D’Costa LJ, Ronald AR, Maitha GM, Gakinya MN, et al. Female to male transmission of human immunodeficiency virus type 1: risk factors for seroconversion in men. Lancet. 1989;2(8660):403–7.PubMedCrossRef Cameron DW, Simonsen JN, D’Costa LJ, Ronald AR, Maitha GM, Gakinya MN, et al. Female to male transmission of human immunodeficiency virus type 1: risk factors for seroconversion in men. Lancet. 1989;2(8660):403–7.PubMedCrossRef
57.
Zurück zum Zitat Laga M, Manoka A, Kivuvu M, Malele B, Tuliza M, Nzila N, et al. Non-ulcerative sexually transmitted diseases as risk factors for HIV-1 transmission in women: results from a cohort study. AIDS. 1993;7(1):95–102.PubMedCrossRef Laga M, Manoka A, Kivuvu M, Malele B, Tuliza M, Nzila N, et al. Non-ulcerative sexually transmitted diseases as risk factors for HIV-1 transmission in women: results from a cohort study. AIDS. 1993;7(1):95–102.PubMedCrossRef
58.
Zurück zum Zitat Torrone EA, Morrison CS, Chen PL, Kwok C, Francis SC, Hayes RJ, et al. Prevalence of sexually transmitted infections and bacterial vaginosis among women in sub-Saharan Africa: an individual participant data meta-analysis of 18 HIV prevention studies. PLoS Med. 2018;15(2):e1002511.PubMedPubMedCentralCrossRef Torrone EA, Morrison CS, Chen PL, Kwok C, Francis SC, Hayes RJ, et al. Prevalence of sexually transmitted infections and bacterial vaginosis among women in sub-Saharan Africa: an individual participant data meta-analysis of 18 HIV prevention studies. PLoS Med. 2018;15(2):e1002511.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Yuh T, Micheni M, Selke S, Oluoch L, Kiptinness C, Magaret A, et al. Sexually transmitted infections among Kenyan adolescent girls and young women with limited sexual experience. Front Public Health. 2020;8:303.PubMedPubMedCentralCrossRef Yuh T, Micheni M, Selke S, Oluoch L, Kiptinness C, Magaret A, et al. Sexually transmitted infections among Kenyan adolescent girls and young women with limited sexual experience. Front Public Health. 2020;8:303.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Deese J, Philip N, Lind M, Ahmed K, Batting J, Beksinska M, Edward VA, Louw CE, Onono M, Palanee-Phillips T, Smit JA, Baeten JM, Donnell D, Mastro TD, Mugo NR, Nanda K, Rees H, Morrison C. Sexually transmitted infections among women randomised to depot medroxyprogesterone acetate, a copper intrauterine device or a levonorgestrel implant. Sex Transm Infect. 2021;97(4):249–55. https://doi.org/10.1136/sextrans-2020-054590.CrossRefPubMed Deese J, Philip N, Lind M, Ahmed K, Batting J, Beksinska M, Edward VA, Louw CE, Onono M, Palanee-Phillips T, Smit JA, Baeten JM, Donnell D, Mastro TD, Mugo NR, Nanda K, Rees H, Morrison C. Sexually transmitted infections among women randomised to depot medroxyprogesterone acetate, a copper intrauterine device or a levonorgestrel implant. Sex Transm Infect. 2021;97(4):249–55. https://​doi.​org/​10.​1136/​sextrans-2020-054590.CrossRefPubMed
61.
Zurück zum Zitat Delany-Moretlwe S, Mgodi N, Bekker LG, Baeten JM, Li C, Donnell D, Agyei Y, Lennon D, Rose SM, Mokgatle M, Kassim S, Mukaka S, Adeyeye A, Celum C. High prevalence and incidence of gonorrhoea and chlamydia in young women eligible for HIV pre-exposure prophylaxis in South Africa and Zimbabwe: results from the HPTN 082 trial. Sex Transm Infect. 2023;99(7):433–9. https://doi.org/10.1136/sextrans-2022-055696.CrossRefPubMed Delany-Moretlwe S, Mgodi N, Bekker LG, Baeten JM, Li C, Donnell D, Agyei Y, Lennon D, Rose SM, Mokgatle M, Kassim S, Mukaka S, Adeyeye A, Celum C. High prevalence and incidence of gonorrhoea and chlamydia in young women eligible for HIV pre-exposure prophylaxis in South Africa and Zimbabwe: results from the HPTN 082 trial. Sex Transm Infect. 2023;99(7):433–9. https://​doi.​org/​10.​1136/​sextrans-2022-055696.CrossRefPubMed
65.
Zurück zum Zitat Avuvika E, Masese LN, Wanje G, Wanyonyi J, Nyaribo B, Omoni G, et al. Barriers and facilitators of screening for sexually transmitted infections in adolescent girls and young women in Mombasa, Kenya: A Qualitative Study. PLoS one. 2017;12(1):e0169388.PubMedPubMedCentralCrossRef Avuvika E, Masese LN, Wanje G, Wanyonyi J, Nyaribo B, Omoni G, et al. Barriers and facilitators of screening for sexually transmitted infections in adolescent girls and young women in Mombasa, Kenya: A Qualitative Study. PLoS one. 2017;12(1):e0169388.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Adamson PC, Loeffelholz MJ, Klausner JD. Point-of-care testing for sexually transmitted infections: a review of recent developments. Arch Pathol Lab Med. 2020;144(11):1344–51.PubMedPubMedCentralCrossRef Adamson PC, Loeffelholz MJ, Klausner JD. Point-of-care testing for sexually transmitted infections: a review of recent developments. Arch Pathol Lab Med. 2020;144(11):1344–51.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Hatzold K, Gudukeya S, Mutseta MN, Chilongosi R, Nalubamba M, Nkhoma C, et al. HIV self-testing: breaking the barriers to uptake of testing among men and adolescents in sub-Saharan Africa, experiences from STAR demonstration projects in Malawi, Zambia and Zimbabwe. J Int AIDS Soc. 2019;22(S1):e25244.PubMedPubMedCentralCrossRef Hatzold K, Gudukeya S, Mutseta MN, Chilongosi R, Nalubamba M, Nkhoma C, et al. HIV self-testing: breaking the barriers to uptake of testing among men and adolescents in sub-Saharan Africa, experiences from STAR demonstration projects in Malawi, Zambia and Zimbabwe. J Int AIDS Soc. 2019;22(S1):e25244.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Indravudh PP, Sibanda EL, d’Elbée M, Kumwenda MK, Ringwald B, Maringwa G, et al. ‘I will choose when to test, where I want to test’: investigating young people’s preferences for HIV self-testing in Malawi and Zimbabwe. Aids. 2017;31 Suppl 3(Suppl 3):S203-s12.PubMedCrossRef Indravudh PP, Sibanda EL, d’Elbée M, Kumwenda MK, Ringwald B, Maringwa G, et al. ‘I will choose when to test, where I want to test’: investigating young people’s preferences for HIV self-testing in Malawi and Zimbabwe. Aids. 2017;31 Suppl 3(Suppl 3):S203-s12.PubMedCrossRef
71.
Metadaten
Titel
Awareness of Heightened Sexual and Behavioral Vulnerability as a Trigger for PrEP Resumption Among Adolescent Girls and Young Women in East and Southern Africa
verfasst von
Krishnaveni Reddy
Thesla Palanee-Phillips
Renee Heffron
Publikationsdatum
05.12.2023
Verlag
Springer US
Erschienen in
Current HIV/AIDS Reports / Ausgabe 6/2023
Print ISSN: 1548-3568
Elektronische ISSN: 1548-3576
DOI
https://doi.org/10.1007/s11904-023-00680-y

Weitere Artikel der Ausgabe 6/2023

Current HIV/AIDS Reports 6/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.