U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Consolidated Guidelines on HIV Prevention, Testing, Treatment, Service Delivery and Monitoring: Recommendations for a Public Health Approach [Internet]. Geneva: World Health Organization; 2021 Jul.

Cover of Consolidated Guidelines on HIV Prevention, Testing, Treatment, Service Delivery and Monitoring: Recommendations for a Public Health Approach

Consolidated Guidelines on HIV Prevention, Testing, Treatment, Service Delivery and Monitoring: Recommendations for a Public Health Approach [Internet].

Show details

3HIV PREVENTION

3.1. Combination HIV prevention

Combination prevention programmes use a mix of evidence-based biomedical, behavioural and structural interventions to meet the current HIV prevention needs of individuals and communities to have the greatest possible impact on reducing the number of people newly infected. Well-designed combination prevention programmes need to reflect the local HIV epidemiology and context. They should focus resources to reach populations at greatest HIV risk with effective, acceptable prevention to address both immediate risks and underlying vulnerability. Combination prevention mobilizes communities, civil society, the private sector, governments and global resources in a collective undertaking. It requires and benefits from enhanced partnership and coordination and should incorporate mechanisms for learning, capacity building and flexibility to permit continual improvement and adaptation to the changing epidemiological environment.

ARV drugs play a key role in HIV prevention. People taking ART who achieved viral suppression (<200 copies/mL) do not transmit HIV to sexual partners. ARV drugs taken by people without HIV as pre exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP) are both highly effective in preventing HIV acquisition.

Other biomedical interventions that reduce HIV risk practices and/or the probability of HIV transmission per contact event include the following.

  • Male and female condoms and condom-compatible lubricant. Male condoms are estimated to reduce heterosexual transmission by at least 80% and to offer 64% protection in anal sex among men who have sex with men, if used consistently and correctly (1,2). Fewer data are available for the efficacy of female condoms, but evidence suggests they can have a similar prevention effect (3).
  • Harm reduction for people who inject drugs. This is provided through a range of interventions and services.

    Needle and syringe programmes are highly effective in reducing HIV transmission through injecting drug use (4).

    Opioid substitution therapy with methadone or buprenorphine is the most effective form of treatment for opioid dependence and has the additional benefit of effectively reducing HIV transmission through injecting drug use. Opioid substitution therapy is also effective in improving ART uptake and adherence for people dependent on opioids (5,6).

    Overdose management with community distribution of naloxone to prevent opioid overdose death (7).

    WHO is developing new recommendations on behavioural interventions to reduce the risk of transmission of HIV, hepatitis and sexually transmitted infections through chemsex, which may include injecting drug use.

  • Voluntary medical male circumcision. The 2020 WHO guideline on voluntary medical male circumcision recommends that the intervention should continue to be promoted as an additional efficacious HIV prevention option within combination prevention for adolescent boys aged 15 years and older and adult men in settings with generalized epidemics to reduce the risk of heterosexually acquired HIV infection. High-certainty evidence from three randomized controlled trials (8) is supported by observational studies conducted between 1986 and 2017, showing that voluntary medical male circumcision reduced the risk of heterosexual acquisition of HIV by about 60%. Other benefits of medical male circumcision include the reduced risk of some other sexually transmitted infections among women and men, including human papillomavirus, the cause of cervical cancer (9).

In deciding whether to offer voluntary medical male circumcision to younger adolescents, ages 10–14 years old, safety, which is affected by their evolving physical development, and capacity to provide informed consent should be considered along with factors such as public health impact, the acceptability and feasibility of delivering voluntary medical male circumcision along with other services and maintaining the benefits of high voluntary medical male circumcision coverage (8).

A minimum package of services, including safer sex education, condom promotion, the offer of HIV testing services and management of sexually transmitted infections, must be delivered along with the male circumcision procedure. Other health services, such as hypertension and/or TB screening, malaria management and tetanus toxoid-containing boosters could be added (8).

Reaching men with HIV prevention, testing, treatment and care important is important for men’s health and for preventing HIV among their sexual partners. Interventions are needed that address barriers to reach men and build on facilitators to enhance adult men’s uptake of voluntary medical male circumcision and other health services. Evidence and case examples are available in the 2020 voluntary medical male circumcision guidance (8), with new examples being updated with experience.

Embedding voluntary medical male circumcision service delivery within the overall health system is key to achieving sustainability and aligns with global efforts to strengthen health systems and achieve universal health coverage. The WHO health systems building blocks can serve as a framework to consider issues and opportunities for sustaining voluntary medical male circumcision services (8).

Behavioural interventions can reduce the frequency of potential transmission events, including the following:

  • Targeted information and education. These are programmes that use various communication approaches, such as school-based comprehensive sexuality education, peer counselling and community-level and interpersonal counselling (including brief interventions) to disseminate messages. These messages encourage people to reduce behaviour that may increase the risk of acquiring HIV and increase behaviour that is protective (such as safer drug use, delaying sexual debut, reducing the frequency of unprotected sex, using male and female condoms correctly and consistently and knowing your and your partner’s HIV status). Recognition is growing that social media and mobile technology are important tools that can be integrated in HIV prevention programmes and can be particularly critical in informing about and providing prevention services to populations such as men who have sex with men.
  • Enabling interventions to address structural barriers to accessing services may increase access to, uptake of and adherence to prevention as well as testing and treatment services. Such interventions address the critical social, legal, political and enabling environment that contribute to HIV transmission, including legal and policy reform towards decriminalizing behaviour (such as drug use, sex work and same-sex sex), measures to reduce stigma and discrimination (including in the health sector), promoting gender equality and preventing gender-based violence and violence towards key populations, economic and social empowerment, access to schooling and supportive interventions designed to enhance referrals, adherence, retention and community mobilization.

Combination prevention for key populations

WHO recommended a comprehensive package of evidence-based HIV-related interventions and services for all key populations in 2014, updated in 2016 (10,11). The package comprised clinical interventions and a set of critical enablers required for successful implementation of programmes and access for the five key populations1. WHO is in the process of updating these guidelines to have a broader health focus and integrate HIV, hepatitis and sexually transmitted infections. The updated guidelines will recommend interventions and prevention packages by population, which will continue to include the critical enablers as outlined in Box 3.1.

Box 3.1Essential strategies for an enabling environment

  1. Supportive legislation, policy and financial commitment, including decriminalization of behaviour such as sex work, same-gender sex, gender identity and expression and drug use
  2. Addressing stigma and discrimination, including by making health services available, accessible and acceptable
  3. Community empowerment
  4. Addressing violence against people from key populations

Source: Consolidated guidelines on HIV prevention, diagnosis, treatment and care for key populations (11).

3.2. Pre-exposure prophylaxis for preventing the acquisition of HIV

PrEP is the use of ARV drugs by HIV-negative individuals to reduce the acquisition of HIV infection. Based on evidence from randomized trials, open-label extension studies and demonstration projects, WHO recommended daily oral PrEP containing tenofovir in 2015 as an additional prevention choice for people at substantial risk of HIV infection. In 2019, WHO updated this recommendation to include an additional dosing regimen, called event-driven PrEP, for cisgender men who have sex with men.

In 2021, WHO released a conditional recommendation that the dapivirine vaginal ring may be offered as an additional prevention choice for women2 at substantial risk of HIV infection as part of combination prevention approaches (12). As evidence for other PrEP products, including long-acting formulations, becomes available, WHO may make new or updated recommendations for PrEP.

3.2.1. Oral pre-exposure prophylaxis for preventing the acquisition of HIV

Recommendations (2016)

Oral pre-exposure prophylaxis (PrEP) containing TDF should be offered as an additional prevention choice for people at substantial riska of HIV infection as part of combination HIV prevention approaches(strong recommendation, high certainty evidence).

Source: Guidelines: updated recommendations on HIV prevention, infant diagnosis, antiretroviral initiation and monitoring (12).

a

See Box 3.2 for reflections on the definition of substantial risk of HIV infection.

Background

In the 2016 consolidated HIV guidelines, WHO recommended oral PrEP containing TDF as an additional prevention choice for people at substantial risk of HIV infection as part of combination HIV prevention approaches (13), replacing previous recommendations (10,14). This recommendation was based on a systematic review of 12 trials that addressed the effectiveness of oral PrEP and were conducted among serodiscordant couples, heterosexual men, women, men who have sex with men, people who inject drugs and transgender women (15). The review showed that, where adherence was high, significant levels of efficacy were achieved, demonstrating the value of this intervention as part of combination prevention approaches.

By recommending PrEP for people at substantial risk (see Box 3.2), the offer of PrEP can be focused based on local epidemiology and individual assessment rather than solely on a risk group, enabling a wider range of populations to benefit and ensuring that implementation is informed by local information regarding the settings and circumstances of HIV transmission.

In 2017, WHO released the PrEP implementation tool (16) and a technical brief on preventing HIV during pregnancy and breastfeeding in the context of PrEP (17). In 2018, WHO published a report on PrEP policy adoption by countries and an update on the interchangeability of FTC and 3TC in PrEP regimens (18).

In 2019, WHO published a technical brief updating the WHO recommendation on oral PrEP to include the option of event-driven dosing for cisgender men who have sex with men (19). This consists of the use of a double dose of oral PrEP 2–24 hours before sex, followed by a third dose 24 hours after the first two doses and a fourth dose 48 hours after the first two doses. This has been described as 2+1+1. If more sex acts take place in the following days, a single dose can be continued daily as long as sexual risk continues, with a single daily dose taken for each of two days after the last sex act. In 2019, WHO also published a technical brief on prevention and control of sexually transmitted infections in the era of oral PrEP for HIV (20).

Since WHO released the recommendation on oral PrEP in 2015, more than 100 countries have incorporated PrEP into their national HIV guidelines, and PrEP users have been reported in 77 countries (21).

Box 3.2“Substantial risk of HIV acquisition”

When this recommendation was initially made in 2016, WHO defined substantial risk of HIV infection provisionally as HIV incidence greater than 3 per 100 person–years in the absence of PrEP. HIV incidence greater than 3 per 100 person–years has been identified among men who have sex with men, transgender women and heterosexual men and women who have sexual partners with undiagnosed or untreated HIV infection. In 2016, it was suggested that implementing PrEP in a population with this level of HIV incidence was considered cost-effective or cost saving, although PrEP may still be cost-effective at lower HIV incidence levels.

However, individual risk varies considerably within populations depending on individual behaviour and the characteristics of sexual partners. In locations with a low overall incidence of HIV infection, there may be individuals at substantial risk who should be offered PrEP services (22). PrEP programmes should consider local context and heterogeneity in risk. Individual characteristics and behaviour that could lead to exposure to HIV, rather than population-level HIV incidence, are most important when considering those who might benefit from PrEP.

Individuals requesting PrEP should be given priority to be offered PrEP, since requesting PrEP likely indicates there is a risk of acquiring HIV. Cost–effectiveness should not be the only consideration when implementing PrEP programmes, since remaining HIV negative and having control over HIV risk has intangible value to people and communities.

Rationale and supporting evidence

This section summarizes the rationale and supporting evidence for the recommendation on daily oral PrEP from the 2016 consolidated HIV guidelines. This was based on a systematic review of 12 randomized controlled trials on TDF-containing oral PrEP. These findings have been published in detail elsewhere (15). A more recent systematic review had similar findings (23).

Summary of review findings

A systematic review and meta-analysis of PrEP trials containing TDF demonstrated that PrEP is effective in reducing the risk of acquiring HIV infection. The level of protection did not differ by age, sex, regimen (TDF versus FTC + TDF) and mode of acquiring HIV (rectal, penile or vaginal) (15). The level of protection was strongly correlated with adherence.

HIV infection

HIV infection was measured in 11 randomized controlled trials comparing PrEP to placebo, three randomized controlled trials comparing PrEP to no PrEP (such as delayed PrEP or “no pill”) and three observational studies. A meta-analysis of data from 10 trials comparing PrEP with placebo demonstrated a 51% reduction in risk of HIV infection for PrEP versus placebo (15,24,25).

Adherence

When all studies were analysed together, the results produced significant heterogeneity. The results from meta-regression conducted to evaluate whether certain variables moderated the effect of PrEP on reducing the risk of acquiring HIV infection demonstrated that adherence is a significant moderator.

When studies were stratified according to adherence levels (high, moderate and low based on proportion in the active arms with detectable drug in blood), heterogeneity in effectiveness was greatly reduced within adherence subgroups, demonstrating that most heterogeneity between studies can be explained by differing adherence levels. Within adherence subgroups, PrEP was most effective among the high-adherence group (defined as higher than 70% drug detection, but all studies in this group had adherence at or above 80%) and significantly reduced the risk of acquiring HIV in studies with moderate levels of adherence (41–70% drug detection). Among studies with low adherence (40% or lower drug detection), PrEP showed no effect in reducing HIV infection (15).

Mode of acquisition

When studies were stratified by mode of acquisition (rectal, vaginal or penile exposure), PrEP showed similar effectiveness across groups. The relative risk of HIV infection for PrEP versus placebo for rectal exposure was 0.34 (95% CI 0.15–0.80). For penile or vaginal exposure, the relative risk of HIV infection for PrEP versus placebo was 0.54 (95% CI 0.32–0.90) (15). Parenteral exposure to HIV was not analysed separately because only one study explicitly included people who inject drugs, and their exposure to HIV arose from sexual practices and incomplete access to sterile injection equipment.

Sex and gender

The 10 randomized PrEP trials reporting HIV outcomes included largely cisgender men and women. Women were included in six studies and men in seven studies. PrEP was effective for both cisgender men and women.

Since the recommendation was made in 2016, additional subgroup analyses of transgender women found PrEP to be effective when taken but there are still challenges with adherence (26). A recent review found high willingness to use oral PrEP among transgender women (27).

Safety

Ten randomized controlled trials comparing PrEP with placebo included data on any adverse event. Across studies, the rates of any adverse event did not differ for PrEP versus placebo. Similarly, there were no differences across subgroups, including mode of acquisition, adherence, sex, drug regimen, drug dosing or age.

Eleven randomized controlled trials comparing PrEP with placebo presented the results for any grade 3 or 4 adverse event. Across studies, there was no statistical difference in rates of any grade 3 or 4 adverse event for PrEP versus placebo and no statistical differences across subgroups, including adherence, sex, drug regimen, drug dosing or age (15).

Several studies noted subclinical declines in renal functioning and bone mineral density among PrEP users (2830). These subclinical changes did not result in clinical events, were not progressive over time and reversed after discontinuing PrEP.

People who start PrEP may experience side-effects in the first few weeks of use. In 2019, a systematic review of 12 randomized controlled trials comparing PrEP with placebo found that people taking PrEP were more likely to report gastrointestinal adverse events, such as vomiting, nausea and abdominal pain (23), although less than 10% of PrEP users across studies reported such adverse events. These side-effects are typically mild and self-limited.

Drug resistance

The risk of drug resistance to FTC was low overall – 11 people with FTC- or TDF-resistant HIV infection among 9222 PrEP users, or 0.1% – and this occurred mainly among people who were acutely infected with HIV when initiating PrEP: seven of the 11 people with FTC- or TDF-resistant HIV infection among 9222 PrEP users. The proportion of people with drug-resistant HIV did not differ in the PrEP and placebo groups among everyone at risk, although the number of events was low (six people infected). Multiple HIV infections (850) were averted for every case of FTC resistance associated with starting PrEP in the presence of acute HIV infection (15). Modelling the HIV drug resistance resulting from ART use is predicted to far exceed that resulting from PrEP use (31). Although mathematical models inform the risk of resistance, their results rely on data from clinical trials and make assumptions about the risk of drug resistance selection during PrEP. A more recent review, conducted in 2019, similarly found that HIV drug resistance with PrEP is uncommon and breakthrough infection despite high adherence to PrEP is rare (32). Countries are encouraged to monitor drug resistance against HIV drugs used for PrEP.

Sexual and reproductive health outcomes

At the time this recommendation was made, no evidence indicated that PrEP use led to risk compensation in sexual practices, such as decreased condom use or more sexual partners (33,34). Since then, changes in sexual behaviour after PrEP initiation have not been widely observed, although this has been documented in some settings (3537).

PrEP does not appear to affect the effectiveness of hormonal contraception, although two studies found trends towards higher rates of pregnancy among oral contraceptive users who also took PrEP. When multivariate analysis accounted for confounders, this relationship was not significant. Oral PrEP was not associated with increased adverse pregnancy-related events among women taking PrEP during early pregnancy (38,39). Drug–drug interactions between PrEP and gender-affirming hormone therapy for transgender women have been observed in some studies, with lower blood plasma TDF exposures among transgender women compared to cisgender men (4042). However, among transgender women and transgender men enrolled in directly observed daily dosing of PrEP, PrEP concentrations similar to cisgender men were observed (43). Protective concentrations can be reached with daily use of oral PrEP even in the presence of gender-affirming hormones, and daily oral PrEP should be offered to transgender and non-binary people at substantial risk of HIV. Serum hormone concentrations are not affected by TDF + FTC PrEP use (43).

Cost and cost–effectiveness

The HIV incidence threshold for cost-saving implementation of PrEP will vary depending on the relative costs of PrEP versus treatment for HIV infection and the anticipated effectiveness of PrEP. In some situations, PrEP may be cost saving, but other interventions may be more cost saving and scalable. Monetary costs should not be the only consideration, since staying free of HIV and having control over HIV risk has intangible value to people and communities. The cost–effectiveness of PrEP may decrease with declining HIV incidence in the context of universal treatment for HIV, but primary prevention, including PrEP, is essential to eradicate HIV, regardless of cost–effectiveness.

Offering PrEP in situations where the incidence of HIV is greater than 3 per 100 person-years is expected to be cost saving in many situations. Offering PrEP at lower incidence thresholds may still be cost-effective.

A review of cost–effectiveness studies for PrEP found that, in generalized epidemics, giving priority for PrEP use to people at substantial risk of acquiring HIV infection increases impact (34). Some of these studies found PrEP to be cost-effective within the context of ART expansion; others found no benefit. In concentrated epidemics (such as among men who have sex with men in the United States of America), PrEP could have significant impact. Studies have found PrEP to be cost-effective depending on the cost of the drug and delivery systems when PrEP uptake is higher among people at substantial risk. Higher PrEP uptake and adherence have been observed among men who have sex with men in demonstration projects (44,45). The results vary widely depending on epidemic type, location and model parameters, including efficacy, cost, HIV incidence and target population (46).

Equity and acceptability

Preventing HIV among PrEP users will contribute to equitable health outcomes by sustaining their health and the health of their sexual partners. People at substantial risk of HIV are often underserved, have barriers to accessing health services and have few effective HIV prevention options. Access to PrEP provides opportunities to engage these individuals in health care, including sexual and reproductive health services. Broadening PrEP recommendations beyond narrowly defined groups (such as men who have sex with men and serodiscordant couples) enables more equitable access and is likely to be less stigmatizing than targeting specific risk groups. Effective PrEP services will reduce future treatment costs overall by preventing HIV infection in populations with high incidence.

PrEP acceptability has been reported in multiple populations, including cisgender women (and pregnant and breastfeeding women), serodiscordant couples, female sex workers, young women, people who inject drugs, transgender people and men who have sex with men. A qualitative literature review of 131 peer-reviewed articles and 46 abstracts (47) showed that individuals have considerable interest in accessing PrEP as an additional choice for HIV prevention. Population support for providing PrEP was based on knowledge of safety and effectiveness and the compatibility of PrEP with other prevention strategies.

Feasibility

Providing oral PrEP to diverse populations has proven feasible in multiple trial settings, demonstration projects and national programmes. Although placebo-controlled trials among cisgender women (39,48) found significant barriers to uptake and adherence, PrEP adherence among women has generally been high when open-label PrEP is provided (49,50). The iPrEx OLE project and the Partners Demonstration Project both show that PrEP implementation is feasible for various populations, including men and women (44,51). The PROUD study in the United Kingdom demonstrated that PrEP is feasible and effective and is not associated with significant changes in behavioural risk (52). Other PrEP demonstration projects in Botswana, South Africa, Thailand and the United States of America confirm that protective levels of adherence are feasible for most PrEP users (49,50,5356) although challenges remain to achieve high levels of adherence among some young people (56). In 2019, following the publication of the initial recommendation, PrEP users were reported from 77 countries, and preliminary data suggests that considerable growth in global PrEP use continued in 2020 despite disruptions by the COVID-19 pandemic (21). More than 100 countries in the world have adopted the WHO recommendations on PrEP into national guidelines (21).

Implementation considerations

WHO published a comprehensive implementation tool for oral PrEP in 2017 (16). This tool includes practical suggestions for clinicians, laboratory monitoring, pharmacy services, testing services, counselling, community engagement and integration of services (including ART, sexually transmitted infections, PEP and other sexual and reproductive health services). WHO will revise this implementation tool in 2021–2022.

As an additional HIV prevention option, PrEP should not displace other effective and well-established HIV prevention interventions, such as condom programming and harm reduction, but rather should be integrated into existing health services. Stigma is a driver of HIV and could decrease or increase depending on how PrEP is implemented. PrEP should be promoted as a positive choice among people for whom it is suitable and their communities, in conjunction with other appropriate prevention interventions and services, including sexual and reproductive health services. Legal environments in which the rights of people at substantial risk of HIV are violated may represent an important barrier to PrEP implementation.

Provider training

Health-care providers should be trained and supported to have conversations to explore sexual and injecting risk behaviour with their clients and help clients consider their risk of acquiring HIV and the range of prevention options, including PrEP. This involves providing respectful and inclusive services, a familiarity with techniques for discussing sensitive behaviour and a strong patient–provider relationship that enables discussions of facilitators and barriers to engagement in health-care services, adherence and self-care. Service providers should be aware of the emotional and physical trauma that people at substantial risk of acquiring HIV infection may have experienced (57). The capacity for respectful work with people who have experienced trauma involves communication and skills development. Services that are appropriate for young people – especially young women and key populations – are essential for the success of all HIV treatment and prevention programmes, including PrEP. Service providers should consider all health, social, and emotional needs of people interested in and using PrEP and provide or refer to appropriate services as needed, including mental health support, intimate partner and gender-based violence services, family planning services, sexually transmitted infection testing and management, among others.

PrEP services can involve different types of health-care and lay providers for different aspects of PrEP service delivery. This includes nurses, pharmacists and lay and peer providers. Using a range of providers for PrEP service delivery has the potential to remove barriers to PrEP uptake and adherence, although adequate training of all service providers is necessary to ensure high-quality services.

Involving communities

Meeting the needs of populations at substantial risk of HIV infection requires the full participation of communities in developing and implementing programmes. The following are good participatory practices that apply to all priority and key populations.

  • Recognize the leadership and resilience of priority and key populations in addressing the HIV epidemic at both the local and global levels and sustain their participation through adequate funding and support for community-based organizations.
  • Ensure access to accurate knowledge and information about PrEP and early treatment by strengthening the capacity of the community-based organizations in educating and training their communities about their use.
  • Promote and expand community-based services, especially services led by priority and key populations.
  • Ensure that PrEP is offered as a choice, free of coercion and with access to other prevention strategies that may be preferred by the individuals at substantial risk.
  • Increase political commitment to rights, including the rights of priority and key populations, by decriminalizing consensual sexual activity and gender expression.
Linking PrEP with other health and community services

People at substantial risk of acquiring HIV are often medically underserved, have few other effective HIV prevention options, and frequently face social and legal challenges. Providing PrEP may give opportunities for increased access to a range of other health services and social support, including reproductive and sexual health services (including managing sexually transmitted infections), and mental health services, primary health care and legal services.

Community-based organizations – especially those working with priority and key populations – should play a significant role in delivering PrEP by engaging people at substantial risk, providing information about PrEP availability and use and promoting links between PrEP providers and other health, social and community support services. Community-based organizations can also be directly involved in delivering PrEP services, including by integrating peer and lay providers into services.

PrEP as part of combination prevention

PrEP should always be provided together with other HIV prevention options. Harm-reduction interventions – including access to sterile or new injection materials – are the mainstay of preventing HIV transmission through unsafe injecting practices, and such supplies should be made available to anyone using injected substances or medications. Condoms and lubricants should be made available, including for sex workers, who should be empowered to insist on their use (58).

Recommendations for early initiation of ART and PrEP in these guidelines are expected to facilitate the identification of people recently infected with HIV. Whenever possible, people in their social and sexual networks should be offered HIV testing, treatment and prevention services. PrEP should be considered, in combination with other prevention services, for HIV-uninfected partners of recently diagnosed people.

HIV testing

HIV testing is required prior to starting or restarting PrEP and should be conducted regularly (such as every three months) during PrEP use. Additional HIV testing conducted after one month of PrEP use can detect acute infection that may have been present when PrEP was started. If the initial HIV serology test result is non-reactive (negative) and there is no history or signs or symptoms of an acute viral syndrome, the person could be offered and initiated on PrEP. If the person has had a recent high-risk HIV exposure (such as within the past 72 hours) they can be offered PEP and transition to PrEP after the completion of PEP and following additional HIV testing.

Frequent HIV testing during PrEP use is also an opportunity to provide sexually transmitted infection screening and management as well as other health services. Using quality-assured HIV testing, according to the national algorithm is important, and should include good counselling, linkage to earlier HIV diagnosis and treatment and minimize the risk of drug resistance during PrEP and PEP.

WHO recommends testing using the same strategy and algorithm for PrEP users as for other individuals. More expensive and complex testing strategies may hinder access and are unlikely to provide any greater benefit in settings where NAT assays or fourth generation serology assays are not routinely used for HIV diagnosis.

During COVID-19, some settings experiencing disruptions to HIV services began utilizing HIV self-testing to maintain essential services – including for initiating, and monitoring ongoing, PrEP. WHO has supported the use of HIV self-testing during COVID-19 as an interim measure and is currently reviewing evidence on the use of HIV self-testing for oral PrEP initiation and monitoring.

Monitoring renal function

Reduced kidney function, indicated by a creatinine clearance of <60 ml/min, is a contraindication for using oral PrEP containing TDF. A systematic review and individual patient data meta-analysis of global programme data (59) found that less than 1% of individuals who were screened before starting oral PrEP had abnormal creatinine clearance levels and less than 3% of oral PrEP users experienced a decline in creatinine clearance to <60 mL/min. Older individuals, especially those over 50 years, with baseline creatinine clearance of <90 mL/min and with kidney-related comorbidities such as diabetes or hypertension, had a higher probability of declining to abnormal levels of creatinine clearance. Less than 1% of oral PrEP users younger than 30 years experience abnormal creatinine clearance. Some programmes may opt to screen for creatinine clearance for all oral PrEP users. However, since creatinine elevation is so rare among individuals younger than 30 years with no kidney-related comorbidities, creatinine screening may be considered optional in this group. To simplify the delivery and cost of PrEP, all individuals 30 years and older and those younger than 30 years who have comorbidities can be screened for serum creatinine once within 1–3 months after oral PrEP initiation. These suggestions by age and risk factors apply for both daily and event-driven dosing regimens.

More frequent screening than once is only suggested for individuals of any age with a history of comorbidities such as diabetes or hypertension, those 50 years or older and those who have had a previous creatinine clearance result of <90 mL/min. For these oral PrEP users, a further test after the baseline screening and every 6–12 months thereafter can be considered. When creatinine screening is conducted, any individuals with an estimated creatinine clearance of ≥60 mL/min can safely be prescribed TDF-containing oral PrEP. Waiting for creatinine screening results should not delay starting oral PrEP, since the results can be reviewed at a follow-up visit. Abnormal creatinine clearance of <60 mL/min should be repeated on a separate day before stopping TDF-containing oral PrEP. Other HIV prevention options should be discussed with the client. Creatinine clearance usually returns to normal levels after stopping PrEP, and PrEP can be restarted if creatinine clearance is confirmed to be to ≥60 mL/min 1–3 months after stopping PrEP. If creatinine clearance does not return to normal levels after stopping PrEP, other causes of renal insufficiency should be evaluated, such as diabetes and hypertension.

Hepatitis B and C

PrEP services provide a unique opportunity to screen for hepatitis B and hepatitis C infection and thus address multiple public health issues. Hepatitis B is endemic in some parts of the world where there is also a high burden of HIV. Testing oral PrEP users for hepatitis B surface antigen (HBsAg) once, at PrEP initiation, is preferred and has several advantages in these settings. Rapid point-of-care tests are available for HBsAg, and WHO has prequalified several rapid diagnostic tests. People with detectable HBsAg and clinical evidence of compensated or decompensated cirrhosis or people older than 30 years with persistently abnormal ALT levels and evidence of high-level hepatitis B replication (who do not have clinical evidence of cirrhosis) are eligible for long-term therapy for hepatitis B (60). People at risk of acquiring hepatitis B with non-reactive HBsAg test may be considered for hepatitis B vaccination depending on endemicity and country recommendations (61). The medications used for oral PrEP are active against hepatitis B. Withdrawing active therapy against hepatitis B can lead to virological and clinical relapse. Clinical relapse did not occur during or after PrEP use in trials that included people with chronic hepatitis B infection (62,63) and are considered very rare. hepatitis B infection is not a contraindication for daily oral PrEP use, but event-driven use of oral PrEP is inappropriate for individuals with chronic hepatitis B infection. Daily oral PrEP can be initiated before hepatitis B testing results are available.

Hepatitis C antibody testing can be considered at PrEP initiation and every 12 months thereafter depending on local epidemiological context, especially when PrEP services are provided to men who have sex with men, people who use drugs and people in prisons and other closed settings. Individuals with reactive serology test results should be referred for further assessment and treatment for hepatitis C infection (64). Hepatitis C infection is not a contraindication for daily or event-driven oral PrEP use, and PrEP can be initiated before hepatitis C test results are available.

Adherence

Support for adherence should include information that PrEP is highly effective when used as prescribed. For daily oral PrEP users, brief client-centred counselling that links daily medication use with a daily habit (such as waking up, going to sleep or a regular meal) may be helpful. Tailored interventions to facilitate adherence among particular groups – such as young people – may be needed. Support groups for PrEP users, including social media groups, may be helpful for peer-to-peer sharing of experience and challenges.

People who start PrEP may report side-effects in the first few weeks of use. These side-effects include nausea, abdominal cramping or headache, are typically mild and self-limited and do not require discontinuing PrEP. People starting PrEP who are advised of this start-up syndrome may be more adherent.

PrEP should be used effectively – during periods of substantial HIV risk – but is unlikely to be for life. PrEP can be discontinued if a person taking PrEP is no longer at risk. It is not unusual for people to start and stop PrEP repeatedly depending on periods of higher and lower HIV risk. Engaging with PrEP users and community support groups is important to facilitate the recognition of circumstances that involve substantial risk of acquiring HIV. Such periods of risk may begin and end with changes in relationship status, alcohol and drug use, leaving school, leaving home, trauma, migration or other events (55,65).

For cisgender men who have sex with men, event-driven PrEP is an effective strategy to reduce HIV risk. It entails a double dose of oral PrEP 2–24 hours before sex, followed by one dose each 24 and 48 hours after the first dose. This is sufficient to achieve high levels of protection against HIV. Event-driven PrEP is not an appropriate option for other PrEP users such as cisgender women, transgender women or transgender men having vaginal sex. Other populations are advised to take seven consecutive days of TDF-based oral PrEP to reach protective levels (66). For event-driven PrEP users, a single dose of PrEP can be taken daily as long as potential sexual exposure to HIV continues, with a daily dose for each of the two days after the last sex act. For people using daily PrEP, it has been suggested that PrEP may be discontinued 28 days after the last potential exposure to HIV if people do not have continuing substantial risk for HIV. However, pharmacokinetic data suggest that PrEP may be discontinued earlier (i.e. seven days) after the last potential exposure. WHO will release revised guidance on this topic in 2021–2022.

People who report a potential high-risk exposure to HIV in the 72 hours before presenting for PrEP should be considered for PEP (67). If substantial HIV risk continues after 28 days, PEP can be transitioned to PrEP.

Pregnancy and breastfeeding

Pregnancy and the postpartum period are characterized by substantial risk of acquiring HIV in some settings. HIV acquired during pregnancy or breastfeeding is associated with an increased risk of HIV transmission to the infant. An increasing body of evidence has demonstrated that TDF-containing oral PrEP is safe during pregnancy and breastfeeding (68). Antenatal and postnatal care services offer an opportunity to integrate PrEP services for women at substantial risk of HIV, but more operational experience and research are needed to understand the unique needs and challenges of this population and how to best address them. Contraception services, safer conception management and links to antenatal care should be available when providing PrEP services for women and transgender men.

Research gaps

Since WHO recommended offering oral PrEP for people at substantial risk of HIV acquisition in 2015, there has been considerable global research on PrEP use, including pilot projects, demonstration studies, and national programmes for PrEP. In addition, oral PrEP was used in at least 77 countries in 2019 (21). This expansion of oral PrEP services has generated substantial evidence on how to implement PrEP at scale but has also highlighted challenges with uptake, effective use and continuation of PrEP. Operational research is especially needed in diverse settings to generate demand for prevention services among adolescents and young people and support effective PrEP use. It is recognized that many PrEP users will not choose to take PrEP continuously for several years. Therefore, support to start, stop and restart PrEP related to periods of sexual risk is an important part of PrEP counselling. This includes innovative platforms such as using social media and mobile applications to engage with potential and existing PrEP users.

The global COVID-19 pandemic has accelerated a trend towards simplified, differentiated, and demedicalized oral PrEP service delivery. This includes using telehealth consultations for initiating and continuing PrEP and PrEP delivery at home and via pharmacies and other community-based locations. HIV self-tests have been used for initiating and continuing PrEP. In some places, peer and lay providers have been included in PrEP service delivery. All of these approaches have the potential to remove barriers to uptake and improve the effective use of PrEP. However, although the feasibility of these different forms of community-based PrEP delivery has been demonstrated in some settings, more operational research is needed on their effectiveness and scalability.

Some evidence indicates that using PrEP leads to changes in sexual practices that increase the risk for acquiring HIV. Although this form of risk compensation has been demonstrated in some settings, PrEP is likely to provide net benefits for HIV prevention, and potential changes in sexual behaviour underscore the need to integrate PrEP services with broader sexual and reproductive health services (35). The broader impact of PrEP on sexual health, the use of other HIV prevention methods, emotional well-being and stigma against people living with HIV may vary according to social and cultural contexts and remains a topic of interest. Research on PrEP is encouraged to consider diverse biological, behavioural and social outcomes, and operational research is needed across settings on how to optimally integrate PrEP with other health and social services. Research has shown high prevalence and incidence of sexually transmitted infections among PrEP users, and integrating sexually transmitted infection screening and treatment into PrEP services could have broad sexual health benefits. Similarly, integrating PrEP and reproductive health services could lead to broad health improvements. Family planning services may offer an opportunity for providing PrEP services, but more evidence on real-world programme integration is needed. Moreover, while an increasing amount of evidence is highlighting that oral PrEP is safe during pregnancy and breastfeeding, more operational research is needed on how to provide PrEP services to pregnant and breastfeeding women.

PEP started after recent exposure can be transitioned to PrEP if substantial HIV risk continues, so PrEP and PEP services should be integrated where appropriate. Operational research on PrEP should also consider social outcomes such as gender-based and intimate partner violence and how to effectively provide gender-based and intimate partner violence services to people accessing PrEP services.

Globally, the largest numbers of PrEP users have been among cisgender men who have sex with men and cisgender women at risk of acquiring HIV. More research is needed on the specific needs of transgender women, transgender men and non-binary people, including additional support for adherence in this population and integration of gender-affirming care with HIV services, including PrEP. Research involving transgender men and non-binary people is particularly lacking, including how to improve awareness and uptake of and adherence to PrEP.

PrEP awareness and use among people who use drugs is limited, and more research on improving the engagement of people who use drugs with PrEP services is needed (69). This includes more research on the feasibility and effectiveness of integrating harm-reduction and PrEP services. People in prisons and other closed settings and individuals recently released from those settings may also be at substantial risk of HIV infection in some geographical locations but are often not adequately reached by HIV prevention services, including PrEP.

WHO released guidance on event-driven PrEP for cisgender men who have sex with men in 2019, since evidence from randomized controlled trials showed high efficacy of non-daily PrEP dosing regimens. Although high use of event-driven PrEP has been reported in some settings, awareness and use of event-driven PrEP is low globally, which is partly because of slow adoption of event-driven PrEP into national guidelines. Further work at the country level is required on how to raise awareness and provide options for various oral PrEP dosing regimens. Moreover, because of the pharmacokinetics of TDF-containing oral PrEP, event-driven PrEP is not recommended for cisgender women and transgender men or non-binary people who have frontal or vaginal sex. Event-driven PrEP may be an appropriate option for all cisgender men (not just those who have sex with men), but little is known on oral PrEP dosing preferences among heterosexual cisgender men. Moreover, event-driven PrEP may be appropriate for transgender men and non-binary people assigned female at birth who exclusively have anal sex. However, there has been very limited research involving members from these diverse populations on preferences for different PrEP dosing regimens and the pharmacokinetics of TDF-containing oral PrEP, including in the context of gender-affirming care (49,53,54).

Oral PrEP has been shown to be cost-effective when provided to individuals at substantial risk of HIV in a range of settings and populations. However, differentiated and integrated oral PrEP service delivery, including in settings such as pharmacies and through community-based dispensing, and considering varying patterns of use, may offer opportunities for cost savings and efficiency. More research on the cost and cost–effectiveness implications of these evolving models of PrEP services is required.

With the dapivirine vaginal ring (see below), WHO has recommended additional PrEP modalities, and may recommend additional PrEP modalities, such as long-acting injectable cabotegravir, in the future. Research is needed on how to integrate these new PrEP modalities and dosing regimens into existing oral PrEP services, including cost and cost–effectiveness implications and on user preferences in diverse settings.

3.2.2. PrEP using the dapivirine vaginal ring

Recommendations (2021)

The dapivirine vaginal ring may be offered as an additional prevention choice for womena at substantial risk of HIV infection as part of combination prevention approaches(conditional recommendation, moderate-certainty evidence).

Source: Guidelines: updated recommendations on HIV prevention, infant diagnosis, antiretroviral initiation and monitoring (12).

a

For the recommendation on the dapivirine vaginal ring, the term women applies to cisgender women, meaning women assigned female at birth. There is no research at this time to support the dapivirine vaginal ring for other populations.

Background

PrEP delivered through a vaginal ring containing dapivirine, a novel non-nucleoside reverse-transcriptase inhibitor (NNRTI), as the active PrEP agent could provide an acceptable option for women who are unable or do not want to take oral PrEP. The dapivirine vaginal ring is a woman-initiated option to reduce the risk of HIV infection. It is made of silicone and contains dapivirine, which is released from the ring into the vagina slowly over one month. The ring should be continuously worn in the vagina for one month and then should be replaced by a new ring (70). The risk of HIV-1 infection is reduced 24 hours after ring insertion (71).

Adolescent girls and women in parts of sub-Saharan Africa continue to experience high HIV incidence. Current prevention options present challenges and barriers to use. The results from the recent ECHO trial (72) highlighted the high HIV incidence among women attending family planning clinics in parts of South Africa and Eswatini and that much greater focus is needed on integrating HIV prevention strategies for women receiving sexual and reproductive health services. In addition, adolescent girls and young women reported a preference for obtaining PrEP at service locations they are already comfortable attending, especially family planning and sexually transmitted infection services (73).

Initial outcomes from oral PrEP programmes for women are mixed (74). Some programmes report low uptake and low continuation (7577). Some women report facing challenges to taking daily oral PrEP. These include the need to take a pill every day, opposition to their taking oral PrEP from partners and side-effects that may occur during the first month of use. These concerns suggest that additional options are needed for PrEP delivery, including long-acting PrEP products that are potentially more discrete, do not rely on daily adherence and have less systemic adverse events. Supporting this evidence are studies demonstrating that women’s needs and preferences for sexual and reproductive health are heterogeneous (78). Expanding PrEP options to include a long-acting, woman-controlled option, such as the dapivirine vaginal ring, could help to meet unmet HIV prevention needs for women (78,79).

Rationale and supporting evidence

Summary of review findings

A systematic review and meta-analysis of dapivirine vaginal ring trials demonstrated that the ring is effective in reducing the risk of acquiring HIV infection. Two randomized controlled trials – the Ring Study (IPM-027) (80) and ASPIRE (MTN-020) (81) reported that the dapivirine vaginal ring was approximately 30% effective in reducing HIV infection in intention-to-treat analysis. A subgroup analysis by age did not show efficacy among women 18–24 years old, who had lower adherence (82). The results from two open-label extension studies – DREAM and HOPE – found increased efficacy, increased adherence and increased retention relative to the randomized controlled trials (83,84). The results from one of the open-label extension studies indicated a 62% reduction in HIV transmission, comparing study results to the simulated control (83). Further studies are underway or planned to help understand whether this lack of effect among younger women results from non-adherence or other factors and to identify ways to support adherence for younger women who choose the dapivirine vaginal ring for HIV prevention (85). Safety and acceptability are also being studied among women 15–19 years old, who were not included in the trials. The dapivirine ring acts locally, and systemic absorption is low (80). The trials reported no notable difference in the treatment and placebo arms of adverse events related to pregnancy, fetal outcomes and/or infant outcomes. However, since the number of pregnancies was small, ongoing trials are assessing further safety data during pregnancy and breastfeeding (86,87).

Reduction in HIV infection

The evidence for HIV infection measured as an outcome in five studies was of moderate certainty. A meta-analysis of HIV infection reported in the two Phase III placebo-controlled randomized controlled trials (ASPIRE and the Ring Study) found a 29% reduction in HIV risk (95% CI 11–43%). This was similar to a pooled analysis using time-to-event data conducted by investigators from both trials that found a 27% relative reduction in HIV risk comparing the dapivirine vaginal ring to the placebo arms (95% CI 9–42%) (88). Individually, ASPIRE found a 27% relative reduction in HIV risk (95% CI 1–46%) (81), and the Ring Study found a 33% relative reduction in HIV risk (95% CI 5–53%) (89) for active dapivirine vaginal ring versus placebo arms.

For ASPIRE, efficacy increased when observations from the two research sites with low adherence were dropped, yielding a 37% relative reduction in HIV risk (95% CI 12–56%) (81). ASPIRE conducted an age-stratified analysis excluding the two sites with low adherence and found that the dapivirine vaginal ring did not significantly reduce the risk of acquiring HIV among women younger than 25 years (the reduction in HIV incidence was 10%, 95% CI −41% to +43%), whereas HIV incidence was 61% lower for the dapivirine vaginal ring versus placebo among women 25 years and older (95% CI 32–77%) (81). A post hoc analysis showed no efficacy and lower adherence among women 18–21 years old. The Ring Study also conducted an age-stratified analysis but found no significant difference in risk reduction for women 21 years and younger versus women older than 21 years (90). However, when the results across the two trials were pooled using individual-level data in analysis conducted by investigators, the reduction in the risk of acquiring HIV-1 was significantly higher among participants older than 21 years; no risk reduction was observed for participants 21 years or younger (88).

The results from the two open-label extension studies, DREAM and HOPE, found increased efficacy, increased adherence and increased retention relative to the randomized controlled trials (83,84). The results from DREAM indicated a 62% reduction in HIV risk compared with the simulated control, and the results from HOPE demonstrated a 39% relative reduction in HIV risk (95% CI 14–69%) compared to the simulated control. Of note, the participants in HOPE were given the choice of using the dapivirine vaginal ring at every study visit, whereas the participants in DREAM had to be willing to use the dapivirine vaginal ring as part of the study’s eligibility criteria. In HOPE, 92% of the participants accepted the dapivirine vaginal ring at enrolment and 73% accepted the dapivirine vaginal ring for the duration of the study (84).

Adverse events

All randomized controlled trials and open-label extension studies presented data on any adverse events with overall moderate-certainty evidence at 24 months. Overall, rates of adverse events were similar across study arms, and the safety endpoints from the open-label extension studies were similar to those found in the randomized controlled trials. When the results from the three randomized controlled trials were combined in a meta-analysis, the rates of any adverse event for dapivirine vaginal ring versus placebo arms did not differ significantly (RR = 1.0, 95% CI 0.95–1.06). When meta-analysis was restricted to the two Phase III randomized controlled trials, the results also showed no difference for the dapivirine vaginal ring versus placebo arms (RR = 1.02, 95% CI 0.98–1.06). In addition, when restricted to assessing differences between grade 3 or 4 adverse events across studies, the results of the meta-analysis showed no difference between the dapivirine vaginal ring and placebo arms (RR = 1.18, 95% CI 0.68–2.05; low-certainty evidence) (91).

ASPIRE reported on study-related social harm, defining this as “nonmedical adverse consequences of dapivirine vaginal ring use or of trial participation more generally” (92). The results from ASPIRE found 94 instances of social harm with 4680 person-years of follow-up. Almost all (n = 87, 93%) were partner-related and were reported by 85 women, of whom 61% had disclosed study participation to their primary partners. Common triggers of social harm included the partner’s discovery of the ring during foreplay or sex, notifying the partner of a sexually transmitted infection or the partner suspecting that the ring was associated with ill health, “promiscuity” or “witchcraft”. The consequences in the small group of women experiencing social harm included destruction of the ring, physical violence or ending the relationship. About 60% of the cases of social harm were categorized as having a minimal impact on the quality of life. Younger women (18–26 years old) were more than twice as likely to experience social harm as older women, and reporting social harm was associated with short-term decreased product adherence (92).

Drug resistance

ASPIRE, the Ring Study, DREAM and HOPE analysed resistance to NNRTIs. The prevalence of NNRTI-resistant infections among seroconverters within these studies ranged from 10% to 28%. When combined in meta-analysis, the results from the two Phase 3 randomized controlled trials show no increased risk for NNRTI-resistant HIV infection for the dapivirine vaginal ring compared to placebo arms (RR = 1.13, 95% CI 0.64–2.01; low-certainty evidence) (91).

Sexual and reproductive health outcomes

All five studies reported on pregnancy incidence among participants, with no differences in incidence noted across the dapivirine vaginal ring and placebo arms. One analysis from ASPIRE evaluated contraceptive efficacy and found no differences for the dapivirine vaginal ring versus placebo arms (moderate-certainty evidence) (93). However, the study identified significant differences in pregnancy incidence by contraceptive method, with women using oral contraceptive pills having much higher pregnancy incidence than those using implants or injectables.

Two analyses, one from ASPIRE and one from a research site in the Ring Study, examined pregnancy-related outcomes and found no difference in adverse pregnancy-related outcomes for the dapivirine vaginal ring versus placebo arms (very-low-certainty evidence) (94,95). However, being on a stable form of contraception was an eligibility requirement for all studies included in this review, since the safety of taking dapivirine while pregnant and/or breastfeeding is unknown. In addition, all studies provided pregnancy tests to women monthly (quarterly during the latter half of the open-label extension studies), and participants immediately discontinued the study product if they became pregnant.

Behavioural outcomes, including incidence of curable sexually transmitted infections

One study described behavioural outcomes, including the number of sexual partners and condom use, observed at one research site in south-western Uganda within the Ring Study (96). The study found no significant change in reports of non-condom use at last sex as reported at baseline and week 104 (64% and 67%, respectively; moderate-certainty evidence). Over the same time span, 57% reported two or more sexual partners at four weeks compared to 56% at 104 weeks (moderate-certainty evidence). Four studies, including the Phase II safety study, ASPIRE, the Ring Study and DREAM, reported on incidence rates of curable sexually transmitted infections identified post-baseline. No differences between study arms were reported (moderate certainty of evidence). However, one research site from the Ring Study found, significant decreases in diagnoses of Trichomonas vaginalis and Neisseria gonorrhoea infection from baseline to 104 weeks of follow-up (96).

Cost and cost–effectiveness

According to the International Partnership for Microbicides, the current cost to produce the ring alone is US$ 7 per ring. It is anticipated that, in low- and middle-income countries, the ring will be provided free of charge to women at public health facilities. Based on several modelling and cost–effectiveness studies, the dapivirine vaginal ring is expected to cost less than oral PrEP since, from a provider perspective, it requires fewer health system resources. In previous studies addressing the cost–effectiveness of oral PrEP, the costs of HIV testing, creatinine clearance and hepatitis B surface antigen tests were all considered in the estimated cost of delivering oral PrEP. For delivering the dapivirine vaginal ring, the only required test is for HIV. One study from South Africa found that the dapivirine vaginal ring would be a cost-saving intervention for KwaZulu-Natal if the intervention were given priority for female sex workers (97,98). Another modelling study from South Africa found that the dapivirine vaginal ring could have a modest impact on the HIV epidemic and be a cost-effective intervention, even with low efficacy, if uniform coverage across all high-risk groups was achieved (99). Two other studies used the Goals model to assess the impact of the dapivirine vaginal ring across countries with a high burden of HIV infection and found that, although the dapivirine vaginal ring has potential to significantly affect epidemics, the impact is highly variable and depends on many factors, such as reaching UNAIDS targets and potential intervention cost (100,101).

Feasibility

Multiple studies of the dapivirine vaginal ring have been conducted in countries in southern and eastern Africa, thus proving its feasibility across certain settings where the ring is intended to be implemented. In addition to the safety study, two Phase 3 randomized controlled trials and two open-label extension projects, additional safety studies were successfully conducted among adolescent young women and postmenopausal women in the United States of America and among healthy women in Europe (102104). The dapivirine vaginal ring is relatively easy to transport and store. It does not require refrigeration and can be stored at room temperature. Several countries in sub-Saharan Africa (Kenya, South Africa, Zambia and Zimbabwe) are already considering initial steps on how to implement the dapivirine vaginal ring.

Acceptability and values and preferences

A review that included 11 articles and abstracts specifically relevant for vaginal rings containing dapivirine for HIV prevention found that the use of vaginal rings was highly acceptable (71–98% in randomized controlled trials and 62–100% in observational studies), and the vast majority of participants across studies reported that the rings are easy to insert and remove (105). Most women disclosed ring use to their male partners, although some women feared violence or anger from partners if ring use was discovered (106). The rings were not felt by 70–92% of participants during sexual intercourse and not felt by 48–97% of male partners. Ring acceptability increased over time as women became more comfortable using the ring and as the ring became more common in their community (105).

Women expressed preferences for devices that were easily accessible, long-acting and partner-approved that could prevent both HIV infection and pregnancy and that could also be used without the partner’s awareness, with minimal impact on sex, and with few side-effects (105). Similarly, a review specific to the dapivirine vaginal ring use identified 21 studies, all conducted in sub-Saharan Africa, and found high acceptability. The review also noted that partner influence can affect ring use and that perceived community awareness and acceptance of the ring is important (106).

A comprehensive systematic review and meta-analysis assessing the global acceptability of vaginal rings (agnostic to active pharmaceutical ingredient) similarly found that rings were highly acceptable (107). The overall acceptability (proportion of women reporting a favourable experience) across 46 studies and 19 080 women was 87% (95% CI 83–91%). This review also found that most women who used the dapivirine vaginal ring liked it, whereas women with no direct experience using a dapivirine vaginal ring stated that they did not think they would like such a product.

The vast majority of women found the dapivirine vaginal ring acceptable. Among the 280 participants who participated in a safety study conducted in sub-Saharan Africa, 95% reported that they would be willing to use the ring if proven effective (80). The results from safety studies among postmenopausal women and adolescents in the United States of America also found the ring highly acceptable (104,108). Qualitative results from ASPIRE found that women grew more accepting of the ring once they used it and developed a sense of ownership and empowerment related to ring use. Women also found the ring easy to use and integrate into their daily lives (109). The most commonly reported concerns were related to hygiene, especially during menses; potential negative health outcomes such as infertility; concerns the ring would get lost or stuck in the body; and concerns over partners feeling the ring during sex or not liking the ring (80,110114).

Equity

The Guideline Development Group judged that the introduction of the dapivirine vaginal ring as an additional prevention option would probably increase equity. The dapivirine vaginal ring offers an additional, discrete, woman-controlled biomedical HIV prevention option. Expanding PrEP options through offering the dapivirine vaginal ring, in addition to oral PrEP, could help meet the diverse needs and preferences of women. Evidence from the field of contraception has demonstrated an association between increased contraceptive choice and increased contraceptive use among women. This has shown that increasing biomedical HIV prevention options could have a similar effect (increased options may lead to increased use) (115). In addition, access to the dapivirine ring for women could also provide additional opportunities for sexual and reproductive health services.

Rationale for decision

The Guideline Development Group formulated a conditional recommendation favouring the dapivirine vaginal ring. The Group assessed that the benefits probably outweighed the harm based on the overall moderate-certainty evidence presented in the systematic review and meta-analysis, the cost–effectiveness of the dapivirine vaginal ring, widespread acceptability and demonstrated feasibility and the potential to increase equity as an additional prevention choice, noting some variability in younger age groups and concerns about use among pregnant and breastfeeding women because of a lack of sufficient evidence.

Implementation considerations

Comprehensive services

Similar to oral PrEP, the dapivirine vaginal ring should be provided to women in combination with other prevention interventions and health services. This should include provision of condoms, a range of contraceptive methods, testing and treatment of sexually transmitted infections and providing or referring to services that prevent and protect against gender-based violence. Where feasible, providing voluntary partner services should also be considered (116). HIV testing should be provided before initiating the use of the dapivirine vaginal ring and every three months while using it as part of the service provision package.

Choice

Although the studies reviewed for this question did not directly compare oral PrEP to using the dapivirine vaginal ring, current evidence suggests that oral daily PrEP, when taken as prescribed, has greater efficacy for HIV prevention than the dapivirine vaginal ring. Oral PrEP should be offered at sites where the dapivirine vaginal ring is provided to enable women to make a choice. Women should be provided with full information and counselling on the available prevention options and their relative efficacy and safety and counselled to help them to make an informed choice regarding the best option for them.

The dapivirine vaginal ring for adolescent girls and young women

The data from the trials were not able to demonstrate efficacy among women younger than 21 years, who had low adherence to ring use. More data are needed to understand dapivirine vaginal ring use among younger women. Experience from oral PrEP services for adolescent girls and young women has shown that younger women may need more support, especially during the early stages of taking oral PrEP, to support continuation. This may be similar for dapivirine vaginal ring use, and studies are ongoing and/or planned in this age group to understand implementation issues and adherence challenges and to ascertain effectiveness, if these can be overcome.

The dapivirine vaginal ring for women from key populations

Although there is no experience with providing the dapivirine vaginal ring to women from key populations, including sex workers and women who use drugs, the dapivirine vaginal ring is expected to protect sex workers and women who use drugs from HIV transmission via vaginal sex. However, before focused implementation is planned for these populations, understanding and considering the values and preferences of women from key populations will be key to ascertain whether they would consider the dapivirine vaginal ring an acceptable and helpful additional prevention choice and, if so, what would be the most acceptable way to deliver it.

Service delivery

Currently there is no experience with providing the dapivirine vaginal ring outside of research and open-label extension projects. Careful consideration, including engagement with women and providers, is needed when deciding where the dapivirine vaginal ring could be offered. These could include reproductive health services, sexually transmitted infection services, contraception services, gender-based violence services and services specific to adolescent girls and young women or youth-friendly services and other services that make oral PrEP available to women. Special considerations will be needed for acceptable and safe approaches for women from key populations. Implementing demonstration projects can be helpful in furthering the understanding of the service delivery models best suited to offer the dapivirine vaginal ring.

HIV testing

Similar to using oral PrEP, HIV testing is required before the dapivirine vaginal ring is offered and should be conducted regularly (such as every three months) while using the ring. Unlike oral PrEP, no creatinine monitoring or hepatitis B testing is required for the safe use of the dapivirine ring. People who test HIV-negative but report substantial risk or who request the ring can be linked to HIV prevention services where the potential for dapivirine vaginal ring use can be assessed. The frequent HIV testing while using the ring is also an opportunity to offer contraceptives, provide sexually transmitted infection screening and management as well as other health services. Using quality-assured HIV testing according to the national algorithm is important and should include counselling and linkage to confirmatory HIV testing and treatment for anyone who has an HIV reactive (positive) test while using the ring. WHO recommends testing using the same strategy and algorithm for dapivirine ring users as for other individuals. More expensive and complex testing strategies may hinder access and are unlikely to provide any greater benefit in settings where NAT assays or fourth-generation serology assays are not routinely used for HIV diagnosis.

Adherence support

Similar to oral daily PrEP, the dapivirine vaginal ring needs to be used continuously during periods of risk for effectiveness. Adherence support should therefore be a key part of service provision. Flexible and tailored support will be needed, especially as women start to use this new product. The opportunity for frequent check-ins with a health (or lay) provider may be needed to support use as women start to use the product. Additional adherence support should be considered for younger women. Partner and peer support should also be considered.

Demand creation

The dapivirine vaginal ring is a new product. In many communities where women experience higher HIV risk, it could be provided even if there is little or no awareness or experience with using other vaginal ring products, such as the contraceptive vaginal ring. If a community is considering implementing the dapivirine vaginal ring, it will be important to develop an awareness programme for both the community and providers that is rolled out before and during introduction of the product. This should include engagement with women’s networks, women’s key population networks and the opportunity to understand concerns and respond to questions about this new product. Messages for men and male partners should also be considered. Some women reported that being able to discuss ring use with partners was supportive and helpful in continuing ring use.

Training and support for providers

The dapivirine vaginal ring is a new product. In settings with a high burden of HIV infection considering implementing the dapivirine vaginal ring, provider experience in offering vaginal ring products is unlikely. National programmes should work to provide adequate training support, since this will be needed to develop and provide this service. Ongoing mentoring and supportive supervision, as programmes continue, should also be considered. Understanding provider issues and concerns and addressing these concerns will be key.

Research gaps

Safety in pregnancy and breastfeeding

Monthly use of the dapivirine vaginal ring has been shown to be safe and effective for HIV prevention among non-pregnant women of childbearing potential. However, data on how dapivirine affects pregnancy outcomes and infants are limited.

Data from animal toxicity studies that evaluated various concentrations of dapivirine vaginal gel, including concentrations substantially higher than the concentration available in the vaginal ring, did not identify any adverse effects on the maternal animals or the developing embryo or fetus (94).

In the MTN-020/ASPIRE trial, 169 of the 2629 women enrolled became pregnant during the trial (94). From this small data set, dapivirine use in the periconception period does not appear to be associated with adverse effects on pregnancy or infant outcomes. However, additional safety studies are needed of dapivirine vaginal ring use during pregnancy and breastfeeding. Two ongoing studies (MTN-042 (DELIVER) and MTN-043 (B-PROTECTED)) will provide further safety data by the end of 2021 (86,87). If these conclude that there are no safety concerns, continuing post-market surveillance activities will be needed to monitor for adverse pregnancy and fetal outcomes through the ARV drug pregnancy registration system.

Effective use among women younger than 21 years

A subanalysis of women younger than 21 years did not demonstrate efficacy in this age group, and adherence to the product was also low. Further studies are currently underway (such as MTN-034 (REACH) (117)) to assess adherence and safety in this age group and to understand barriers to use and ways to support adherence and continuation.

Acceptability among women from key population groups

There has been no research to date on implementing the dapivirine vaginal ring with key population groups, including sex workers and women who use drugs. Conducting values and preferences surveys with members of both communities will be important to understand their views on this intervention. Based on the results of these surveys, and if the communities feel that the dapivirine vaginal ring could be an important additional HIV prevention option, involving the community in designing and developing programmes will be critical.

The dapivirine vaginal ring as part of combination prevention

Women will be counselled on the dapivirine vaginal ring along with other prevention options such as daily oral PrEP. Male and female condoms and partner services must also be available and offered alongside the dapivirine vaginal ring. Some women may switch from oral daily PrEP to using the dapivirine vaginal ring and potentially back to oral PrEP use. These possible patterns of using ARV drugs for prevention are currently not known or understood and require careful support and assessment.

Some women may decide to use both the dapivirine vaginal ring and oral daily PrEP at the same time. Although using oral PrEP and the dapivirine vaginal ring together is probably safe, no evidence indicates that using them together will result in any additive advantage. Whatever the choice, adherence is important to optimize protection from either one. Further, inconsistent use of either or both when used simultaneously would be ineffective for HIV prevention. Using the dapivirine vaginal ring in combination with other prevention interventions and intermittent use of the dapivirine vaginal ring needs to be studied further, which could also include moving from oral PrEP to the dapivirine vaginal ring and back again according to circumstances.

It is not known whether introducing the dapivirine vaginal ring, and by increasing choice, will support more women at substantial HIV risk overall to access ARV drug–based prevention or whether the dapivirine vaginal ring will replace existing oral PrEP use for some users. Monitoring this will be important.

Cost and cost–effectiveness

Oral daily PrEP and the dapivirine vaginal ring are costly prevention interventions. This is why WHO suggests that these prevention options should be given priority for women at substantial HIV risk, since their use could have the greatest benefit and be most cost-effective. Further cost–effectiveness analysis using real-world data in various settings and population groups would be useful to guide future implementation for maximum impact.

3.3. Post-exposure prophylaxis

Recommendations (2016)

Overall

An HIV PEP regimen with two ARV drugs is effective, but three drugs are preferred(conditional recommendation, low-certainty evidence).a

Adults and adolescents

TDF + 3TC (or FTC) is recommended as the preferred backbone regimen for HIV PEP(strong recommendation, low-certainty evidence).a

DTG is recommended as the preferred third drug for HIV PEP(strong recommendation, low-certainty evidence).

When available, ATV/r, DRV/r, LPV/r and RAL may be considered as alternative third drug options for PEP(conditional recommendation, low-certainty evidence).

Childrenb

AZT + 3TC is recommended as the preferred backbone regimen for HIV PEP for children 10 years and younger. ABC + 3TC or TDF + 3TC (or FTC) can be considered as alternative regimens(strong recommendation, low-certainty evidence).a

DTG is recommended as the preferred third drug for HIV PEP with approved DTG dosing(strong recommendation, low-certainty evidence).

When available, ATV/r, DRV/r, LPV/r and RAL may be considered as alternative third drug options for PEP(conditional recommendation, low-certainty evidence).

a

Source: Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: recommendations for a public health approach – second edition (13).

b

The choice of ARV drugs for children will depend on the availability of approved dosing and age-appropriate formulations for children.

Background

WHO guidelines for HIV PEP, formulated in 2014, aimed to provide a simplified approach to delivering PEP, given the suboptimal uptake and completion of PEP (67,118,119). The guidelines aimed to align recommendations for HIV PEP with the ARV drugs available in low- and middle-income countries for treating and preventing HIV.

The 2016 WHO consolidated HIV guidelines provided additional recommendations on eligibility, timing, prescribing and adherence support and clinical considerations (13). The WHO clinical guidelines on responding to children and adolescents who have been sexually abused describe further clinical considerations in providing appropriate care to children and adolescents who have been sexually abused (120). In 2018, new evidence provided information about the tolerability and completion rates of the WHO-recommended HIV PEP regimens and data on newer ARV drugs, notably DTG. In response, WHO provide updated recommendations on ARV drugs for HIV PEP (121).

Rationale and supporting evidence

A systematic review completed in 2018 assessed the tolerability of HIV PEP and completion of different ARV drug regimens recommended by the 2016 WHO consolidated HIV guidelines (13,121). The systematic review identified 16 studies reporting the outcomes of HIV PEP regimens using TDF + 3TC (or FTC) backbones (121). All studies involved adults, and no additional evidence was retrieved for PEP regimens for children or adolescents. Overall, the highest completion rates for HIV PEP were reported for TDF + 3TC (or FTC) in combination with DRV/r (93%, 95% CI 89–97%) or DTG (90%, 95% CI 84–96%). These regimens were also associated with the lowest rates of discontinuation or substitutions because of adverse events (1%, 95% CI 0–2% for DRV/r; 1%, 95% CI 1–4% for DTG).

For adults, the Guideline Development Group recommends that DTG may be used as the preferred third drug for HIV PEP. This recommendation considered the high rates of PEP completion and low rates of adverse events as well as the established high tolerability of DTG when used in ART (121). This preference also considered cost, current and anticipated availability, low potential for drug–drug interactions and the desirability of aligning with recommendations for ART. Alternative third drug options include ATV/r, DRV/r, LPV/r and RAL, with the choice to be based on considerations of tolerability and completion rates as well as cost, availability and acceptability (121) (Table 3.1).

Updated information since 2019

DTG was approved in June 2020 for all children older than 4 weeks weighing more than 3kg and available with dispersible tablets that can be easily administered for all children weighting less than 20 kg. For children weighting more than 20 kg, 50 mg adult film-coated tablets can be use.

The WHO 2014 guidelines on HIV PEP noted that data on using EFV in HIV PEP were lacking and that there are concerns about giving a drug associated with early central nervous system and mental health adverse events to HIVnegative people who may have anxiety related to HIV exposure. Since then, data have been published suggesting that EFV is associated with high rates of discontinuing HIV PEP because of central nervous system events (122). EFV should therefore only be used as a third drug option when no other options are available.

For children, no new evidence has been published since the review carried out for the 2014 guidelines However, the recommendation to provide DTG as a preferred drug option for this population (from four weeks and 3 kg) is now included, extrapolating from data for adults with the goal of aligning the recommendations for adults and adolescents.

Considerations for adolescent girls and women of childbearing potential

As part of comprehensive PEP services, all women should be offered pregnancy testing at baseline and follow-up. Emergency contraception should be offered to girls and women as soon as possible and within five days of sexual exposure. For women not wanting to take emergency contraception, an alternative to DTG should be provided (121).

Assessing eligibility

HIV PEP should be offered and initiated as early as possible for all individuals with exposure that has the potential for HIV transmission, preferably within 72 hours. For individuals who may not be able to access services within this time, providers should consider the range of essential interventions and referrals that should be offered to clients presenting after 72 hours.

Eligibility assessment should be based on the HIV status of the source whenever possible and may include consideration of background prevalence and local epidemiological patterns.

The following types of exposure may warrant HIV PEP.

  • Body fluids: blood, blood-stained saliva, breast-milk, genital secretions; cerebrospinal, amniotic, peritoneal, synovial, pericardial or pleural fluids. Although these fluids carry a high risk of HIV infection, this list is not exhaustive. All cases should be assessed clinically, and the health care workers should decide whether the actual exposure constitutes a significant risk.
  • Types of exposure: (1) mucous membrane from sexual exposure; splashes to the eye, nose, or oral cavity and (2) parenteral exposures.

Exposure that does not require HIV PEP includes:

  • when the exposed individual is already HIV positive;
  • when the source is established to be HIV negative; and
  • exposure to bodily fluids that do not pose a significant risk: tears, non-blood-stained saliva, urine and sweat.

In cases that do not require PEP, the exposed person should be counselled about limiting future exposure risk. Although HIV testing is not required, it may be provided if desired by the exposed person.

Clinical considerations

For PrEP, there is concern about the potential risk of hepatic flares among people with chronic hepatitis B once TDF-, 3TC- or FTC-based PEP is stopped. Assessment of hepatitis B infection status should not be a precondition for offering TDF-, 3TC- or FTC-based PEP, but people with established chronic hepatitis B infection should be monitored for hepatic flare after discontinuing PEP. Among people with unknown hepatitis B status and where hepatitis B testing is readily available, people started on TDF-, 3TC- or FTC-based PEP should be tested for hepatitis B to detect active hepatitis B infection and the need for ongoing hepatitis B therapy after discontinuing PEP.

NVP should not be used for PEP for adults, adolescents and older children because of the risk of life-threatening serious adverse events associated with HIV-negative adults using this drug.

For infants, NVP has been widely and safely used for HIV-uninfected infants for preventing vertical transmission of HIV and should be used for preterm babies or infants younger than four weeks old when DTG cannot be used. However, because the NVP toxicity profile beyond infancy remains unclear, its use should be avoided for children older than two years.

EFV is widely available as a third agent since it is used as part of the preferred first-line ART regimen. EFV is well tolerated for treatment but has limited acceptability for use as PEP since there are concerns about giving a drug associated with early nervous system and mental health adverse events to HIV-negative people who may have anxiety related to HIV exposure.

Full guidance on managing other conditions associated with possible exposure to HIV is provided in the 2014 Guidelines on post-exposure prophylaxis for HIV and the use of cotrimoxazole prophylaxis for HIV-related infections among adults, adolescents and children: recommendations for a public health approach (67).

Implementation considerations

The uptake and completion rates for HIV PEP are suboptimal, and the recommendations for HIV PEP regimens should be considered together with existing WHO recommendations aimed at improving completion rates for HIV PEP, including adherence support and providing a full 28-day course of medication at the first clinic visit (13,67).

Choice of HIV PEP regimen should consider the ARV drugs already being procured within national HIV programmes. Additional considerations include the availability of heat-stable formulations, daily dosing, availability and affordability (Table 3.1).

People may be subject to ongoing high risk of exposure to HIV, leading to multiple prescriptions for PEP. In such situations, health-care providers should discuss with their clients the potential benefits of transitioning to HIV PrEP (16,22).

Table 3.1. Characteristics of third drug options for PEP.

Table 3.1

Characteristics of third drug options for PEP.

3.4. Infant prophylaxis

Good practice statement (2016)

ART should be initiated urgently among all pregnant and breastfeeding women living with HIV, even if they are identified late in pregnancy or postpartum, because the most effective way to prevent HIV vertical transmission is to reduce maternal viral load. a

Source: Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: recommendations for a public health approach – second edition (13).

a

Whenever possible, all efforts should be made to identify HIV-infected pregnant women early enough to avoid the need for enhanced prophylaxis.

Recommendations (2016)

  • Infants born to mothers with HIV who are at high risk of acquiring HIVa should receive dual prophylaxis with daily AZT and NVP for the first six weeks of life, whether they are breastfed or formula fed (strong recommendation, moderate-certainty evidence).
  • Breastfed infants who are at high risk of acquiring HIVa, including those first identified as exposed to HIV during the postpartum period, should continue infant prophylaxis for an additional six weeks (total of 12 weeks of infant prophylaxis) using either AZT and NVP or NVP alone (conditional recommendation, low-certainty evidence).
  • Infants of mothers who are receiving ART and are breastfeeding should receive six weeks of infant prophylaxis with daily NVP. If infants are receiving replacement feeding, they should be given four to six weeks of infant prophylaxis with daily NVP (or twice-daily AZT) (strong recommendation, moderate-certainty evidence for breastfeeding infants; strong recommendation, low-certainty evidence for infants receiving only replacement feeding).

Source: Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: recommendations for a public health approach – second edition (13).

a

High-risk infants are defined as those:

-

born to women with established HIV infection who have received less than four weeks of ART at the time of delivery; or

-

born to women with established HIV infection with viral load >1000 copies/mL in the four weeks before delivery, if viral load is available; or

-

born to women with incident HIV infection during pregnancy or breastfeeding; or

-

born to women identified for the first time during the postpartum period, with or without a negative HIV test prenatally.

Background

Despite decades of progress in decreasing rates of vertical (mother-to-child) transmission, children continue to acquire HIV. Even with expanded treatment coverage for women with HIV, perinatal transmission continues to occur among infants born to women with HIV diagnosed in pregnancy or at delivery. Infants are also at risk of acquiring HIV during breastfeeding from a woman living with HIV. Roughly half of all newly infected children acquire HIV during breastfeeding. Although countries continue to make progress, challenges remain in retaining women living with HIV in health-care services and on effective ART throughout pregnancy and the breastfeeding period as well as detecting women who acquire during pregnancy and breastfeeding and preventing this from happening. Addressing these gaps requires continuing to emphasize promoting universal testing and treatment in the antenatal period and retesting HIV-negative women during pregnancy, at delivery and during breastfeeding to identify incident HIV, especially in settings with a high burden of HIV infection. In addition, it is important to consider the need for enhanced infant prophylaxis for the infants born to mothers that have not received early, effective ART.

The 2016 WHO consolidated HIV guidelines recommend a dual regimen of AZT and NVP for infants deemed to be at high risk of vertical transmission, which can be extended for up to 12 weeks for breastfeeding infants (13). A high-risk infant is defined as an infant whose mother was first identified as HIV-infected at delivery or in the postpartum, infected during pregnancy or breastfeeding, started ART late in pregnancy or did not achieve viral suppression by the time of delivery (Fig. 3.1). All high-risk infants should receive dual drug prophylaxis (AZT plus NVP) for the first six weeks. For breastfeeding infants, this should be followed by either an extra six weeks of AZT plus NVP or an extra six weeks of NVP alone.

WHO guidelines on infant feeding (123) in relation to HIV reaffirm the position of WHO that the best way to prevent vertical transmission in the postpartum period and optimize infant survival is to ensure that mothers living with HIV are receiving ART, have suppressed viral loads and are able to breastfeed their infants for up to two years, with the infant being exclusively breastfed in the initial six months. If a mother receiving ART has suppressed viral loads, the risk of breast-milk transmission is very low and infant prophylaxis confers minimal additional benefit beyond 4–6 weeks of life.

Since the release of the 2016 WHO consolidated HIV guidelines, countries have adopted enhanced postnatal prophylaxis using a variety of different approaches to adapt to the country context and challenges. In about one third of AIDS Free priority countries (Burundi, Eswatini, Ethiopia, Ghana, Kenya, Mozambique and Zambia), enhanced postnatal prophylaxis has been adopted for all breastfeeding HIV-exposed infants, and the remaining countries have adopted enhanced postnatal prophylaxis for high-risk infants identified primarily based on maternal ART duration and, when available, maternal viral load close to delivery. Most countries opted for at least 12 weeks of prophylaxis, usually AZT + NVP for the first six weeks followed by NVP alone. Five countries (Eswatini, Kenya, Namibia, South Africa and Zambia) link the duration of enhanced postnatal prophylaxis to the maternal viral load and extend enhanced postnatal prophylaxis over the entire breastfeeding period when viral suppression is not achieved. Finally, in three countries (Botswana, United Republic of Tanzania and Zambia), administration of three drugs in a fixed-dose dispersible tablet formulation based on AZT + 3TC + NVP has been adopted to address the challenges of procuring individual liquid formulations.

Evidence for the recommendation

This recommendation is based on evidence from randomized clinical trials (124) and considers the risk–benefit ratio of enhanced postnatal prophylaxis: potential for increased drug toxicity versus additional protection from HIV transmission (13). The systematic review (124) focused on studies that report on outcomes following the use of combined and/or prolonged infant prophylaxis regimens compared with the current standard of care. Although some of the studies reviewed were conducted in settings in which formula feeding is the norm, the findings can still be applied to breastfeeding populations since intrapartum HIV transmission is an important driver of vertical HIV transmission in both settings. Four studies met the criteria for inclusion, of which two were randomized trials (125,126) and two were observational studies (127,128).

  • The intrapartum transmission rate was found to be significantly lower with the two-drug and the three-drug regimens versus AZT alone (126). Serious adverse events possibly or probably related to study drugs were more frequent with the three-drug regimen than with AZT-alone or the two-drug group.
  • In a breastfed population, infants who received six months of NVP experienced a 54% lower transmission rate at six months compared with those who received only six weeks of NVP (126). However, among the infants born to mothers receiving ART at the time of randomization, the postnatal transmission rate was extremely low and did not differ between those who received longer duration NVP prophylaxis versus placebo.
  • In a large European cohort of “high-risk” mother–infant pairs, no difference was reported in serious adverse events between infants who received one, two or three drugs. When neutropaenia was compared between the two-drug and three-drug arms, there was a trend towards more events in the three-drug arm, but this was not statistically significant (127).
  • A single-arm study in non-breastfeeding infants of mothers who received less than eight weeks of antepartum ART in Thailand gave infant prophylaxis with AZT + 3TC + NVP for two weeks, followed by AZT + 3TC for an additional two weeks; no intrapartum infections were observed and the rate of serious adverse events among infants receiving intensified prophylaxis was lower than in a historical observational cohort (128).
  • None of the studies reviewed addressed infants identified in the postpartum period or infants exposed to an incident HIV infection either during pregnancy or while breastfeeding. However, the findings of the systematic review could probably be applied to these settings as well.

The recommendations for extended prophylaxis for breastfeeding infants are predicated on maternal ART being initiated at or before the time when infant prophylaxis is begun (whether at birth or when maternal HIV is first detected postpartum), since infant prophylaxis is intended only to provide a bridge of protection to the infant during the period that maternal viral load is decreasing on ART.

Defining high-risk infants

The 2016 WHO consolidated HIV guidelines acknowledge a range of factors to be considered when assessing risk, including examples from countries that stratify HIV-exposed infants according to risk (13). Factors such as prolonged rupture of membranes, preterm delivery and low birth weight are no longer associated with increased risk of transmission when mothers are receiving ART. The critical determinants of transmission risk in the ART era are maternal viral load and duration of maternal ART.

Whenever possible, use same-day point-of-care testing for viral load testing of pregnant and breastfeeding women to expedite the return of results and clinical decision-making (see subsection 4.72). If this is not available, viral load specimens and results for pregnant and breastfeeding women should be given priority across the laboratory referral process (including specimen collection, testing and return of results). The 2021 WHO clinical guidelines (12) also state that for all pregnant women, regardless of ART initiation timing, viral load testing should be performed at 34–36 weeks of gestation (or at the latest at delivery) to identify women who may be at risk of treatment failure and/or may deliver infants at higher risk of perinatal transmission.

In this context, the following scenarios may be considered as working definitions of high risk:

  • born to women with established HIV infection who have received less than four weeks of ART at the time of delivery; or
  • born to women with established HIV infection with viral load >1000 copies/mL in the four weeks before delivery, if viral load is available; or
  • born to women with incident HIV infection during pregnancy or breastfeeding; or
  • born to women identified for the first time during the postpartum period, with or without a negative HIV test prenatally.

Fig. 3.1. Algorithm for risk assessment at the time of delivery to help identify infants at high and low risk of infection.

Fig. 3.1

Algorithm for risk assessment at the time of delivery to help identify infants at high and low risk of infection.

Implementation considerations

Providing multiple drugs to newborns is challenging from an operational perspective, and although AZT and NVP are proposed based on the available data, one is administered once daily and the other twice daily. Provider training will be critical to the successful uptake of these recommendations, and innovative approaches to dosing (such as using twice-daily dosing of NVP) may help to simplify administration. When the recommended regimen is not available or feasible, use of alternative options such as RAL, 3TC, LPV/r solid formulations or triple-drug fixed-dose combinations containing AZT, NVP and 3TC may be considered (the dosing recommendations can be found in Annex 1).

Consistent with the recommendations on early infant diagnosis, no specific approach to the testing of high-risk newborns is recommended. However, birth infant diagnostic NAT testing may be considered. In addition, infants who are first identified as HIV exposed postpartum have a high cumulative risk of already having acquired HIV by the time prophylaxis is initiated, and an HIV NAT test should be performed around the time of initiating prophylaxis. This will help to minimize the risk that extended prophylaxis among infected infants will lead to the development of resistance and will help promote linkage to timely initiation of ART.

Some programmes have adopted enhanced prophylaxis for all HIV-exposed infants. Although this may simplify decision-making, it increases costs and exposes many infants who may not need enhanced prophylaxis to added toxicity. This type of approach ought to be reserved for selected situations in which a majority of mothers are at high risk of transmitting HIV. Data on the average duration of ART at delivery and, where available, the proportion of pregnant women with viral load >1000 copies/mL at the end of the third trimester might help policy-makers to determine whether the potential benefit outweighs the added costs and toxicity. Even then, it should only be an interim measure while strategies to increase the coverage of maternal testing, early treatment and improved adherence are being implemented.

However, there are several situations where suppression of viral loads throughout the breastfeeding period cannot be ensured in the mother, for example:

  • if a mother refuses or is unable to start or continue ART and intends to breastfeed her infant;
  • if the provider knows the mother is poorly adherent to ART while breastfeeding; and
  • if maternal viral load is known to be elevated when the infant prophylaxis regimen is about to be stopped.

There is no formal recommendation for these types of situations and no evidence to guide the best course of action. It is reasonable, however, to assume that infant prophylaxis serves as a back-up solution for preventing postnatal transmission of HIV, and national programmes could consider the merits of giving clinical providers the option of continuing infant prophylaxis beyond the recommended 6- or 12-week period. If this option is provided in the national guidelines, there ought to be clearly defined scenarios in which continuing prophylaxis is warranted. National guidelines should also emphasize that the best way to prevent breast-milk transmission of HIV is by optimal maternal treatment for the entire duration of exposure. Continuing prophylaxis should therefore be seen as an interim measure while efforts are made to support and improve maternal treatment adherence. Deciding to continue prophylaxis should consider the factors that led to poor maternal adherence since they may affect adherence to infant prophylaxis. Once stopped, infant prophylaxis should not be restarted if there are fresh concerns about maternal adherence. No evidence supports such an approach; instead the focus should be on determining why the mother was unable to remain adherent. If the decision has been made to continue infant prophylaxis, mothers and infants should be evaluated at regular intervals to assess the need for ongoing infant prophylaxis.

Research gaps

Potential areas for research include clinical and pharmacological studies to inform the development of improved ARV drug formulations, including fixed-dose combinations in appropriate doses for newborns and infants. Research into the use of alternative drugs for prophylaxis that are better tolerated and that may have greater efficacy for infant prophylaxis such as integrase inhibitors could also be considered. Studies to evaluate the clinical relevance of postnatal viraemic episodes in breastfeeding mothers and the relative contribution of such episodes to infant HIV infection are needed, coupled with evaluation of whether enhanced prophylaxis adds additional benefit in a population with a well-implemented effective maternal ART programme but known difficulty with adherence and viraemia in the postpartum period.

Finally, implementation science research to evaluate the optimal definition of high risk in the context of universal maternal ART and the impact of various service delivery models and how they affect adherence to enhanced postnatal prophylaxis and retention would be of great value.

Box 3.2Improving service delivery and implementing a postnatal package of care

There are several barriers to the uptake of effective infant HIV services, and no single intervention can address all the barriers facing women and their infants at different times and places. Socioeconomic and traditional factors that keep mother–infant pairs together are among the enabling circumstances that improve service uptake. Programmes could benefit from combining effective interventions into service packages to support service provision and focus on the community engagement that supports uptake.

Interventions that have been proven to improve the provision and uptake of infant HIV services and the retention of mother–infant pairs include:

  • client-focused interventions that provide support to individual clients using reminder text messaging, conditional cash incentives and male partner involvement; and
  • health system-focused interventions, including measures to enhance the programme (point-of-care testing technologies, provider training and support, enhanced counselling services and peer support), to strengthen the health system (initiatives to improve quality and integrating maternal, newborn and child health and HIV services) and to support community-based services and health-care workers.

Promoting integration to reduce fragmentation of care for mothers and infants and ensure that infants remain in the testing cascade until final diagnosis should be a priority. Strong antenatal and well-baby care systems provide opportunities to strengthen service delivery for HIV-exposed infants. Integration within a well-established maternal, neonatal and child services platform, which traditionally provides services closest to clients, facilitates mother–infant pair follow-up and reduces the cost and time-visit burden on clients (integrated information systems that link mother and infant information improve client tracking and facilitate the continuity of care provision). Examples include longitudinal follow-up registers and cohort analysis and linkage with information on community services. Programmes should, however, consider the increasing burden on the maternal, newborn and child platform within the context of existing human resources and the challenges of changing services with new packages.

Community engagement and community-based services play an important role in supporting HIV-exposed infant care. These clear and highly context-specific services play a boosting role in supporting facility-focused services and include community-based HIV testing. The engagement of networks of women living with HIV has been effective in several countries and has been used to improve community HIV literacy to create demand, form support groups at the facility and community levels, strengthen linkage to care by escorting newly diagnosed clients to treatment clinics, conducting defaulter tracking and providing active follow-up of mother–infant pairs. In several settings, these interventions led to reduced loss to follow-up among mother–infant pairs.

Ensuring provision of a comprehensive integrated postnatal package of HIV services will promote delivery of a set of interventions that contributes not only to improved HIV outcomes but better early childhood development overall.

References

1.
Smith DK, Herbst JH, Zhang X, Rose CE. Condom effectiveness for HIV prevention by consistency of use among men who have sex with men in the United States. J Acquir Immune Defic Syndr. 2015;68:337–44. [PubMed: 25469526]
2.
Weller SC, Davis-Beaty K. Condom effectiveness in reducing heterosexual HIV transmission. Cochrane Database Syst Rev. 2002;(1):CD003255. [PubMed: 11869658]
3.
French P, Latka M, Gollub E, Rogers C, Hoover D, Stein Z. Use-effectiveness of the female versus male condom in preventing sexually transmitted disease in women. Sex Transm Dis. 2003;30:433–9. [PubMed: 12916135]
4.
Effectiveness of sterile needle and syringe programming in reducing HIV/AIDS among injecting drug users. Geneva: World Health Organization; 2004 (https://apps​.who.int​/iris/handle/10665/43107, accessed 1 June 2021).
5.
Effectiveness of drug dependence treatment in preventing HIV among injecting drug users. Geneva: World Health Organization; 2005 (https://apps​.who.int​/iris/handle/10665/43259, accessed 1 June 2021).
6.
Guidelines for the psychosocially assisted pharmacological treatment of opioid dependence. Geneva: World Health Organization; 2009 (https://apps​.who.int​/iris/handle/10665/43948, accessed 1 June 2021). [PubMed: 23762965]
7.
Community management of opioid overdose. Geneva: World Health Organization; 2014 (https://apps​.who.int​/iris/handle/10665/137462, accessed 1 June 2021). [PubMed: 25577941]
8.
Preventing HIV through safe voluntary medical male circumcision for adolescent boys and men in generalized HIV epidemics: recommendations and key considerations. Geneva: World Health Organization; 2020 (https://apps​.who.int​/iris/handle/10665/333841, accessed 1 June 2021) [PubMed: 32986340]
9.
Farley TM, Samuelson J, Grabowski MK, Ameyan W, Gray RH, Baggaley R. Impact of male circumcision on risk of HIV infection in men in a changing epidemic context–systematic review and meta-analysis. J Int AIDS Soc. 2020;23:e25490. [PMC free article: PMC7303540] [PubMed: 32558344]
10.
Consolidated guidelines on HIV prevention, diagnosis, treatment and care for key populations. Geneva: World Health Organization; 2014 (https://apps​.who.int​/iris/handle/10665/128048, accessed 1 June 2021). [PubMed: 25996019]
11.
Consolidated guidelines on HIV prevention, diagnosis, treatment and care for key populations. Geneva: World Health Organization; 2016 (https://apps​.who.int​/iris/handle/10665/246200, accessed 1 June 2021). [PubMed: 27559558]
12.
Guidelines: updated recommendations on HIV prevention, infant diagnosis, antiretroviral initiation and monitoring. Geneva: World Health Organization; 2021 (https://apps​.who.int​/iris/handle/10665/340190, accessed 1 June 2021). [PubMed: 33822559]
13.
Consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection: recommendations for a public health approach – second edition. Geneva: World Health Organization; 2016 (https://apps​.who.int​/iris/handle/10665/208825, accessed 1 June 2021). [PubMed: 27466667]
14.
Guidance on oral pre-exposure prophylaxis (PrEP) for serodiscordant couples, men and transgender women who have sex with men at high risk of HIV: recommendations for use in the context of demonstration projects. Geneva: World Health Organization; 2012 (https://apps​.who.int​/iris/handle/10665/75188, accessed 1 June 2021). [PubMed: 23586123]
15.
Fonner VA, Dalglish SL, Kennedy CE, Baggaley R, O’Reilly KR, Koechlin FM et al. Effectiveness and safety of oral HIV pre-exposure prophylaxis for all populations. AIDS. 2016;30:1973–83. [PMC free article: PMC4949005] [PubMed: 27149090]
16.
WHO implementation tool for pre-exposure prophylaxis of HIV infection. Geneva: World Health Organization; 2017 (https://apps​.who.int​/iris/handle/10665/255890, accessed 1 June 2021).
17.
WHO technical brief: preventing HIV during pregnancy and breastfeeding in the context of PrEP. Geneva: World Health Organization; 2017 (https://apps​.who.int​/iris/handle/10665/255866, accessed 1 June 2021).
18.
Update on antiretroviral regimens for treating and preventing HIV infection and update on early infant diagnosis of HIV: interim guidance. Geneva: World Health Organization; 2018 (https://apps​.who.int​/iris/handle/10665/273129, accessed 1 June 2021).
19.
What’s the 2+1+1? Event driven PrEP to prevent HIV in gay men and other men who have sex with men: update to WHO’s recommendation on oral PrEP. Geneva: World Health Organization 2019 (https://apps​.who.int​/iris/handle/10665/325955, accessed 1 June 2021).
20.
Technical brief: prevention and control of sexually transmitted infections (STIs) in the era of pre-exposure prophylaxis (PrEP) for HIV. Geneva: World Health Organization; 2019 (https://apps​.who.int​/iris/handle/10665/325908, accessed 1 June 2021).
21.
Schaefer R, Schmidt H-M, Ravasi G, Mozalevskis A, Rewari BB, Lule F et al. Global adoption of guidelines on and use of oral pre-exposure prophylaxis (PrEP): current situation and future projects. Lancet HIV. In press.
22.
Guideline on when to start antiretroviral therapy and on pre-exposure prophylaxis for HIV. Geneva: World Health Organization; 2015 (https://apps​.who.int​/iris/handle/10665/186275, accessed 1 June 2021). [PubMed: 26598776]
23.
Chou R, Evans C, Hoverman A, Sun C, Dana T, Bougatsos C et al. Preexposure prophylaxis for the prevention of HIV infection: evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2019;321:2214–30. [PubMed: 31184746]
24.
Baeten JM, Donnell D, Mugo NR, Ndase P, Thomas KK, Campbell JD et al. Single-agent tenofovir versus combination emtricitabine plus tenofovir for pre-exposure prophylaxis for HIV-1 acquisition: an update of data from a randomised, double-blind, phase 3 trial. Lancet Infect Dis. 2014;14:1055–64. [PMC free article: PMC4252589] [PubMed: 25300863]
25.
Wilton J, Senn H, Sharma M, Tan DH. Pre-exposure prophylaxis for sexually-acquired HIV risk management: a review. HIV AIDS (Auckl). 2015;7:125. [PMC free article: PMC4422285] [PubMed: 25987851]
26.
Deutsch MB, Glidden DV, Sevelius J, Keatley J, McMahan V, Guanira J et al. HIV pre-exposure prophylaxis in transgender women: a subgroup analysis of the iPrEx trial. Lancet HIV. 2015;2:e512–9. [PMC free article: PMC5111857] [PubMed: 26614965]
27.
Pacifico de Carvalho N, Mendicino CCP, Candido RCF, Alecrim DJD, Menezes de Padua CA. HIV pre-exposure prophylaxis (PrEP) awareness and acceptability among trans women: a review. AIDS Care. 2019;31:1234–40. [PubMed: 31043069]
28.
Martin M, Vanichseni S, Suntharasamai P, Sangkum U, Mock PA, Gvetadze RJ et al. Renal function of participants in the Bangkok tenofovir study – Thailand, 2005–2012. Clin Infect Dis. 2014;59:716–24. [PubMed: 24829212]
29.
Solomon MM, Lama JR, Glidden DV, Mulligan K, McMahan V, Liu AY et al. Changes in renal function associated with oral emtricitabine/tenofovir disoproxil fumarate use for HIV pre-exposure prophylaxis. AIDS. 2014;28:851. [PMC free article: PMC3966916] [PubMed: 24499951]
30.
Liu AY, Vittinghoff E, Sellmeyer DE, Irvin R, Mulligan K, Mayer K et al. Bone mineral density in HIV-negative men participating in a tenofovir pre-exposure prophylaxis randomized clinical trial in San Francisco. PLoS One. 2011;6:e23688. [PMC free article: PMC3163584] [PubMed: 21897852]
31.
Van De Vijver DA, Nichols BE, Abbas UL, Boucher CA, Cambiano V, Eaton JW et al. Preexposure prophylaxis will have a limited impact on HIV-1 drug resistance in sub-Saharan Africa: a comparison of mathematical models. AIDS. 2013;27:2943–51. [PubMed: 23939237]
32.
Gibas KM, van den Berg P, Powell VE, Krakower DS. Drug resistance during HIV pre-exposure prophylaxis. Drugs. 2019;79:609–19. [PMC free article: PMC6606557] [PubMed: 30963509]
33.
Marcus JL, Glidden DV, Mayer KH, Liu AY, Buchbinder SP, Amico KR et al. No evidence of sexual risk compensation in the iPrEx trial of daily oral HIV preexposure prophylaxis. PLoS One. 2013;8:e81997. [PMC free article: PMC3867330] [PubMed: 24367497]
34.
Guest G, Shattuck D, Johnson L, Akumatey B, Clarke EEK, Chen P-L et al. Changes in sexual risk behavior among participants in a PrEP HIV prevention trial. Sex Transm Dis. 2008;35:1002–8. [PubMed: 19051397]
35.
Holt M, Broady TR, Mao L, Chan C, Rule J, Ellard J et al. Increasing preexposure prophylaxis use and ‘net prevention coverage’ in behavioural surveillance of Australian gay and bisexual men. AIDS. 2021;35:835–40. [PubMed: 33587442]
36.
Quaife M, MacGregor L, Ong JJ, Gafos M, Torres-Rueda S, Grant H et al. Risk compensation and sexually transmitted infection incidence in PrEP programmes. Lancet HIV. 2020;7:e222–3. [PubMed: 31767536]
37.
Rojas Castro D, Delabre RM, Molina JM. Give PrEP a chance: moving on from the “risk compensation” concept. J Int AIDS Soc. 2019;22(Suppl. 6):e25351. [PMC free article: PMC6715948] [PubMed: 31468693]
38.
Baeten JM, Donnell D, Ndase P, Mugo NR, Campbell JD, Wangisi J et al. Antiretroviral prophylaxis for HIV prevention in heterosexual men and women. N Engl J Med. 2012;367:399–410. [PMC free article: PMC3770474] [PubMed: 22784037]
39.
Van Damme L, Corneli A, Ahmed K, Agot K, Lombaard J, Kapiga S et al. Preexposure prophylaxis for HIV infection among African women. N Engl J Med. 2012;367:411–22. [PMC free article: PMC3687217] [PubMed: 22784040]
40.
Yager JL, Anderson PL. Pharmacology and drug interactions with HIV PrEP in transgender persons receiving gender affirming hormone therapy. Expert Opin Drug Metab Toxicol. 2020;16:463–74. [PubMed: 32250177]
41.
Hiransuthikul A, Janamnuaysook R, Himmad K, Kerr SJ, Thammajaruk N, Pankam T et al. Drug–drug interactions between feminizing hormone therapy and pre-exposure prophylaxis among transgender women: the iFACT study. J Int AIDS Soc. 2019;22:e25338. [PMC free article: PMC6625338] [PubMed: 31298497]
42.
Shieh E, Marzinke MA, Fuchs EJ, Hamlin A, Bakshi R, Aung W et al. Transgender women on oral HIV pre-exposure prophylaxis have significantly lower tenofovir and emtricitabine concentrations when also taking oestrogen when compared to cisgender men. J Int AIDS Soc. 2019;22:e25405. [PMC free article: PMC6832671] [PubMed: 31692269]
43.
Grant RM, Pellegrini M, Defechereux PA, Anderson PL, Yu M, Glidden DV et al. Sex hormone therapy and tenofovir diphosphate concentration in dried blood spots: primary results of the iBrEATHe Study. Clin Infect Dis. 2020;ciaa1160. [PMC free article: PMC8492111] [PubMed: 32766890]
44.
Grant RM, Anderson PL, McMahan V, Liu A, Amico KR, Mehrotra M et al. Uptake of pre-exposure prophylaxis, sexual practices, and HIV incidence in men and transgender women who have sex with men: a cohort study. Lancet Infect Dis. 2014;14:820–9. [PMC free article: PMC6107918] [PubMed: 25065857]
45.
Hoagland B, Moreira RI, De Boni RB, Kallas EG, Madruga JV, Vasconcelos R et al. High pre-exposure prophylaxis uptake and early adherence among men who have sex with men and transgender women at risk for HIV Infection: the PrEP Brasil demonstration project. J Int AIDS Soc. 2017;20:21472. [PMC free article: PMC5515021] [PubMed: 28418232]
46.
Untangling the web of antiretroviral price reductions. Geneva: Médecins Sans Frontières; 2014 (https://www​.msfaccess​.org/sites/default/files​/MSF_UTW_17th_Edition_4_b.pdf, accessed 1 June 2021).
47.
Koechlin FM, Fonner VA, Dalglish SL, O’Reilly KR, Baggaley R, Grant RM et al. Values and preferences on the use of oral pre-exposure prophylaxis (PrEP) for HIV prevention among multiple populations: a systematic review of the literature. AIDS Behav. 2017;21:1325–35. [PMC free article: PMC5378753] [PubMed: 27900502]
48.
Marrazzo JM, Ramjee G, Richardson BA, Gomez K, Mgodi N, Nair G et al. Tenofovir-based preexposure prophylaxis for HIV infection among African women. N Engl J Med. 2015;372:509–18. [PMC free article: PMC4341965] [PubMed: 25651245]
49.
Bekker L-G, Hughes J, Amico R, Roux S, Hendrix C, Anderson PL et al. HPTN 067/ADAPT Cape Town: a comparison of daily and nondaily PrEP dosing in African women. 22nd Conference on Retroviruses and Opportunistic Infections, Seattle, WA, USA, 23–26 February 2015 (https://www​.croiconference​.org/abstract/hptn-067adapt-cape-town-comparison-daily-and-nondaily-prep-dosing-african-women, accessed 1 June 2021).
50.
Henderson F, Taylor A, Chirwa L, Williams T, Borkowf C, Kasonde M et al. Characteristics and oral PrEP adherence in the TDF2 open-label extension in Botswana. J Int AIDS Soc. 2015;18.
51.
Baeten J, Heffron R, Kidoguchi L, Mugo N, Katabira E, Bukusi E. Partners Demonstration Project Team. Near elimination of HIV transmission in a demonstration project of PrEP and ART. 22nd Conference on Retroviruses and Opportunistic Infections, Seattle, WA, USA, 23–26 February 2015 (https://www​.croiconference​.org/abstract/near-elimination-hiv-transmission-demonstration-project-prep-and-art, accessed 1 June 2021).
52.
McCormack S, Dunn D. Pragmatic open-label randomised trial of preexposure prophylaxis: the PROUD study. 22nd Conference on Retroviruses and Opportunistic Infections, Seattle, WA, USA, 23–26 February 2015 (https://www​.croiconference​.org/abstract/pragmatic-open-label-randomised-trial-preexposure-prophylaxis-proud-study, accessed 1 June 2021).
53.
Grant RM, Mannheimer S, Hughes JP, Hirsch-Moverman Y, Loquere A, Chitwarakorn A. Daily and nondaily oral preexposure prophylaxis in men and transgender women who have sex with men: the Human Immunodeficiency Virus Prevention Trials Network 067/ADAPT Study. Clin Infect Dis. 2018;66:1712–21. [PMC free article: PMC5961078] [PubMed: 29420695]
54.
Holtz T, Chitwarakorn A, Curlin M, Hughes J, Amico K, Hendrix C et al. HPTN 067/ADAPT study: a comparison of daily and non-daily pre-exposure prophylaxis dosing in Thai men who have sex with men, Bangkok, Thailand. J Int AIDS Soc. 2015;18.
55.
Liu A, Cohen S, Vittinghoff E, Anderson P, Doblecki-Lewis S, Bacon O. Adherence, sexual behavior and HIV/STI incidence among men who have sex with men (MSM) and transgender women (TGW) in the US PrEP demonstration (Demo) project. 8th IAS Conference on HIV Pathogenesis, Treatment and Prevention, Vancouver, Canada, 18–22 July 2015 (https://www​.natap.org/2015/IAS/IAS_80​.htm, accessed 1 June 2021).
56.
Hosek S, Rudy B, Landovitz R, Kapogiannis B, Siberry G, Rutledge B et al. An HIV pre-exposure prophylaxis (PrEP) demonstration project and safety study for young gay men and other men who have sex with men. J Acquir Immune Defic Syndr. 2017;74:21. [PMC free article: PMC5140725] [PubMed: 27632233]
57.
Machtinger EL, Cuca YP, Khanna N, Rose CD, Kimberg LS. From treatment to healing: the promise of trauma-informed primary care. Womens Health Issues. 2015;25:193–7. [PubMed: 25965151]
58.
Bekker L-G, Johnson L, Cowan F, Overs C, Besada D, Hillier S et al. Combination HIV prevention for female sex workers: what is the evidence? Lancet. 2015;385:72–87. [PMC free article: PMC10318470] [PubMed: 25059942]
59.
Schaefer R, Amparo da Costa Leite P, Silva R, Abdool Karim Q, Akolo C, Caceres C et al. Kidney function in oral pre-exposure prophylaxis users: a systematic literature review and individual patient data meta-analysis. In preparation.
60.
Guidelines for the prevention, care and treatment of persons with chronic hepatitis B infection. Geneva: World Health Organization; 2015 (https://apps​.who.int​/iris/handle/10665/154590, accessed 1 June 2021). [PubMed: 26225396]
61.
World Health Organization. Hepatitis B vaccines: WHO position paper – July 2017. Wkly Epidemiol Rec. 2017;92:369–92 (https://apps​.who.int​/iris/handle/10665/255873, accessed 1 June 2021). [PubMed: 28685564]
62.
Grant RM, Lama JR, Anderson PL, McMahan V, Liu AY, Vargas L et al. Preexposure chemoprophylaxis for HIV prevention in men who have sex with men. N Engl J Med. 2010;363:2587–99. [PMC free article: PMC3079639] [PubMed: 21091279]
63.
Peterson L, Taylor D, Roddy R, Belai G, Phillips P, Nanda K et al. Tenofovir disoproxil fumarate for prevention of HIV infection in women: a phase 2, double-blind, randomized, placebo-controlled trial. PLOS Clin Trial. 2007;2:e27. [PMC free article: PMC1876601] [PubMed: 17525796]
64.
Guidelines for the care and treatment of persons diagnosed with chronic hepatitis C virus infection. Geneva: World Health Organization; 2018 (http://apps​.who.int/iris​/bitstream/handle​/10665/273174/9789241550345-eng.pdf, accessed 1 June 2021). [PubMed: 30307724]
65.
Holtz T, Chitwarakorn A, Curlin M, Hughes J, Amico K, Hendrix C. HPTN 067/ADAPT study: a comparison of daily and non-daily pre-exposure prophylaxis dosing in Thai men who have sex with men, Bangkok, Thailand. J Int AIDS Soc. 2015;18.
66.
Cottrell ML, Yang KH, Prince HM, Sykes C, White N, Malone S et al. A translational pharmacology approach to predicting outcomes of preexposure prophylaxis against HIV in men and women using tenofovir disoproxil fumarate with or without emtricitabine. J Infect Dis. 2016;214:55–64. [PMC free article: PMC4907409] [PubMed: 26917574]
67.
Guidelines on post-exposure prophylaxis for HIV and the use of co-trimoxazole prophylaxis for HIV-related infections among adults, adolescents and children: recommendations for a public health approach. Geneva: World Health Organization; 2014 (https://apps​.who.int​/iris/handle/10665/145719, accessed 1 June 2021). [PubMed: 26042326]
68.
Joseph Davey DL, Pintye J, Baeten JM, Aldrovandi G, Baggaley R, Bekker LG et al. Emerging evidence from a systematic review of safety of pre-exposure prophylaxis for pregnant and postpartum women: where are we now and where are we heading? J Int AIDS Soc. 2020;23:e25426. [PMC free article: PMC6948023] [PubMed: 31912985]
69.
Mistler CB, Copenhaver MM, Shrestha R. The pre-exposure prophylaxis (PrEP) care cascade in people who inject drugs: a systematic review. AIDS Behav. 2021;25:1490–506. [PMC free article: PMC7858689] [PubMed: 32749627]
70.
Dapivirine vaginal ring 25 mg (dapivirine): an overview of dapivirine vaginal ring 25 mg and why it received a positive opinion. Amsterdam: European Medicines Agency; 2020 (https://www​.ema.europa​.eu/en/documents/medicine-outside-eu​/dapivirine-vaginal-ring-25-mg-medicine-overview_en.pdf, accessed 1 June 2021).
71.
Assessment report: dapivirine vaginal ring 25 mg. Amsterdam: European Medicines Agency; 2020 (https://www​.ema.europa​.eu/en/documents/medicine-outside-eu​/dapivirine-vaginal-ring-25-mg-public-assessment-report_en.pdf, accessed 1 June 2021).
72.
Ahmed K, Baeten JM, Beksinska M, Bekker L-G, Bukusi EA, Donnell D et al. HIV incidence among women using intramuscular depot medroxyprogesterone acetate, a copper intrauterine device, or a levonorgestrel implant for contraception: a randomised, multicentre, open-label trial. Lancet. 2019;394:303–13. [PMC free article: PMC6675739] [PubMed: 31204114]
73.
Celum C, Delany-Moretlwe S, Hosek S, Dye B, Bekker L-G, Mgodi N. Risk behavior, perception, and reasons for PrEP among young African women in HPTN 082. 23rd Conference on Retroviruses and Opportunistic Infections, Boston, MA, USA, 22–25 February 2016 (https://www​.croiconference​.org/abstract/risk-behavior-perception-and-reasons-prep-among-young-african-women-hptn-082, accessed 1 June 2021).
74.
Koss C, Havlir D, Ayieko J, Kwarisiima D, Kabami J, Atukunda M. Lower than expected HIV incidence among men and women at elevated HIV risk in a population-based PrEP study in rural Kenya and Uganda: interim results from the SEARCH study. 23rd International AIDS Conference, virtual, 6–10 July 2020 (https://aids2020​.org​/wp-content/uploads/2020​/07/HIV-Highlights-Press-Conference-Abstracts.pdf, accessed 1 June 2021)
75.
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:e25463. [PMC free article: PMC7060297] [PubMed: 32144874]
76.
Scorgie F, Khoza N, Baron D, Lees S, Harvey S, Ramskin L et al. Disclosure of PrEP use by young women in South Africa and Tanzania: qualitative findings from a demonstration project. Cult Health Sex. 2021;23:257–72. [PubMed: 32129720]
77.
Corneli A, Perry B, McKenna K, Agot K, Ahmed K, Taylor J et al. Participants’ explanations for nonadherence in the FEM-PrEP clinical trial. J Acquir Immune Defic Syndr. 2016;71:452–61. [PubMed: 26536315]
78.
Montgomery ET, Beksinska M, Mgodi N, Schwartz J, Weinrib R, Browne EN et al. End-user preference for and choice of four vaginally delivered HIV prevention methods among young women in South Africa and Zimbabwe: the Quatro Clinical Crossover Study. J Int AIDS Soc. 2019;22:e25283. [PMC free article: PMC6506690] [PubMed: 31069957]
79.
van der Straten A, Agot K, Ahmed K, Weinrib R, Browne EN, Manenzhe K et al. The Tablets, Ring, Injections as Options (TRIO) study: what young African women chose and used for future HIV and pregnancy prevention. J Int AIDS Soc. 2018;21:e25094. [PMC free article: PMC5876496] [PubMed: 29600595]
80.
Nel A, Bekker L-G, Bukusi E, Hellstrӧm E, Kotze P, Louw C et al. Safety, acceptability and adherence of dapivirine vaginal ring in a microbicide clinical trial conducted in multiple countries in sub-Saharan Africa. PLoS One. 2016;11:e0147743. [PMC free article: PMC4786336] [PubMed: 26963505]
81.
Baeten JM, Palanee-Phillips T, Brown ER, Schwartz K, Soto-Torres LE, Govender V et al. Use of a vaginal ring containing dapivirine for HIV-1 prevention in women. N Engl J Med. 2016;375:2121–32. [PMC free article: PMC4993693] [PubMed: 26900902]
82.
Brown ER, Hendrix CW, van der Straten A, Kiweewa FM, Mgodi NM, Palanee-Philips T et al. Greater dapivirine release from the dapivirine vaginal ring is correlated with lower risk of HIV-1 acquisition: a secondary analysis from a randomized, placebo-controlled trial. J Int AIDS Soc. 2020;23:e25634. [PMC free article: PMC7673220] [PubMed: 33206462]
83.
Nel A, van Niekerk N, Van Baelen B, Malherbe M, Mans W, Carter A et al. Safety, adherence, and HIV-1 seroconversion among women using the dapivirine vaginal ring (DREAM): an open-label, extension study. Lancet HIV. 2021;8:e77–86. [PubMed: 33539761]
84.
Baeten JM, Palanee-Phillips T, Mgodi NM, Mayo AJ, Szydlo DW, Ramjee G et al. Safety, uptake, and use of a dapivirine vaginal ring for HIV-1 prevention in African women (HOPE): an open-label, extension study. Lancet HIV. 2021;8:e87–95. [PMC free article: PMC8038210] [PubMed: 33539762]
85.
Brown ER, Hendrix CW, van der Straten A, Kiweewa FM, Mgodi NM, Palanee-Philips T et al. Greater dapivirine release from the dapivirine vaginal ring is correlated with lower risk of HIV-1 acquisition: a secondary analysis from a randomized, placebo-controlled trial. J Int AIDS Soc. 2020;23:e25634. [PMC free article: PMC7673220] [PubMed: 33206462]
86.
MTN-042 – A study of PrEP and the dapivirine ring in pregnant women. Microbicide Trials Network; 2020 (https://mtnstopshiv​.org​/research/studies/mtn-042).
87.
MTN-043 – B-PROTECTED: Breastfeeding. PrEP & ring open-label trial. Microbicide Trials Network; 2020 (https://mtnstopshiv​.org​/news/studies/mtn043).
88.
Rosenberg Z, Nel A, van Niekerk N, Van Baelen B, Van Roey J, Palanee-Phillips T et al. Pooled efficacy analysis of two Phase III trials of dapivirine vaginal ring for the reduction of HIV-1 infection risk in HIV-uninfected women in sub-Saharan Africa. 9th IAS Conference on HIV Science, Paris, France, 23–26 July 2017 (https://www​.ipmglobal​.org/sites/default/files​/ias_dpv_ring_pooled​_analysis_poster_21_july_2017.pdf, accessed 1 June 2021).
89.
Nel A, Van Baelen BE, Mans W, Louw C, Gama C, Mabude Z et al. Dapivirine vaginal ring reduces the risk of HIV-1 infection among women in Africa. 9th South Africa AIDS Conference, Durban, South Africa, 11–14 June 2019.
90.
Nel A, van Niekerk N, Kapiga S, Bekker L-G, Gama C, Gill K et al. Safety and efficacy of a dapivirine vaginal ring for HIV prevention in women. N Engl J Med. 2016;375:2133–43. [PubMed: 27959766]
91.
Fonner V, Dalglish S. Dapivirine intervaginal ring as pre-exposure prophylaxis to prevent HIV among women at substantial risk of infection: a systematic review and meta-analysis. Unpublished.
92.
Palanee-Phillips T, Roberts ST, Reddy K, Govender V, Naidoo L, Siva S et al. Impact of partner-related social harms on women’s adherence to the dapivirine vaginal ring during a phase III trial. J Acquir Immune Defic Syndr. 2018;79:580. [PMC free article: PMC6231955] [PubMed: 30239426]
93.
Balkus JE, Palanee-Phillips T, Reddy K, Siva S, Harkoo I, Nakabiito C et al. Dapivirine vaginal ring use does not diminish the effectiveness of hormonal contraception. J Acquir Immune Defic Syndr. 2017;76:e47. [PMC free article: PMC5597463] [PubMed: 28542081]
94.
Makanani B, Balkus JE, Jiao Y, Noguchi LM, Palanee-Phillips T, Mbilizi Y et al. Pregnancy and infant outcomes among women using the dapivirine vaginal ring in early pregnancy. J Acquir Immune Defic Syndr. 2018;79:566. [PMC free article: PMC6231990] [PubMed: 30383589]
95.
Kusemererwa S, Abaasa A. Pregnancy incidence and outcomes among women using dapivirine vaginal ring for HIV prevention in a phase III clinical trial in south western Uganda. HIV Research for Prevention Meeting, Madrid, Spain, 21–25 October 2018 (https://www​.liebertpub​.com/doi/10.1089/aid​.2018.5000.abstracts, accessed 1 June 2021).
96.
Kusemererwa S, Abaasa A. Does the use of the dapivirine vaginal ring result in change in risk sexual behavior? HIV Research for Prevention Meeting, Madrid, Spain, 21–25 October 2018 (https://www​.liebertpub​.com/doi/10.1089/aid​.2018.5000.abstracts, accessed 1 June 2021).
97.
Glaubius R, Ding Y, Penrose KJ, Hood G, Engquist E, Mellors JW et al. Dapivirine vaginal ring for HIV prevention: modelling health outcomes, drug resistance and cost-effectiveness. J Int AIDS Soc. 2019;22:e25282. [PMC free article: PMC6510112] [PubMed: 31074936]
98.
Glaubius R, Penrose KJ, Hood G, Parikh UM, Abbas U. Dapivirine vaginal ring preexposure prophylaxis for HIV prevention in South Africa. Topics Antivir Med. 2016;24(E-1):458.
99.
Smith J, Harris K, Garnett G, Van Damme L, Hallett T. Cost–effectiveness of the intravaginal dapivirine ring: a modeling analysis. Topics Antivir Med. 2016;24(E-1):458.
100.
Reidy M, Gardiner E, Pretorius C, Glaubius R, Torjesen K, Kripke K. Evaluating the potential impact and cost–effectiveness of dapivirine vaginal ring pre-exposure prophylaxis for HIV prevention. PLoS One. 2019;14:e0218710. [PMC free article: PMC6594614] [PubMed: 31242240]
101.
Kripke K, Reidy M, Bhavaraju N, Torjesen K, Gardiner E. Modeling the potential impact of the dapivirine ring for HIV prevention. 22nd International AIDS Society Conference, Amsterdam, Netherlands, 23–27 July 2018 (https://www​.prepwatch​.org/wp-content/uploads​/2018/08/OPTIONS​_DapRingModeling_AIDS2018poster-1.pdf, accessed 1 June 2021).
102.
Nel A, Haazen W, Nuttall J, Romano J, Rosenberg Z, van Niekerk N. A safety and pharmacokinetic trial assessing delivery of dapivirine from a vaginal ring in healthy women. AIDS. 2014;28:1479–87. [PubMed: 24901365]
103.
Chen BA, Zhang J, Gundacker HM, Hendrix CW, Hoesley CJ, Salata RA et al. Phase 2a safety, pharmacokinetics, and acceptability of dapivirine vaginal rings in US postmenopausal women. Clin Infect Dis. 2019;68:1144–51. [PMC free article: PMC6424088] [PubMed: 30289485]
104.
Bunge KE, Levy L, Szydlo DW, Zhang J, Gaur AH, Reirden D et al. Brief report: phase IIa safety study of a vaginal ring containing dapivirine in adolescent young women. J Acquir Immune Defic Syndr. 2020;83:135–9. [PMC free article: PMC8577288] [PubMed: 31929401]
105.
Griffin JB, Ridgeway K, Montgomery E, Torjesen K, Clark R, Peterson J et al. Vaginal ring acceptability and related preferences among women in low-and middle-income countries: a systematic review and narrative synthesis. PLoS One. 2019;14:e0224898. [PMC free article: PMC6839883] [PubMed: 31703094]
106.
Schwartz K, Bhavaraju N, Ridgeway K, Gomez A. End-user perspectives on their ability, motivation and opportunity to use the dapivirine vaginal ring. 23rd International AIDS Conference, virtual, 6–10 July 2020 (https://programme​.aids2020​.org/Abstract/AbstractList?abstractGrid-sort​=AbstractNumber-asc&abstractGrid-page=168, accessed 1 June 2021).
107.
Ridgeway KM, Montgomery ET, Smith K, Torjesen K, van der Straten A, Achilles SL. Vaginal ring acceptability: a systematic review and meta-analysis of vaginal ring experiences from around the world. In preparation.
108.
van der Straten A, Panther L, Laborde N, Hoesley CJ, Cheng H, Husnik MJ et al. Adherence and acceptability of a multidrug vaginal ring for HIV prevention in a phase I study in the United States. AIDS Behav. 2016;20:2644–53. [PMC free article: PMC4970965] [PubMed: 26837628]
109.
Montgomery ET, van der Straten A, Chitukuta M, Reddy K, Woeber K, Atujuna M et al. Acceptability and use of a dapivirine vaginal ring in a phase III trial. AIDS. 2017;31:1159. [PMC free article: PMC5557083] [PubMed: 28441175]
110.
Chitukuta M, Duby Z, Katz A, Nakyanzi T, Reddy K, Palanee-Phillips T et al. Negative rumours about a vaginal ring for HIV-1 prevention in sub-Saharan Africa. Culture Health Sexuality. 2019;21:1209–24. [PMC free article: PMC6639142] [PubMed: 30657023]
111.
Duby Z, Katz AW, Browne EN, Mutero P, Etima J, Zimba CC et al. Hygiene, blood flow, and vaginal overload: why women removed an HIV prevention vaginal ring during menstruation in Malawi, South Africa, Uganda and Zimbabwe. AIDS Behav. 2020;24:617–28. [PMC free article: PMC6815681] [PubMed: 31030301]
112.
Laborde ND, Pleasants E, Reddy K, Atujuna M, Nakyanzi T, Chitukuta M et al. Impact of the dapivirine vaginal ring on sexual experiences and intimate partnerships of women in an HIV prevention clinical trial: managing ring detection and hot sex. AIDS Behav. 2018;22:437–46. [PMC free article: PMC5866044] [PubMed: 29151197]
113.
Nair G, Roberts S, Baeten J, Palanee-Philips T, Katie S, Reddy K et al. Disclosure of vaginal ring use to male partners in an HIV prevention study: impact on adherence. HIV Research for Prevention Meeting, Madrid, Spain, 21–25 October 2018 (https://www​.liebertpub​.com/doi/10.1089/aid​.2018.5000.abstracts, accessed 1 June 2021).
114.
van der Straten A, Browne EN, Shapley-Quinn MK, Brown ER, Reddy K, Scheckter R et al. First impressions matter: how initial worries influence adherence to the dapivirine vaginal ring. J Acquir Immune Defic Syndr. 2019;81:304–10. [PMC free article: PMC6571014] [PubMed: 30844995]
115.
Ross J, Stover J. Use of modern contraception increases when more methods become available: analysis of evidence from 1982–2009. Global Health: Sci Pract. 2013;1:203–12. [PMC free article: PMC4168565] [PubMed: 25276533]
116.
Consolidated guidelines on HIV testing services. Geneva: World Health Organization; 2019 (https://apps​.who.int​/iris/handle/10665/336323, accessed 1 June 2021).
117.
NCT03074786: MTN-034/REACH (Reversing the Epidemic in Africa with Choices in HIV Prevention). 2020 (https:​//clinicaltrials​.gov/ct2/show/NCT03074786, accessed 1 June 2021).
118.
Ford N, Shubber Z, Calmy A, Irvine C, Rapparini C, Ajose O et al. Choice of antiretroviral drugs for postexposure prophylaxis for adults and adolescents: a systematic review. Clin Infect Dis. 2015;60:S170–6. [PubMed: 25972499]
119.
Penazzato M, Dominguez K, Cotton M, Barlow-Mosha L, Ford N. Choice of antiretroviral drugs for postexposure prophylaxis for children: a systematic review. Clin Infect Dis. 2015;60:S177–81. [PubMed: 25972500]
120.
Responding to children and adolescents who have been sexually abused: WHO clinical guidelines. Geneva: World Health Organization; 2017 (https://apps​.who.int​/iris/handle/10665/259270, accessed 1 June 2021). [PubMed: 29630189]
121.
Updated recommendations on first-line and second-line antiretroviral regimens and post-exposure prophylaxis and recommendations on early infant diagnosis of HIV: interim guidelines: supplement to the 2016 consolidated guidelines on the use of antiretroviral drugs for treating and preventing HIV infection. Geneva: World Health Organization; 2018 (https://apps​.who.int​/iris/handle/10665/277395, accessed 1 June 2021).
122.
Wiboonchutikul S, Thientong V, Suttha P, Kowadisaiburana B, Manosuthi W. Significant intolerability of efavirenz in HIV occupational postexposure prophylaxis. J Hosp Infect. 2016;92:372–7. [PubMed: 26876748]
123.
Guideline – updates on HIV and infant feeding: the duration of breastfeeding, and support from health services to improve feeding practices among mothers living with HIV. Geneva: World Health Organization; 2016 (https://apps​.who.int​/iris/handle/10665/246260?mode=simple, accessed 1 June 2021). [PubMed: 27583316]
124.
Beste S, Essajee S, Siberry G, Hannaford A, Dara J, Sugandhi N et al. Optimal antiretroviral prophylaxis in infants at high risk of acquiring HIV: a systematic review. Pediatr Infect Dis J. 2018;37:169–75. [PubMed: 29319636]
125.
Nielsen-Saines K, Watts DH, Veloso VG, Bryson YJ, Joao EC, Pilotto JH et al. Three postpartum antiretroviral regimens to prevent intrapartum HIV infection. N Engl J Med. 2012;366:2368–79. [PMC free article: PMC3590113] [PubMed: 22716975]
126.
Coovadia HM, Brown ER, Fowler MG, Chipato T, Moodley D, Manji K et al. Efficacy and safety of an extended nevirapine regimen in infant children of breastfeeding mothers with HIV-1 infection for prevention of postnatal HIV-1 transmission (HPTN 046): a randomised, double-blind, placebo-controlled trial. Lancet. 2012;379:221–8. [PMC free article: PMC3539769] [PubMed: 22196945]
127.
Chiappini E, Galli L, Giaquinto C, Ene L, Goetghebuer T, Judd A et al. Use of combination neonatal prophylaxis for the prevention of mother-to-child transmission of HIV infection in European high-risk infants. AIDS. 2013;27:991–1000. [PubMed: 23211776]
128.
Lallemant M, Amzal B, Urien S, Sripan P, Cressey T, Ngo-Giang-Huong N et al. Antiretroviral intensification to prevent intrapartum HIV transmission in late comers. J Int AIDS Soc.2015;18(5Suppl. 4):20479.

Footnotes

1

WHO defines key populations as men who have sex with men, people in prisons and other closed settings, people who inject drugs, sex workers and transgender people.

2

For the recommendation on the dapivirine vaginal ring, the term women applies to cisgender women, meaning women assigned female at birth. There is no research at this time to support the dapivirine vaginal ring for other populations.

© World Health Organization 2021.

Sales, rights and licensing. To purchase WHO publications, see http://apps.who.int/bookorders. To submit requests for commercial use and queries on rights and licensing, see http://www.who.int/about/licensing.

Third-party materials. If you wish to reuse material from this work that is attributed to a third party, such as tables, figures or images, it is your responsibility to determine whether permission is needed for that reuse and to obtain permission from the copyright holder. The risk of claims resulting from infringement of any third-party-owned component in the work rests solely with the user.

Some rights reserved. This work is available under the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 IGO licence (CC BY-NC-SA 3.0 IGO; https://creativecommons.org/licenses/by-nc-sa/3.0/igo).

Under the terms of this licence, you may copy, redistribute and adapt the work for non-commercial purposes, provided the work is appropriately cited, as indicated below. In any use of this work, there should be no suggestion that WHO endorses any specific organization, products or services. The use of the WHO logo is not permitted. If you adapt the work, then you must license your work under the same or equivalent Creative Commons licence. If you create a translation of this work, you should add the following disclaimer along with the suggested citation: “This translation was not created by the World Health Organization (WHO). WHO is not responsible for the content or accuracy of this translation. The original English edition shall be the binding and authentic edition”.

Any mediation relating to disputes arising under the licence shall be conducted in accordance with the mediation rules of the World Intellectual Property Organization (http://www.wipo.int/amc/en/mediation/rules/).

Bookshelf ID: NBK572725

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...